1
|
Ma N, Liu P, Li N, Hu Y, Kang L. Exploring the pharmacological mechanisms for alleviating OSA: Adenosine A2A receptor downregulation of the PI3K/Akt/HIF‑1 pathway (Review). Biomed Rep 2025; 22:21. [PMID: 39720297 PMCID: PMC11668141 DOI: 10.3892/br.2024.1899] [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: 08/29/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Obstructive sleep apnea (OSA) is the most common type of sleep apnea, which leads to episodes of intermittent hypoxia due to obstruction of the upper airway. A key feature of OSA is the upregulation and stabilization of hypoxia-inducible factor 1 (HIF-1), a crucial metabolic regulator that facilitates rapid adaptation to changes in oxygen availability. Adenosine A2A receptor (A2AR), a major adenosine receptor, regulates HIF-1 under hypoxic conditions, exerting anti-inflammatory properties and affecting lipid metabolism. The present study explored the roles of A2AR in OSA regulation, specifically focusing on its effects via the PI3K/Akt/HIF-1 pathway. The findings enhance our understanding the pharmacological potential of A2AR in OSA management and suggest future research directions in exploring its clinical applications.
Collapse
Affiliation(s)
- Nini Ma
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| | - Peijie Liu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| | - Ning Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| | - Yushi Hu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| | - Liang Kang
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| |
Collapse
|
2
|
Zhang H, Ouyang Z, Zhou T, Su F, Wang M. Novel anoikis-related diagnostic biomarkers for aortic dissection based on machine learning. Sci Rep 2024; 14:31314. [PMID: 39732886 PMCID: PMC11682115 DOI: 10.1038/s41598-024-82655-6] [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: 02/14/2023] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
Aortic dissection (AD) is one of the most dangerous diseases of the cardiovascular system, which is characterized by acute onset and poor prognosis, while the pathogenesis of AD is still unclear and may affect or even delay the diagnosis of AD. Anchorage-dependent cell death (Anoikis) is a special mode of cell death, which is programmed cell death caused by normal cells after detachment from extracellular matrix (ECM) and has been widely studied in the field of oncology in recent years. In this study, we applied bioinformatics analysis, according to the results of research analysis and Gene Ontology (GO), as well as Kyoto Encyclopedia of Genes and Genomes (KEGG), finally found 3 anoikis-related genes (ARGs) based on machine learning. Among these, TP53 and TUBB3 were further verified by receiver operating characteristic (ROC), gene set enrichment analysis (GSEA), gene set variation analysis (GSVA)and other methods. We hypothesize ARGs may be involved in the pathogenesis of AD through pathways such as oxidative stress, inflammatory response, and ECM. Therefore, we conclude that these ARGs can be potential factors in determining the diagnosis of AD.
Collapse
Affiliation(s)
- Hanyi Zhang
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, Hunan, China
- Xiang Ya School of Medcine, Central South University, 172 Tongzipo Road, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhen Ouyang
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, Hunan, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tianji Zhou
- Xiang Ya Nursing School, Central South University, 172 Tongzipo Road, Changsha, Hunan, China
| | - Feng Su
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, Hunan, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Department of Department of Emergency Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410000, Hunan, China.
| | - Mi Wang
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, Hunan, China.
- Department of Mental Health Center, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
3
|
Lu C, Zhang Z, Fan Y, Wang X, Qian J, Bian Z. Shikonin induces ferroptosis in osteosarcomas through the mitochondrial ROS-regulated HIF-1α/HO-1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156139. [PMID: 39423479 DOI: 10.1016/j.phymed.2024.156139] [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: 05/29/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND The most common malignant bone tumour is osteosarcoma, which has an unsatisfactory prognosis and unsatisfactory treatment. Ferroptosis shows promise as an effective OS therapy. A substance extracted from the Lithospermum erythrorhizon, Shikonin (SHK), inhibits a number of tumours, including ovarian, gastric, and lung cancers. However, whether SHK induces OS ferroptosis and its mechanisms are not clear. PURPOSE Our study is aimed at investigating whether SHK causes ferroptosis and elucidating its molecular mechanism. METHODS Cell counting Kit-8, cell cycle and cell apoptosis assay were utilised to assess therapeutic effect of SHK against OS. Normal cells, including H9C2, AML-12 and HK-2, haematoxylin and eosin staining and mice serum biomarker tests were used to assess SHK biosafety. Malondialdehyde (MDA) levels, the glutathione (GSH)/oxidized glutathione (GSSG) ratio, reactive oxygen species (ROS), lipid peroxide (LPO), and intracellular Fe2+ detection, quantitative reverse transcription PCR (qRT-PCR), Western blotting (WB) and rescue experiments were employed to confirm whether SHK induced ferroptosis in OS cells. Molecular docking, cellular thermal shift assay (CETSA), and drug affinity responsive target stability (DARTS) assay were used to evaluate the direct binding between SHK and hypoxia-inducible factor-1α (HIF-1α). Protein stability and degradation analysis, small RNA interference, flow cytometry, qRT-PCR, and WB were used to investigate the molecular mechanism of ferroptosis. In vivo xenografts of nude mouse was used to study the anti-OS effect of SHK. RESULTS SHK significantly reduced OS cell viability and induced apoptosis and G2/M arrest. SHK increased intracellular levels of MDA, ROS, LPO, and Fe2+ while simultaneously reducing the GSH/GSSG ratio and GPX4 and SLC7A11 expression. CETSA and DARTS results demonstrated that SHK did not bind targetly to HIF-1α. Instead, mitochondrial ROS (MitoROS) promoted HIF-1α expression, resulting in HO-1 overexpression, excess Fe2+ production, ROS accumulation and GPX depletion, and ferroptosis. Furthermore, inhibition of MitoROS using Mito-TEMPO downregulated HIF-1α/HO-1 axis and mitigated the SHK-induced ferroptosis. In vivo, SHK effectively suppressed OS growth with favourable biosafety, confirming the molecular mechanism underlying SHK-induced ferroptosis in OS. CONCLUSION We observe that HIF-1α/HO-1 axis is the crucial factor in SHK-induced OS ferroptosis. Importantly, we demonstrate that HIF-1α is indirectly regulated by MitoROS rather than SHK bound directly to HIF-1α. Our research suggest that SHK is a potential drug candidate for OS treatment and may help in identifying novel therapeutic targets for OS.
Collapse
Affiliation(s)
- Congcong Lu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, PR China; Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Zhen Zhang
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Yuhao Fan
- Zhejiang University School of Medicine, Hangzhou, 310000, PR China
| | - Xiyu Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou 310053, PR China; Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China
| | - Jin Qian
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China.
| | - Zhenyu Bian
- Department of Orthopedics, Affiliated Hangzhou First People's Hospital, WestLake University School of Medicine, Hangzhou 310006, PR China.
| |
Collapse
|
4
|
Naushad S, Gaucher J, Mezdari Z, Détrait M, Belaidi E, Zhang Y, Vial G, Bouyon S, Czibik G, Pini M, Aldekwer S, Liang H, Pelloux V, Aron-Wisnewsky J, Tamisier R, Pépin JL, Derumeaux G, Sawaki D, Arnaud C. Chronic intermittent hypoxia triggers cardiac fibrosis: Role of epididymal white adipose tissue senescent remodeling? Acta Physiol (Oxf) 2024; 240:e14231. [PMID: 39263916 DOI: 10.1111/apha.14231] [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: 05/30/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024]
Abstract
AIM Obstructive sleep apnea (OSA) is a growing health problem affecting nearly 1 billion people worldwide. The landmark feature of OSA is chronic intermittent hypoxia (CIH), accounting for multiple organ damage, including heart disease. CIH profoundly alters both visceral white adipose tissue (WAT) and heart structure and function, but little is known regarding inter-organ interaction in the context of CIH. We recently showed that visceral WAT senescence drives myocardial alterations in aged mice without CIH. Here, we aimed at investigating whether CIH induces a premature visceral WAT senescent phenotype, triggering subsequent cardiac remodeling. METHODS In a first experiment, 10-week-old C57bl6J male mice (n = 10/group) were exposed to 14 days of CIH (8 h daily, 5%-21% cyclic inspired oxygen fraction, 60 s per cycle). In a second series, mice were submitted to either epididymal WAT surgical lipectomy or sham surgery before CIH exposure. Finally, we used p53 deficient mice or Wild-type (WT) littermates, also exposed to the same CIH protocol. Epididymal WAT was assessed for fibrosis, DNA damages, oxidative stress, markers of senescence (p16, p21, and p53), and inflammation by RT-qPCR and histology, and myocardium was assessed for fibrosis and cardiomyocyte hypertrophy. RESULTS CIH-induced epididymal WAT remodeling characterized by increased fibrosis, oxidative stress, DNA damage response, inflammation, and increased expression of senescent markers. CIH-induced epididymal WAT remodeling was associated with subtle and early myocardial interstitial fibrosis. Both epididymal WAT surgical lipectomy and p53 deletion prevented CIH-induced myocardial fibrosis. CONCLUSION Short-term exposure to CIH induces epididymal WAT senescent remodeling and cardiac interstitial fibrosis, the latter being prevented by lipectomy. This finding strongly suggests visceral WAT senescence as a new target to mitigate OSA-related cardiac disorders.
Collapse
Affiliation(s)
- Suzain Naushad
- Université Paris Est Créteil, INSERM U955, Créteil, France
| | - Jonathan Gaucher
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Zaineb Mezdari
- Université Paris Est Créteil, INSERM U955, Créteil, France
| | - Maximin Détrait
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Elise Belaidi
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Yanyan Zhang
- Université Paris Est Créteil, INSERM U955, Créteil, France
| | - Guillaume Vial
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Sophie Bouyon
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Gabor Czibik
- Université Paris Est Créteil, INSERM U955, Créteil, France
| | - Maria Pini
- Université Paris Est Créteil, INSERM U955, Créteil, France
| | - Sahar Aldekwer
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Hao Liang
- Université Paris Est Créteil, INSERM U955, Créteil, France
| | - Véronique Pelloux
- Nutrition and Obesities, Systemic Approaches, NutriOmics, Laboratory, Sorbonne University, Paris, France
- Nutrition Department, CRNH Ile de France, AP-HP, Pitie-Salpêtrière Hospital, Paris, France
| | - Judith Aron-Wisnewsky
- Nutrition and Obesities, Systemic Approaches, NutriOmics, Laboratory, Sorbonne University, Paris, France
- Nutrition Department, CRNH Ile de France, AP-HP, Pitie-Salpêtrière Hospital, Paris, France
| | - Renaud Tamisier
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Jean-Louis Pépin
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| | - Geneviève Derumeaux
- Université Paris Est Créteil, INSERM U955, Créteil, France
- Department of Physiology, AP-HP, Henri Mondor Hospital, FHU-SENEC, Créteil, France
| | - Daigo Sawaki
- Université Paris Est Créteil, INSERM U955, Créteil, France
| | - Claire Arnaud
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, HP2, Grenoble, France
| |
Collapse
|
5
|
El Amine B, Fournier J, Minoves M, Baillieul S, Roche F, Perek N, Pépin JL, Tamisier R, Khouri C, Rome C, Briançon-Marjollet A. Cerebral oxidative stress, inflammation and apoptosis induced by intermittent hypoxia: a systematic review and meta-analysis of rodent data. Eur Respir Rev 2024; 33:240162. [PMID: 39694586 DOI: 10.1183/16000617.0162-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/21/2024] [Indexed: 12/20/2024] Open
Abstract
Obstructive sleep apnoea (OSA) contributes to cerebrovascular diseases and cognitive decline. Preclinical studies support the deleterious impact on the brain of intermittent hypoxia (IH), one of the main components of OSA, but heterogeneity in rodent species and brain regions studied, or induced by IH paradigms, can challenge interpretation of the studies. Hence, we conducted a systematic review and meta-analysis to evaluate the impact of IH on rodent brain oxidative stress, inflammation, apoptosis and the expression of brain-derived neurotrophic factor (BDNF) and hypoxia-inducible factor 1 (HIF-1). PubMed and Web of Science searches identified 663 articles related to IH exposure, of which 60 were included. The examined outcomes were oxidative stress, inflammation, apoptosis, HIF-1 or BDNF in brains. Standardised mean difference was used to compare studies. Metaregressions were performed to clarify the impact of IH exposure parameters, rodent characteristics or cerebral localisation on these outcomes. IH-induced oxidative stress (increased malondialdehyde (MDA) and NADPH oxidase (NOX) and decreased superoxide dismutase), increased inflammation (tumour necrosis factor-α, NF-κB and inducible nitric oxide synthase), HIF-1 and apoptosis evaluated by terminal deoxynucleotidyl transferase dUTP nick-end labelling and cleaved caspase-3. In contrast, B-cell lymphoma 2 (BCL2) and BDNF expression were not significantly modified. Metaregressions showed that MDA, NOX and BDNF were associated with determinants of IH cycles (inspired oxygen fraction and duration of hypoxia) and some parameters depended on localisation. Rodent characteristics had little impact on the outcomes. Our meta-analysis robustly establishes that IH, independently of other confounders, has a strong effect on the brain by inducing oxidative stress, inflammation and apoptosis in rodent models. Our findings support the interest of considering and treating cerebral consequences of OSA in clinical practice.
Collapse
Affiliation(s)
- Bayan El Amine
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
- Univ. Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Joey Fournier
- Univ. Grenoble Alpes, Inserm CIC1406, CHU de Grenoble, Grenoble, France
| | - Mélanie Minoves
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Sébastien Baillieul
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Frédéric Roche
- Université Jean Monnet, Inserm U1059 Sainbiose, Saint Etienne, France
- Physiologie Clinique et de l'Exercice, CHU, Saint Etienne, France
| | - Nathalie Perek
- Université Jean Monnet, Inserm U1059 Sainbiose, Saint Etienne, France
| | - Jean-Louis Pépin
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Renaud Tamisier
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Charles Khouri
- Univ. Grenoble Alpes, Inserm U1300, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
- Univ. Grenoble Alpes, Inserm CIC1406, CHU de Grenoble, Grenoble, France
| | - Claire Rome
- Univ. Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, Grenoble, France
| | | |
Collapse
|
6
|
Elias-Llumbet A, Sharmin R, Berg-Sorensen K, Schirhagl R, Mzyk A. The Interplay between Mechanoregulation and ROS in Heart Physiology, Disease, and Regeneration. Adv Healthc Mater 2024; 13:e2400952. [PMID: 38962858 DOI: 10.1002/adhm.202400952] [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: 03/13/2024] [Revised: 05/16/2024] [Indexed: 07/05/2024]
Abstract
Cardiovascular diseases are currently the most common cause of death in developed countries. Due to lifestyle and environmental factors, this problem is only expected to increase in the future. Reactive oxygen species (ROS) are a key player in the onset of cardiovascular diseases but also have important functions in healthy cardiac tissue. Here, the interplay between ROS generation and cardiac mechanical forces is shown, and the state of the art and a perspective on future directions are discussed. To this end, an overview of what is currently known regarding ROS and mechanosignaling at a subcellular level is first given. There the role of ROS in mechanosignaling as well as the interplay between both factors in specific organelles is emphasized. The consequences at a larger scale across the population of heart cells are then discussed. Subsequently, the roles of ROS in embryogenesis, pathogenesis, and aging are further discussed, exemplifying some aspects of mechanoregulation. Finally, different models that are currently in use are discussed to study the topics above.
Collapse
Affiliation(s)
- Arturo Elias-Llumbet
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
- Laboratory of Genomic of Germ Cells, Biomedical Sciences Institute, Faculty of Medicine, University of Chile, Independencia, Santiago, 1027, Chile
| | - Rokshana Sharmin
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
| | | | - Romana Schirhagl
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
| | - Aldona Mzyk
- DTU Health Tech, Ørsteds Plads Bldg 345C, Kongens Lyngby, 2800, Denmark
| |
Collapse
|
7
|
Zheng JH, Zhu YH, Yang J, Ji PX, Zhao RK, Duan ZH, Yao HF, Jia QY, Yin YF, Hu LP, Li Q, Jiang SH, Huo YM, Liu W, Sun YW, Liu DJ. A CLIC1 network coordinates matrix stiffness and the Warburg effect to promote tumor growth in pancreatic cancer. Cell Rep 2024; 43:114633. [PMID: 39154343 DOI: 10.1016/j.celrep.2024.114633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/19/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) features substantial matrix stiffening and reprogrammed glucose metabolism, particularly the Warburg effect. However, the complex interplay between these traits and their impact on tumor advancement remains inadequately explored. Here, we integrated clinical, cellular, and bioinformatics approaches to explore the connection between matrix stiffness and the Warburg effect in PDAC, identifying CLIC1 as a key mediator. Elevated CLIC1 expression, induced by matrix stiffness through Wnt/β-catenin/TCF4 signaling, signifies poorer prognostic outcomes in PDAC. Functionally, CLIC1 serves as a catalyst for glycolytic metabolism, propelling tumor proliferation. Mechanistically, CLIC1 fortifies HIF1α stability by curbing hydroxylation via reactive oxygen species (ROS). Collectively, PDAC cells elevate CLIC1 levels in a matrix-stiffness-responsive manner, bolstering the Warburg effect to drive tumor growth via ROS/HIF1α signaling. Our insights highlight opportunities for targeted therapies that concurrently address matrix properties and metabolic rewiring, with CLIC1 emerging as a promising intervention point.
Collapse
Affiliation(s)
- Jia-Hao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yu-Heng Zhu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Jian Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Pei-Xuan Ji
- Shanghai Institute of Digestive Disease, Division of Gastroenterology and Hepatology, NHC Key Laboratory of Digestive Diseases, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, P.R. China
| | - Rui-Kang Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, P.R. China
| | - Zong-Hao Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hong-Fei Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Qin-Yuan Jia
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yi-Fan Yin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Li-Peng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Qing Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Yan-Miao Huo
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - Wei Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - Yong-Wei Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| | - De-Jun Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China; Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China.
| |
Collapse
|
8
|
Mialet-Perez J, Belaidi E. Interplay between hypoxia inducible Factor-1 and mitochondria in cardiac diseases. Free Radic Biol Med 2024; 221:13-22. [PMID: 38697490 DOI: 10.1016/j.freeradbiomed.2024.04.239] [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] [Received: 02/14/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Ischemic heart diseases and cardiomyopathies are characterized by hypoxia, energy starvation and mitochondrial dysfunction. HIF-1 acts as a cellular oxygen sensor, tuning the balance of metabolic and oxidative stress pathways to provide ATP and sustain cell survival. Acting on mitochondria, HIF-1 regulates different processes such as energy substrate utilization, oxidative phosphorylation and mitochondrial dynamics. In turn, mitochondrial homeostasis modifications impact HIF-1 activity. This underlies that HIF-1 and mitochondria are tightly interconnected to maintain cell homeostasis. Despite many evidences linking HIF-1 and mitochondria, the mechanistic insights are far from being understood, particularly in the context of cardiac diseases. Here, we explore the current understanding of how HIF-1, reactive oxygen species and cell metabolism are interconnected, with a specific focus on mitochondrial function and dynamics. We also discuss the divergent roles of HIF in acute and chronic cardiac diseases in order to highlight that HIF-1, mitochondria and oxidative stress interaction deserves to be deeply investigated. While the strategies aiming at stabilizing HIF-1 have provided beneficial effects in acute ischemic injury, some deleterious effects were observed during prolonged HIF-1 activation. Thus, deciphering the link between HIF-1 and mitochondria will help to optimize HIF-1 modulation and provide new therapeutic perspectives for the treatment of cardiovascular pathologies.
Collapse
Affiliation(s)
- Jeanne Mialet-Perez
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, France
| | - Elise Belaidi
- Univ. Lyon 1, Laboratory of Tissue Biology and Therapeutic Engineering, CNRS, LBTI UMR 5305, 69367, Lyon, France.
| |
Collapse
|
9
|
Du RR, Zhou JC, Qin WJ, Lu KZ, Duan XM, Yang YN, Yuan X, Li K, Zhang XW, Zhang PC. Fourteen new 2-benzylbenzofuran glycosides with cardioprotective activity from Heterosmilax yunnanensis. Bioorg Chem 2024; 143:107079. [PMID: 38185011 DOI: 10.1016/j.bioorg.2023.107079] [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: 09/05/2023] [Revised: 12/15/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
Fourteen new 2-benzylbenzofuran O-glycosides (1-13, 15) and one new key precursor, diarylacetone (14) were isolated from the roots of Heterosmilax yunnanensis Gagnep, which all have characteristic 2,3,4-O-trisubstituted benzyl. Their structures were elucidated by 1D and 2D NMR, HRESIMS, UV and IR. The isolated compounds were assessed for their cardioprotective activities and compounds 1, 3 and 6 could significantly improve cardiomyocytes viability. Moreover, the mechanistic study revealed that these three compounds could significantly decrease intracellular ROS levels and maintain mitochondrial homeostasis upon hypoxia inducement. Consequently, 1, 3 and 6 might serve as potential lead compounds to prevent myocardial ischemia.
Collapse
Affiliation(s)
- Rong-Rong Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ji-Chao Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wen-Jie Qin
- Beijing Zhendong Guangming Pharmaceutical Research Institute, Beijing 100085, China
| | - Kai-Zhou Lu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiu-Mei Duan
- Beijing Zhendong Guangming Pharmaceutical Research Institute, Beijing 100085, China
| | - Ya-Nan Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiang Yuan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Kun Li
- Shanxi Zhendong Pharmaceutical Co. Ltd, Changzhi 047100, China.
| | - Xiao-Wei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Pei-Cheng Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
10
|
Ai L, Li R, Cao Y, Liu Z, Niu X, Li Y. 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (Tempol) alleviates lung injury by inhibiting SIRT6-HIF-1α signaling pathway activation through the upregulation of miR-212-5p expression. Mol Biol Rep 2024; 51:129. [PMID: 38236324 DOI: 10.1007/s11033-023-09039-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 11/28/2023] [Indexed: 01/19/2024]
Abstract
OBJECTIVE Obstructive sleep apnea is closely related to oxidative stress. 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (Tempol) can scavenge reactive oxygen species (ROS) and ameliorate oxidative damage in the body. The mechanism by which Tempol alleviates chronic intermittent hypoxia-induced lung injury has rarely been reported. This study aimed to confirm the molecular mechanism by which Tempol alleviates lung injury. METHODS The levels of miR-212-5p and Sirtuin 6 (SIRT6) in injured lungs were analyzed using bioinformatics. In vitro, intermittent hypoxia (IH) treatment induced hypoxia in BEAS-2B cells and we established a model of chronic intermittent hypoxia (CIH) in mouse using a programmed hypoxia chamber. We used HE staining to observe the morphology of lung tissue, and the changes in lung fibers were observed by Masson staining. The levels of inflammatory factors in mouse serum were detected by ELISA, and the levels of the oxidative stress indicators GSH, MDA, SOD and ROS were detected using commercially available kits. Moreover, a real-time qPCR assay was used to detect miR-212-5p expression, and Western blotting was used to detect the levels of SIRT6, HIF-1α and apoptosis-related proteins. CCK-8 was used to detect cell proliferation. Subsequently, we used flow cytometry to detect cell apoptosis. Dual-luciferase gene reporters determine the on-target binding relationship of miR-212-5p and SIRT6. RESULTS SIRT6 was highly expressed in CIH-induced lung injury, as shown by bioinformatics analysis; however, miR-212-5p expression was decreased. Tempol promoted miR-212-5p expression, and the levels of SIRT6 and HIF-1α were inhibited. In BEAS-2B cells, Tempol also increased proliferation, inhibited apoptosis and inhibited oxidative stress in BEAS-2B cells under IH conditions. In BEAS-2B cells, these effects of Tempol were reversed after transfection with an miR-212-5p inhibitor. miR-212-5p targeted and negatively regulated the level of SIRT6 and overexpression of SIRT6 effectively reversed the enhanced influence of the miR-212-5p mimic on Tempol's antioxidant activity. Tempol effectively ameliorated lung injury in CIH mice and inhibited collagen deposition and inflammatory cell infiltration. Likewise, the therapeutic effect of Tempol could be effectively reversed by interference with the miR-212-5p inhibitor. CONCLUSION Inhibition of the SIRT6-HIF-1α signaling pathway could promote the effect of Tempol by upregulating the level of miR-212-5p, thereby alleviating the occurrence of lung injury and providing a new underlying target for the treatment of lung injury.
Collapse
Affiliation(s)
- Li Ai
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, No 374 Dianmian Road, Kunming, 650101, Yunnan, China
| | - Ran Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, No 374 Dianmian Road, Kunming, 650101, Yunnan, China
| | - Yu Cao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, No 374 Dianmian Road, Kunming, 650101, Yunnan, China
| | - Zhijuan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, No 374 Dianmian Road, Kunming, 650101, Yunnan, China
| | - Xiaoqun Niu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, No 374 Dianmian Road, Kunming, 650101, Yunnan, China
| | - Yongxia Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Kunming Medical University, No 374 Dianmian Road, Kunming, 650101, Yunnan, China.
| |
Collapse
|
11
|
Zhu Z, Wu Y, Qu L, Zou Y, Nie G, Xu S, Zhou Q, Zhang Y, Chen R. Evaluation of nocturnal apnea and airflow limitation as indicators for cognitive dysfunction in patients with chronic obstructive pulmonary disease/obstructive sleep apnea hypopnea syndrome overlap syndrome. Chron Respir Dis 2024; 21:14799731241236492. [PMID: 38411460 PMCID: PMC10901067 DOI: 10.1177/14799731241236492] [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] [Indexed: 02/28/2024] Open
Abstract
OBJECTIVE The aim of this study is to investigate how much intermittent hypoxemia and airflow limitation contribute to cognitive impairment in overlap syndrome (OS), which is the coexistence of two common diseases, obstructive sleep apnea hypopnea syndrome (OSAHS) and chronic obstructive pulmonary disease (COPD). METHODS We conducted a cross-sectional study of patients with OSAHS, COPD or OS, compared with normal controls, to determine the association between sleep apnea/pulmonary function-related indicators and cognitive dysfunction in individuals with OSAHS, COPD or OS. RESULTS A total of 157 participants were recruited. Both OSAHS and OS presented lower adjusted Montreal cognitive assessment (MoCA) scores compared with COPD group. In addition, the MoCA score was significantly lower in COPD group compared with control group. The incidence of cognitive impairment was 57.4% in OSAHS group, and 78% in OS group, which were significantly higher than COPD group (29%) and control group (8.8%). Furthermore, a broader range of cognitive domains were affected in OS group compared with OSAHS group. Elevated levels of oxygen desaturation index (ODI) and/or apnea hypopnea index (AHI) were positively correlated with increased Epworth sleeping scale (ESS) in OSAHS and OS. Forced vital capacity (FVC), forced expiratory volume in 1 s (FEV1) and peak expiratory flow (PEF) were positively correlated with cognitive scores in OSAHS but not in OS. Serum level of hypoxia-inducible factor-1α (HIF-1α) was significantly higher in OS. Logistic regression identified ODI as an independent risk factor for cognitive impairment in OS, while severity of snoring and PEF were independent risk factors in OSAHS. DISCUSSION This study revealed significant cognitive impairment in OS, OSAHS and COPD. Sleep-related indicators are warranted in OS patients for detection, differentiation and grading of cognitive impairment, whereas pulmonary functions are warranted in OSAHS patients for detection and early intervention of cognitive impairment.
Collapse
Affiliation(s)
- Zheng Zhu
- Department of Respiratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Pulmonary and Critical Care Medicine, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Yaohua Wu
- Department of Pulmonary and Critical Care Medicine, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Ling Qu
- Department of Pulmonary and Critical Care Medicine, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Ying Zou
- Department of Pulmonary and Critical Care Medicine, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Guozhong Nie
- Department of Pulmonary and Critical Care Medicine, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Shuguang Xu
- Department of Pulmonary and Critical Care Medicine, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Qixing Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Yunfeng Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Rui Chen
- Department of Respiratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Sleep Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Arnaud C, Billoir E, de Melo Junior AF, Pereira SA, O'Halloran KD, Monteiro EC. Chronic intermittent hypoxia-induced cardiovascular and renal dysfunction: from adaptation to maladaptation. J Physiol 2023; 601:5553-5577. [PMID: 37882783 DOI: 10.1113/jp284166] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
Chronic intermittent hypoxia (CIH) is the dominant pathological feature of human obstructive sleep apnoea (OSA), which is highly prevalent and associated with cardiovascular and renal diseases. CIH causes hypertension, centred on sympathetic nervous overactivity, which persists following removal of the CIH stimulus. Molecular mechanisms contributing to CIH-induced hypertension have been carefully delineated. However, there is a dearth of knowledge on the efficacy of interventions to ameliorate high blood pressure in established disease. CIH causes endothelial dysfunction, aberrant structural remodelling of vessels and accelerates atherosclerotic processes. Pro-inflammatory and pro-oxidant pathways converge on disrupted nitric oxide signalling driving vascular dysfunction. In addition, CIH has adverse effects on the myocardium, manifesting atrial fibrillation, and cardiac remodelling progressing to contractile dysfunction. Sympatho-vagal imbalance, oxidative stress, inflammation, dysregulated HIF-1α transcriptional responses and resultant pro-apoptotic ER stress, calcium dysregulation, and mitochondrial dysfunction conspire to drive myocardial injury and failure. CIH elaborates direct and indirect effects in the kidney that initially contribute to the development of hypertension and later to chronic kidney disease. CIH-induced morphological damage of the kidney is dependent on TLR4/NF-κB/NLRP3/caspase-1 inflammasome activation and associated pyroptosis. Emerging potential therapies related to the gut-kidney axis and blockade of aryl hydrocarbon receptors (AhR) are promising. Cardiorenal outcomes in response to intermittent hypoxia present along a continuum from adaptation to maladaptation and are dependent on the intensity and duration of exposure to intermittent hypoxia. This heterogeneity of OSA is relevant to therapeutic treatment options and we argue the need for better stratification of OSA phenotypes.
Collapse
Affiliation(s)
- Claire Arnaud
- Université Grenoble-Alpes INSERM U1300, Laboratoire HP2, Grenoble, France
| | - Emma Billoir
- Université Grenoble-Alpes INSERM U1300, Laboratoire HP2, Grenoble, France
| | | | - Sofia A Pereira
- iNOVA4Health, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Emilia C Monteiro
- iNOVA4Health, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
13
|
Liang Z, Zheng Z, Guo Q, Tian M, Yang J, Liu X, Zhu X, Liu S. The role of HIF-1α/HO-1 pathway in hippocampal neuronal ferroptosis in epilepsy. iScience 2023; 26:108098. [PMID: 37876811 PMCID: PMC10590818 DOI: 10.1016/j.isci.2023.108098] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/12/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023] Open
Abstract
Epilepsy, a common central nervous system disorder, remains an enigma in pathogenesis. Emerging consensus designates hippocampal neuronal injury as a cornerstone for epileptogenic foci, pivotal in epileptic genesis and progression. Ferroptosis, a regulated cell death modality hinging on iron, catalyzes lipid reactive oxygen species formation through iron and membrane polyunsaturated fatty acid interplay, culminating in oxidative cell death. This research investigates the role of hypoxia-inducible factor (HIF)-1α/heme oxygenase (HO)-1 in hippocampal neuron ferroptosis during epilepsy. Untargeted metabolomics exposes metabolite discrepancies between epilepsy patients and healthy individuals, unveiling escalated oxidative stress, heightened bilirubin, and augmented iron metabolism in epileptic blood. Enrichment analyses unveil active HIF-1 pathway in epileptic pathogenesis, reinforced by HIF-1α signaling perturbations in DisGeNET database. PTZ-kindled mice model confirms increased ferroptotic markers, oxidative stress, HIF-1α, and HO-1 in epilepsy. Study implicates HIF-1α/HO-1 potentially regulates hippocampal neuronal ferroptosis, iron metabolism, and oxidative stress, thereby promoting the propagation of epilepsy.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Zhaoshi Zheng
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qi Guo
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meng Tian
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Jing Yang
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Xiu Liu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Songyan Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Xu H, Li W, Chen J, Zhang P, Rong S, Tian J, Zhang Y, Li Y, Cui Z, Zhang Y. Associations between insomnia and large vessel occlusion acute ischemic stroke: An observational study. Clinics (Sao Paulo) 2023; 78:100297. [PMID: 37924679 PMCID: PMC10660002 DOI: 10.1016/j.clinsp.2023.100297] [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] [Received: 07/08/2023] [Revised: 09/22/2023] [Accepted: 10/05/2023] [Indexed: 11/06/2023] Open
Abstract
OBJECTIVES This study explored the association between insomnia and the clinical outcome of large vessel occlusion Acute Ischemic Stroke (AIS) and attempted to explore its potential mechanisms from the perspectives of inflammation and oxidative stress. METHODS AIS patients who underwent endovascular treatment for large vessel occlusion at Binzhou Central Hospital from 2018 to 2022 (n = 508) were included. Patients were divided into an insomnia group and a non-insomnia group. Insomnia was judged by self-reported Athens Insomnia Scale score. Regression analysis was used to compare the differences in the 24-hour and 7-day National Institutes of Health Stroke Scale (NIHSS) score, Early Neurological Deterioration (END), early adverse event incidence, 90-day prognosis and mortality, and serum biomarkers levels. RESULTS The incidence of insomnia in the study population was 39.6% (n = 144, insomnia group; n = 364, non-insomnia group). Compared with the non-insomnia group, a worse prognosis outcome (63% vs. 49%, adjusted rate ratio: 1.8, 95% Confidence Interval: 1.2-3.7; p = 0.016), higher 24-h and 7-day NIHSS score (17 [9-36] vs. 13 [5-20]; p = 0.024, and 11 [4‒24) vs. 8 [2‒14]; p = 0.031, respectively), higher END (24% vs. 15%, p = 0.022), and higher incidence of adverse events were observed in the insomnia group (79% vs. 59%, p = 0.010). The 90-day mortality was higher in the insomnia group than that in the non-insomnia group (22% vs. 17%), however, such a difference was not statistically significant. CONCLUSION Insomnia is closely related to the clinical outcome of AIS with large vessel occlusion, and inflammation and oxidative stress mechanisms may be involved.
Collapse
Affiliation(s)
- Huali Xu
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China; Department of Neurology, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong Province, China
| | - Weili Li
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jiahao Chen
- Department of Neurobiology, Capital Medical University, Beijing, China
| | - Piao Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Siming Rong
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jinping Tian
- Department of Information, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong Province, China
| | - Yuqian Zhang
- Department of Neurology, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong Province, China
| | - Yanke Li
- Department of Neurology, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong Province, China
| | - Zhenzhen Cui
- Department of Neurology, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong Province, China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
15
|
Breuillard C, Moulin S, Bouyon S, Couchet M, Moinard C, Belaidi E. Chronic intermittent hypoxia due to obstructive sleep apnea slightly alters nutritional status: a pre-clinical study. Front Nutr 2023; 10:1250529. [PMID: 37964925 PMCID: PMC10642957 DOI: 10.3389/fnut.2023.1250529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is associated with chronic intermittent hypoxia (cIH) that causes disturbances in glucose and lipid metabolism. Animals exposed to cIH show lower body weight and food intake, but the protein-energy metabolism has never been investigated. Here, to address the gap, we studied the impact of cIH on nutritional status in rats. A total of 24 male Wistar rats were randomized into 3 groups (n = 8): a control group (Ctrl), a cIH group (cIH) exposed to cIH (30 s 21-30 s 5% fraction of inspired oxygen, 8 h per day, for 14 days), and a pair-fed group (PF) exposed to normoxia with food intake adjusted to the intake of the cIH group rats with anorexia. Body weight and food intake were measured throughout the study. After 14 days, the rats were euthanized, the organs were collected, weighed, and the liver, intestine mucosa, and muscles were snap-frozen to measure total protein content. Food intake was decreased in the cIH group. Body weight was significantly lower in the cIH group only (-11%, p < 0.05). Thymus and liver weight as well as EDL protein content tended to be lower in the cIH group than in the Ctrl and PF groups. Jejunum and ileum mucosa protein contents were lower in the cIH group compared to the PF group. cIH causes a slight impairment of nutritional status and immunity. This pre-clinical work argues for greater consideration of malnutrition in care for OSAS patients. Further studies are warranted to devise an adequate nutritional strategy.
Collapse
Affiliation(s)
- Charlotte Breuillard
- Université Grenoble Alpes, Inserm, Laboratory of Fundamental and Applied Bioenergetics (LBFA), Grenoble, France
| | - Sophie Moulin
- Université Grenoble Alpes, Inserm, Laboratory HP2, Grenoble, France
| | - Sophie Bouyon
- Université Grenoble Alpes, Inserm, Laboratory HP2, Grenoble, France
| | - Morgane Couchet
- Université Grenoble Alpes, Inserm, Laboratory of Fundamental and Applied Bioenergetics (LBFA), Grenoble, France
| | - Christophe Moinard
- Université Grenoble Alpes, Inserm, Laboratory of Fundamental and Applied Bioenergetics (LBFA), Grenoble, France
| | - Elise Belaidi
- Université Grenoble Alpes, Inserm, Laboratory HP2, Grenoble, France
| |
Collapse
|
16
|
Lee J, Lee SA, Son SH, Choi JA, Nguyen TD, Kim J, Son D, Song CH. Impaired mitophagy induces antimicrobial responses in macrophages infected with Mycobacterium tuberculosis. Cell Biosci 2023; 13:158. [PMID: 37649112 PMCID: PMC10470153 DOI: 10.1186/s13578-023-01107-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 08/18/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Mitophagy, mitochondrial selective autophagy, plays a pivotal role in the maintenance of cellular homeostasis in response to cellular stress. However, the role of mitophagy in macrophages during infection has not been elucidated. To determine whether mitophagy regulates intracellular pathogen survival, macrophages were infected with Mycobacterium tuberculosis (Mtb), an intracellular bacterium. RESULTS We showed that Mtb-infected macrophages induced mitophagy through BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3) activation. In contrast, BNIP3-deficient macrophages failed to induce mitophagy, resulting in reduced mitochondrial membrane potential in response to Mtb infection. Moreover, the accumulation of damaged mitochondria due to BNIP3 deficiency generated higher levels of mitochondrial reactive oxygen species (mROS) compared to the control, suppressing the intracellular survival of Mtb. We observed that siBNIP3 suppressed intracellular Mtb in mice lungs. CONCLUSION We found that BNIP3 plays a critical role in the regulation of mitophagy during Mtb infection. The inhibition of mitophagy suppresses Mtb growth in macrophages through increased mROS production. Therefore, BNIP3 might be a novel therapeutic target for tuberculosis treatment.
Collapse
Affiliation(s)
- Junghwan Lee
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea
| | - Seong-Ahn Lee
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Sang-Hun Son
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Ji-Ae Choi
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea
| | - Tam Doan Nguyen
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Jaewhan Kim
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Doyi Son
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea
| | - Chang-Hwa Song
- Department of Microbiology, Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa‑ro, Jung‑gu, Daejeon, 35015, South Korea.
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon, 35015, South Korea.
- Translational Immunology Institute, Chungnam National University, Daejeon, 34134, South Korea.
| |
Collapse
|
17
|
Gharib E, Veilleux V, Boudreau LH, Pichaud N, Robichaud GA. Platelet-derived microparticles provoke chronic lymphocytic leukemia malignancy through metabolic reprogramming. Front Immunol 2023; 14:1207631. [PMID: 37441073 PMCID: PMC10333545 DOI: 10.3389/fimmu.2023.1207631] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/30/2023] [Indexed: 07/15/2023] Open
Abstract
Background It is well established that inflammation and platelets promote multiple processes of cancer malignancy. Recently, platelets have received attention for their role in carcinogenesis through the production of microvesicles or platelet-derived microparticles (PMPs), which transfer their biological content to cancer cells. We have previously characterized a new subpopulation of these microparticles (termed mito-microparticles), which package functional mitochondria. The potential of mitochondria transfer to cancer cells is particularly impactful as many aspects of mitochondrial biology (i.e., cell growth, apoptosis inhibition, and drug resistance) coincide with cancer hallmarks and disease progression. These metabolic aspects are particularly notable in chronic lymphocytic leukemia (CLL), which is characterized by a relentless accumulation of proliferating, immunologically dysfunctional, mature B-lymphocytes that fail to undergo apoptosis. The present study aimed to investigate the role of PMPs on CLL metabolic plasticity leading to cancer cell phenotypic changes. Methods CLL cell lines were co-incubated with different concentrations of human PMPs, and their impact on cell proliferation, mitochondrial DNA copy number, OCR level, ATP production, and ROS content was evaluated. Essential genes involved in metabolic-reprogramming were identified using the bioinformatics tools, examined between patients with early and advanced CLL stages, and then validated in PMP-recipient CLLs. Finally, the impact of the induced metabolic reprogramming on CLLs' growth, survival, mobility, and invasiveness was tested against anti-cancer drugs Cytarabine, Venetoclax, and Plumbagin. Results The data demonstrated the potency of PMPs in inducing tumoral growth and invasiveness in CLLs through mitochondrial internalization and OXPHOS stimulation which was in line with metabolic shift reported in CLL patients from early to advanced stages. This metabolic rewiring also improved CLL cells' resistance to Cytarabine, Venetoclax, and Plumbagin chemo drugs. Conclusion Altogether, these findings depict a new platelet-mediated pathway of cancer pathogenesis. We also highlight the impact of PMPs in CLL metabolic reprogramming and disease progression.
Collapse
Affiliation(s)
- Ehsan Gharib
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Vanessa Veilleux
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Luc H Boudreau
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Nicolas Pichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Gilles A Robichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| |
Collapse
|
18
|
Xin Y, Zhao L, Peng R. HIF-1 signaling: an emerging mechanism for mitochondrial dynamics. J Physiol Biochem 2023:10.1007/s13105-023-00966-0. [PMID: 37178248 DOI: 10.1007/s13105-023-00966-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/02/2023] [Indexed: 05/15/2023]
Abstract
A growing emphasis has been paid to the function of mitochondria in tumors, neurodegenerative disorders (NDs), and cardiovascular diseases. Mitochondria are oxygen-sensitive organelles whose function depends on their structural basis. Mitochondrial dynamics are critical in regulating the structure. Mitochondrial dynamics include fission, fusion, motility, cristae remodeling, and mitophagy. These processes could alter mitochondrial morphology, number, as well as distribution, to regulate complicated cellular signaling processes like metabolism. Meanwhile, they also could modulate cell proliferation and apoptosis. The initiation and progression of several diseases, such as tumors, NDs, cardiovascular disease, were all interrelated with mitochondrial dynamics. HIF-1 is a nuclear protein presented as heterodimers, and its transcriptional activity is triggered by hypoxia. It plays an important role in numerous physiological processes including the development of cardiovascular system, immune system, and cartilage. Additionally, it could evoke compensatory responses in cells during hypoxia through upstream and downstream signaling networks. Moreover, the alteration of oxygen level is a pivotal factor to promote mitochondrial dynamics and HIF-1 activation. HIF-1α might be a promising target for modulating mitochondrial dynamics to develop therapeutic approaches for NDs, immunological diseases, and other related diseases. Here, we reviewed the research progress of mitochondrial dynamics and the potential regulatory mechanism of HIF-1 in mitochondrial dynamics.
Collapse
Affiliation(s)
- Yu Xin
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Li Zhao
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| | - Ruiyun Peng
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| |
Collapse
|
19
|
Minoves M, Hazane-Puch F, Moriondo G, Boutin-Paradis A, Lemarié E, Pépin JL, Godin-Ribuot D, Briançon-Marjollet A. Differential Impact of Intermittent vs. Sustained Hypoxia on HIF-1, VEGF and Proliferation of HepG2 Cells. Int J Mol Sci 2023; 24:ijms24086875. [PMID: 37108039 PMCID: PMC10139223 DOI: 10.3390/ijms24086875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
Obstructive sleep apnea (OSA) is an emerging risk factor for cancer occurrence and progression, mainly mediated by intermittent hypoxia (IH). Systemic IH, a main landmark of OSA, and local sustained hypoxia (SH), a classical feature at the core of tumors, may act separately or synergistically on tumor cells. Our aim was to compare the respective consequences of intermittent and sustained hypoxia on HIF-1, endothelin-1 and VEGF expression and on cell proliferation and migration in HepG2 liver tumor cells. Wound healing, spheroid expansion, proliferation and migration were evaluated in HepG2 cells following IH or SH exposure. The HIF-1α, endothelin-1 and VEGF protein levels and/or mRNA expression were assessed, as were the effects of HIF-1 (acriflavine), endothelin-1 (macitentan) and VEGF (pazopanib) inhibition. Both SH and IH stimulated wound healing, spheroid expansion and proliferation of HepG2 cells. HIF-1 and VEGF, but not endothelin-1, expression increased with IH exposure but not with SH exposure. Acriflavine prevented the effects of both IH and SH, and pazopanib blocked those of IH but not those of SH. Macitentan had no impact. Thus, IH and SH stimulate hepatic cancer cell proliferation via distinct signaling pathways that may act synergistically in OSA patients with cancer, leading to enhanced tumor progression.
Collapse
Affiliation(s)
- Mélanie Minoves
- INSERM U1300, HP2 Laboratory, CHU Grenoble Alpes, University Grenoble Alpes, 38042 Grenoble, France
| | | | - Giorgia Moriondo
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Antoine Boutin-Paradis
- INSERM U1300, HP2 Laboratory, CHU Grenoble Alpes, University Grenoble Alpes, 38042 Grenoble, France
| | - Emeline Lemarié
- INSERM U1300, HP2 Laboratory, CHU Grenoble Alpes, University Grenoble Alpes, 38042 Grenoble, France
| | - Jean-Louis Pépin
- INSERM U1300, HP2 Laboratory, CHU Grenoble Alpes, University Grenoble Alpes, 38042 Grenoble, France
| | - Diane Godin-Ribuot
- INSERM U1300, HP2 Laboratory, CHU Grenoble Alpes, University Grenoble Alpes, 38042 Grenoble, France
| | - Anne Briançon-Marjollet
- INSERM U1300, HP2 Laboratory, CHU Grenoble Alpes, University Grenoble Alpes, 38042 Grenoble, France
| |
Collapse
|
20
|
Xu Y, Hu T, Ding H, Yuan Y, Chen R. miR-485-5p alleviates obstructive sleep apnea syndrome with hypertension by inhibiting PI3K/AKT signaling pathway via downregulating HIF3A expression. Sleep Breath 2023; 27:109-119. [PMID: 35132534 DOI: 10.1007/s11325-022-02580-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The obstructive sleep apnea syndrome (OSAS) is a risk factor for hypertension. MiRNAs are key regulators in hypertension. However, their roles in OSAS with hypertension remain unclear. This study aimed to clarify the function and mechanism of miRNAs in OSAS with hypertension. METHODS A chronic intermittent hypoxia (CIH) model was established to simulate OSAS with hypertension. The proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) were assessed by CCK-8 and wound healing assays. qRT-PCR, Western blot, and immunofluorescence staining were used to detect the expression of HIF3A and PI3K/AKT pathway. Hematoxylin-eosin and Masson staining were used to detect the histopathological changes of pulmonary arterial hypertension (PAH). The regulatory function of miR-485-5p to HIF3A was assessed by dual luciferase reporter gene assay. RESULTS MiR-485-5p was significantly downregulated in the rats with OSAS-induced PAH. MiR-485-5p alleviated proliferation and migration of PASMCs in vitro and ameliorated OSAS-induced PAH in vivo. HIF3A could act as a target of miR-485-5p. HIF3A downregulation regulated by miR-485-5p alleviated proliferation and migration of PASMCs in vitro and ameliorated OSAS-induced PAH in vivo. In addition, we found that PI3K/AKT signaling was significantly inhibited in OSAS-induced PAH. CONCLUSION MiR-485-5p alleviates OSAS with hypertension by inhibiting the PI3K/Akt pathway via downregulating HIF3A expression through the PI3K/AKT pathway. These findings suggest that miR-485-5p has the potential for treating OSAS-associated hypertension.
Collapse
Affiliation(s)
- Yan Xu
- Department of Respiratory, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Jiangsu, 215004, Suzhou, China.,Department of Respiratory, The Affiliated Hospital of Yangzhou University, No. 45 Taizhou Road, Jiangsu, 225001, Yangzhou, China
| | - Tao Hu
- Department of Respiratory, The Affiliated Hospital of Yangzhou University, No. 45 Taizhou Road, Jiangsu, 225001, Yangzhou, China
| | - Huizhen Ding
- Department of Respiratory, The Affiliated Hospital of Yangzhou University, No. 45 Taizhou Road, Jiangsu, 225001, Yangzhou, China
| | - Yi Yuan
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Jiangsu, 212013, Zhenjiang , China
| | - Rui Chen
- Department of Respiratory, The Second Affiliated Hospital of Soochow University, No. 1055 Sanxiang Road, Jiangsu, 215004, Suzhou, China.
| |
Collapse
|
21
|
L-Citrulline Supplementation Reduces Blood Pressure and Myocardial Infarct Size under Chronic Intermittent Hypoxia, a Major Feature of Sleep Apnea Syndrome. Antioxidants (Basel) 2022; 11:antiox11122326. [PMID: 36552534 PMCID: PMC9774116 DOI: 10.3390/antiox11122326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
Intermittent hypoxia (IH) is a landmark of obstructive sleep apnea (OSA) at the core of the cardiovascular consequences of OSA. IH triggers oxidative stress, a major underlying mechanism for elevated blood pressure (BP) and increased infarct size. L-citrulline is an amino acid that has been demonstrated to be protective of the cardiovascular system and exert pleiotropic effects. Therefore, we tested the impact of citrulline supplementation on IH-induced increase in BP and infarct size. Four groups of rats exposed to normoxia (N) or IH [14 days (d), 8 h/day, 30 s-O2 21%/30 s-O2 5%] and were supplemented or not with citrulline (1 g·kg-1·d-1). After 14 d, BP was measured, and hearts were submitted to global ischemia-reperfusion to measure infarct size. Histological and biochemical analyses were conducted on hearts and aorta to assess oxidative stress. Citrulline significantly reduced BP (-9.92%) and infarct size (-18.22%) under IH only. In the aorta, citrulline supplementation significantly decreased superoxide anion and nitrotyrosine levels under IH and abolished the IH-induced decrease in nitrite. Citrulline supplementation significantly decreased myocardial superoxide anion levels and xanthine oxidase enzyme activity under IH. Citrulline shows a cardioprotective capacity by limiting IH-induced pro-oxidant activity. Our results suggest that citrulline might represent a new pharmacological strategy in OSA patients with high cardiovascular risk.
Collapse
|
22
|
Limberg JK, Baker SE, Petersen-Jones HG, Guo W, Huang A, Jensen MD, Singh P. Endothelin-1 as a novel target for the prevention of metabolic dysfunction with intermittent hypoxia in male participants. Am J Physiol Regul Integr Comp Physiol 2022; 323:R351-R362. [PMID: 35816718 PMCID: PMC9423726 DOI: 10.1152/ajpregu.00301.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/22/2022]
Abstract
We examined the effect of intermittent hypoxia (IH, a hallmark feature of sleep apnea) on adipose tissue lipolysis and the role of endothelin-1 (ET-1) in this response. We hypothesized that IH can increase ET-1 secretion and plasma free fatty acid (FFA) concentrations. We further hypothesized that inhibition of ET-1 receptor activation with bosentan could prevent any IH-mediated increase in FFA. To test this hypothesis, 16 healthy male participants (32 ± 5 yr, 26 ± 2 kg/m2) were exposed to 30 min of IH in the absence (control) and presence of bosentan (62.5 mg oral twice daily for 3 days prior). Arterial blood samples for ET-1, epinephrine, and FFA concentrations, as well as abdominal subcutaneous adipose tissue biopsies (to assess transcription of cellular receptors/proteins involved in lipolysis), were collected. Additional proof-of-concept studies were conducted in vitro using primary differentiated human white preadipocytes (HWPs). We show that IH increased circulating ET-1, epinephrine, and FFA (P < 0.05). Bosentan treatment reduced plasma epinephrine concentrations and blunted IH-mediated increases in FFA (P < 0.01). In adipose tissue, bosentan had no effect on cellular receptors and proteins involved in lipolysis (P > 0.05). ET-1 treatment did not directly induce lipolysis in differentiated HWP. In conclusion, IH increases plasma ET-1 and FFA concentrations. Inhibition of ET-1 receptors with bosentan attenuates the FFA increase in response to IH. Based on a lack of a direct effect of ET-1 in HWP, we speculate the effect of bosentan on circulating FFA in vivo may be secondary to its ability to reduce sympathoadrenal tone.
Collapse
Affiliation(s)
- Jacqueline K Limberg
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Sarah E Baker
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota
| | | | - Winston Guo
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota
| | - An Huang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Prachi Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| |
Collapse
|
23
|
Chronic intermittent hypoxia, a hallmark of obstructive sleep apnea, promotes 4T1 breast cancer development through endothelin-1 receptors. Sci Rep 2022; 12:12916. [PMID: 35902610 PMCID: PMC9334573 DOI: 10.1038/s41598-022-15541-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/24/2022] [Indexed: 11/08/2022] Open
Abstract
The association between obstructive sleep apnea (OSA) and cancer is still debated and data are scarce regarding the link between OSA and breast cancer progression. Since conclusive epidemiological studies require large sample sizes and sufficient duration of exposure before incident cancer occurrence, basic science studies represent the most promising approach to appropriately address the topic. Here we assessed the impact of intermittent hypoxia (IH), the major hallmark of OSA, on the development of breast cancer and explored the specific involvement of the endothelin signaling pathway. Original in vitro and in vivo models were used where 3D-spheroids or cultures of murine 4T1 breast cancer cells were submitted to IH cycles, and nude NMRI mice, orthotopically implanted with 4T1 cells, were submitted to chronic IH exposure before and after implantation. The role of the endothelin-1 in promoting cancer cell development was investigated using the dual endothelin receptor antagonist, macitentan. In vitro exposure to IH significantly increased 4T1 cell proliferation and migration. Meta-analysis of 4 independent in vivo experiments showed that chronic IH exposure promoted tumor growth, assessed by caliper measurement (overall standardized mean difference: 1.00 [0.45-1.55], p < 0.001), bioluminescence imaging (1.65 [0.59-2.71]; p < 0.01) and tumor weight (0.86 [0.31-1.41], p < 0.01), and enhanced metastatic pulmonary expansion (0.77 [0.12-1.42]; p = 0.01). Both in vitro and in vivo tumor-promoting effects of IH were reversed by macitentan. Overall, these findings demonstrate that chronic intermittent hypoxia exposure promotes breast cancer growth and malignancy and that dual endothelin receptor blockade prevents intermittent hypoxia-induced tumor development.
Collapse
|
24
|
Intermittent Hypoxia-Induced Cardiomyocyte Death Is Mediated by HIF-1 Dependent MAM Disruption. Antioxidants (Basel) 2022; 11:antiox11081462. [PMID: 36009181 PMCID: PMC9405320 DOI: 10.3390/antiox11081462] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
Rationale: Intermittent hypoxia (IH) is one of the main features of sleep-disordered breathing (SDB). Recent findings indicate that hypoxia inducible factor-1 (HIF-1) promotes cardiomyocytes apoptosis during chronic IH, but the mechanisms involved remain to be elucidated. Here, we hypothesize that IH-induced ER stress is associated with mitochondria-associated ER membrane (MAM) alteration and mitochondrial dysfunction, through HIF-1 activation. Methods: Right atrial appendage biopsies from patients with and without SDB were used to determine HIF-1α, Grp78 and CHOP expressions. Wild-type and HIF-1α+/− mice were exposed to normoxia (N) or IH (21–5% O2, 60 cycles/h, 8 h/day) for 21 days. Expressions of HIF-1α, Grp78 and CHOP, and apoptosis, were measured by Western blot and immunochemistry. In isolated cardiomyocytes, we examined structural integrity of MAM by proximity ligation assay and their function by measuring ER-to-mitochondria Ca2+ transfer by confocal microscopy. Finally, we measured mitochondrial respiration using oxygraphy and calcium retention capacity (CRC) by spectrofluorometry. MAM structure was also investigated in H9C2 cells incubated with 1 mM CoCl2, a potent HIF-1α inducer. Results: In human atrial biopsies and mice, IH induced HIF-1 activation, ER stress and apoptosis. IH disrupted MAM, altered Ca2+ homeostasis, mitochondrial respiration and CRC. Importantly, IH had no effect in HIF-1α+/− mice. Similar to what observed under IH, HIF-1α overexpression was associated with MAM alteration in H9C2. Conclusion: IH-induced ER stress, MAM alterations and mitochondrial dysfunction were mediated by HIF-1; all these intermediate mechanisms ultimately inducing cardiomyocyte apoptosis. This suggests that HIF-1 modulation might limit the deleterious cardiac effects of SDB.
Collapse
|
25
|
Liu T, Wang G, Zhang X, Liu X, Liang Z, Ren X, Yan D, Zhang W. B serum proteome profiles revealed dysregulated proteins and mechanisms associated with insomnia patients: A preliminary study. Front Integr Neurosci 2022; 16:936955. [PMID: 35958162 PMCID: PMC9360785 DOI: 10.3389/fnint.2022.936955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background Insomnia is a clinical problem of significant public health importance; however, the underlying pathogenesis of this disorder is not comprehensively understood. Methods To identify potential treatment targets and unfold one of the gaps that were involved in insomnia pathological mechanisms, we employed a tandem mass tag-based (TMT) quantitative proteomics technology to detect differentially expressed proteins (DEPs) in serum from patients with insomnia and controls. DEPs were further analyzed by bioinformatics platforms. In addition, parallel reaction monitoring (PRM) was used to verify the TMT results. Results Patients with insomnia had poorer sleep quality compared with healthy controls. A total of 106 DEPs were identified among patients with insomnia and controls. They were mainly enriched in immune and inflammation-related biological functions and signaling pathways. Using the protein–protein interaction network, we screened the 10 most connected proteins as key DEPs. We predicted that four key DEPs were subject to targeted regulation by natural compounds of herbs. Eight key DEPs were validated using PRM in an additional 15 patients with insomnia and 15 controls, and the results also supported the experimental findings. Conclusion We identified aberrantly expressed proteins in insomnia that may be involved in the immune-inflammatory response. The 10 key DEPs screened may be potential targets for insomnia, especially FN1, EGF, HP, and IGF1. The results of this study will broaden our understanding of the pathological mechanisms of insomnia and provide more possibilities for pharmacotherapy.
Collapse
Affiliation(s)
- Tao Liu
- Traditional Chinese Medicine Hospital Affiliated with Xinjiang Medical University, Ürümqi, China
- Postdoctoral Workstation of Traditional Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China
| | - Guanying Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xingping Zhang
- Traditional Chinese Medicine Hospital Affiliated with Xinjiang Medical University, Ürümqi, China
- College of Traditional Chinese Medicine, Xinjiang Medical University, Ürümqi, China
- *Correspondence: Xingping Zhang,
| | - Xin Liu
- College of Traditional Chinese Medicine, Xinjiang Medical University, Ürümqi, China
| | - Zhengting Liang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Ürümqi, China
| | - Xiaojuan Ren
- Xinjiang Urumqi Traditional Chinese Medicine Hospital, Ürümqi, China
| | - Deqi Yan
- College of Traditional Chinese Medicine, Xinjiang Medical University, Ürümqi, China
| | - Wenhui Zhang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Ürümqi, China
| |
Collapse
|
26
|
Hypoxia signaling in human health and diseases: implications and prospects for therapeutics. Signal Transduct Target Ther 2022; 7:218. [PMID: 35798726 PMCID: PMC9261907 DOI: 10.1038/s41392-022-01080-1] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Molecular oxygen (O2) is essential for most biological reactions in mammalian cells. When the intracellular oxygen content decreases, it is called hypoxia. The process of hypoxia is linked to several biological processes, including pathogenic microbe infection, metabolic adaptation, cancer, acute and chronic diseases, and other stress responses. The mechanism underlying cells respond to oxygen changes to mediate subsequent signal response is the central question during hypoxia. Hypoxia-inducible factors (HIFs) sense hypoxia to regulate the expressions of a series of downstream genes expression, which participate in multiple processes including cell metabolism, cell growth/death, cell proliferation, glycolysis, immune response, microbe infection, tumorigenesis, and metastasis. Importantly, hypoxia signaling also interacts with other cellular pathways, such as phosphoinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) signaling, nuclear factor kappa-B (NF-κB) pathway, extracellular signal-regulated kinases (ERK) signaling, and endoplasmic reticulum (ER) stress. This paper systematically reviews the mechanisms of hypoxia signaling activation, the control of HIF signaling, and the function of HIF signaling in human health and diseases. In addition, the therapeutic targets involved in HIF signaling to balance health and diseases are summarized and highlighted, which would provide novel strategies for the design and development of therapeutic drugs.
Collapse
|
27
|
Zhou W, Zhao Z, Yu Z, Hou Y, Keerthiga R, Fu A. Mitochondrial transplantation therapy inhibits the proliferation of malignant hepatocellular carcinoma and its mechanism. Mitochondrion 2022; 65:11-22. [DOI: 10.1016/j.mito.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/11/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023]
|
28
|
Gaucher J, Vial G, Montellier E, Guellerin M, Bouyon S, Lemarie E, Pelloux V, Bertrand A, Pernet-Gallay K, Lamarche F, Borel AL, Arnaud C, Belaidi E, Clément K, Godin Ribuot D, Aron-Wisnewsky J, Pépin JL. Intermittent Hypoxia Rewires the Liver Transcriptome and Fires up Fatty Acids Usage for Mitochondrial Respiration. Front Med (Lausanne) 2022; 9:829979. [PMID: 35252260 PMCID: PMC8894659 DOI: 10.3389/fmed.2022.829979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/21/2022] [Indexed: 11/15/2022] Open
Abstract
Sleep Apnea Syndrome (SAS) is one of the most common chronic diseases, affecting nearly one billion people worldwide. The repetitive occurrence of abnormal respiratory events generates cyclical desaturation-reoxygenation sequences known as intermittent hypoxia (IH). Among SAS metabolic sequelae, it has been established by experimental and clinical studies that SAS is an independent risk factor for the development and progression of non-alcoholic fatty liver disease (NAFLD). The principal goal of this study was to decrypt the molecular mechanisms at the onset of IH-mediated liver injury. To address this question, we used a unique mouse model of SAS exposed to IH, employed unbiased high-throughput transcriptomics and computed network analysis. This led us to examine hepatic mitochondrial ultrastructure and function using electron microscopy, high-resolution respirometry and flux analysis in isolated mitochondria. Transcriptomics and network analysis revealed that IH reprograms Nuclear Respiratory Factor- (NRF-) dependent gene expression and showed that mitochondria play a central role. We thus demonstrated that IH boosts the oxidative capacity from fatty acids of liver mitochondria. Lastly, the unbalance between oxidative stress and antioxidant defense is tied to an increase in hepatic ROS production and DNA damage during IH. We provide a comprehensive analysis of liver metabolism during IH and reveal the key role of the mitochondria at the origin of development of liver disease. These findings contribute to the understanding of the mechanisms underlying NAFLD development and progression during SAS and provide a rationale for novel therapeutic targets and biomarker discovery.
Collapse
Affiliation(s)
- Jonathan Gaucher
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France,*Correspondence: Jonathan Gaucher
| | - Guillaume Vial
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France
| | - Emilie Montellier
- CNRS 5309, INSERM U1209, Institute for Advanced Biosciences, University Grenoble-Alpes, Grenoble, France
| | - Maëlle Guellerin
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France
| | - Sophie Bouyon
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France
| | - Emeline Lemarie
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France
| | - Véronique Pelloux
- Nutrition and Obesities, Systemic Approaches, NutriOmics, Laboratory, Sorbonne University, Paris, France,Nutrition Department, CRNH Ile de France, Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
| | - Anne Bertrand
- INSERM U1216, Grenoble Institute of Neurosciences, University Grenoble-Alpes, Grenoble, France
| | - Karin Pernet-Gallay
- INSERM U1216, Grenoble Institute of Neurosciences, University Grenoble-Alpes, Grenoble, France
| | - Frederic Lamarche
- Laboratory of Fundamental and Applied Bioenergetics (LBFA), INSERM U1055, University Grenoble Alpes, Grenoble, France
| | - Anne-Laure Borel
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France
| | - Claire Arnaud
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France
| | - Elise Belaidi
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France
| | - Karine Clément
- Nutrition and Obesities, Systemic Approaches, NutriOmics, Laboratory, Sorbonne University, Paris, France,Nutrition Department, CRNH Ile de France, Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
| | - Diane Godin Ribuot
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France
| | - Judith Aron-Wisnewsky
- Nutrition and Obesities, Systemic Approaches, NutriOmics, Laboratory, Sorbonne University, Paris, France,Nutrition Department, CRNH Ile de France, Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Paris, France
| | - Jean-Louis Pépin
- Hypoxia and PhysioPathology (HP2) Laboratory, INSERM U1300, CHU Grenoble-Alpes, University Grenoble-Alpes, Grenoble, France,Jean-Louis Pépin
| |
Collapse
|
29
|
Zhang X, Chen Z, Zuo S, Sun H, Li X, Lu X, Xing Z, Chen M, Liu J, Xiao G, He Y. Endothelin-A Receptor Antagonist Alleviates Allergic Airway Inflammation via the Inhibition of ILC2 Function. Front Immunol 2022; 13:835953. [PMID: 35222426 PMCID: PMC8873101 DOI: 10.3389/fimmu.2022.835953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Allergic airway inflammation is a universal airway disease that is driven by hyperresponsiveness to inhaled allergens. Group 2 innate lymphoid cells (ILC2s) produce copious amounts of type 2 cytokines, which lead to allergic airway inflammation. Here, we discovered that both peripheral blood of human and mouse lung ILC2s express the endothelin-A receptor (ETAR), and the expression level of ETAR was dramatically induced upon interleukin-33 (IL-33) treatment. Subsequently, both preventive and therapeutic effects of BQ123, an ETAR antagonist, on allergic airway inflammation were observed, which were associated with decreased proliferation and type 2 cytokine productions by ILC2s. Furthermore, ILC2s from BQ123 treatment were found to be functionally impaired in response to an interleukin IL-33 challenged. And BQ123 treatment also affected the phosphorylation level of the extracellular signal-regulated kinase (ERK), as well as the level of GATA binding protein 3 (GATA3) in activated ILC2s. Interestingly, after BQ123 treatment, both mouse and human ILC2s in vitro exhibited decreased function and downregulation of ERK signaling and GATA3 stability. These observations imply that ETAR is an important regulator of ILC2 function and may be involved in ILC2-driven pulmonary inflammation. Therefore, blocking ETAR may be a promising therapeutic strategy for allergic airway inflammation.
Collapse
Affiliation(s)
- Xiaogang Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ziyang Chen
- Department of Neurosurgery Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Shaowen Zuo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hengbiao Sun
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Xinyao Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhe Xing
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Meiqi Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingping Liu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Gang Xiao
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Yumei He
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University, Guangzhou, China
| |
Collapse
|
30
|
Effects of Hypoxic Environment on Periodontal Tissue through the ROS/TXNIP/NLRP3 Inflammasome Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7690960. [PMID: 35083332 PMCID: PMC8786523 DOI: 10.1155/2022/7690960] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 11/30/2021] [Accepted: 12/08/2021] [Indexed: 11/23/2022]
Abstract
There is low evidence for the possible association between obstructive sleep apnea-hypopnea syndrome (OSAHS) and periodontitis, necessitating further research. This study was aimed at investigating this association. For the in vitro study, 8-day-old Wistar rats were divided into the unilateral nasal obstruction group (UNO) and the sham surgery group (SHAM). Rats in the former group were subjected to UNO by cauterization of the external nostril at the age of 8 days. Immunofluorescence analysis, quantitative real-time polymerase chain reaction, and western blot were performed to assess the expression of thioredoxin-interacting protein (TXNIP), NLR family pyrin domain-containing 3 (NLRP3) inflammasome-associated factors, and interleukin-1β (IL-1β). Throughout the experimental period, the weights of rats in the two groups were similar. The mRNA and protein expression of TXNIP and IL-1β was significantly higher in the UNO than in the SHAM groups. Compared with SHAM, NLRP3 inflammasome-associated factors were activated in the UNO group. For the in vitro study, a cellular hypoxia model was established by treating human periodontal ligament cells (HPDLCs) with cobalt chloride. The studies showed that hypoxia can induce an excessive production and accumulation of reactive oxygen species (ROS) in HPDLCs and induce abnormal expression of TNXIP, NLRP3 inflammasome-related factors, and IL-1β. More importantly, N-acetylcysteine induced reduction of ROS in HPDLCs, downregulated TXNIP expression, inhibited the expression and aggregation of NLRP3 inflammasome-related factors, and abrogated the inflammatory response to hypoxia. In conclusion, hypoxia-induced ROS can activate the TXNIP/NLRP3 inflammasome signaling pathway in response to oxidative stress, resulting in the increased expression of inflammatory factors in HPDLCs. Our findings provide evidence for the mechanism underlying the possible association between OSAHS and periodontal disease.
Collapse
|
31
|
Ochoa SV, Otero L, Aristizabal-Pachon AF, Hinostroza F, Carvacho I, Torres YP. Hypoxic Regulation of the Large-Conductance, Calcium and Voltage-Activated Potassium Channel, BK. Front Physiol 2022; 12:780206. [PMID: 35002762 PMCID: PMC8727448 DOI: 10.3389/fphys.2021.780206] [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: 09/20/2021] [Accepted: 11/15/2021] [Indexed: 11/15/2022] Open
Abstract
Hypoxia is a condition characterized by a reduction of cellular oxygen levels derived from alterations in oxygen balance. Hypoxic events trigger changes in cell-signaling cascades, oxidative stress, activation of pro-inflammatory molecules, and growth factors, influencing the activity of various ion channel families and leading to diverse cardiovascular diseases such as myocardial infarction, ischemic stroke, and hypertension. The large-conductance, calcium and voltage-activated potassium channel (BK) has a central role in the mechanism of oxygen (O2) sensing and its activity has been related to the hypoxic response. BK channels are ubiquitously expressed, and they are composed by the pore-forming α subunit and the regulatory subunits β (β1–β4), γ (γ1–γ4), and LINGO1. The modification of biophysical properties of BK channels by β subunits underly a myriad of physiological function of these proteins. Hypoxia induces tissue-specific modifications of BK channel α and β subunits expression. Moreover, hypoxia modifies channel activation kinetics and voltage and/or calcium dependence. The reported effects on the BK channel properties are associated with events such as the increase of reactive oxygen species (ROS) production, increases of intracellular Calcium ([Ca2+]i), the regulation by Hypoxia-inducible factor 1α (HIF-1α), and the interaction with hemeproteins. Bronchial asthma, chronic obstructive pulmonary diseases (COPD), and obstructive sleep apnea (OSA), among others, can provoke hypoxia. Untreated OSA patients showed a decrease in BK-β1 subunit mRNA levels and high arterial tension. Treatment with continuous positive airway pressure (CPAP) upregulated β1 subunit mRNA level, decreased arterial pressures, and improved endothelial function coupled with a reduction in morbidity and mortality associated with OSA. These reports suggest that the BK channel has a role in the response involved in hypoxia-associated hypertension derived from OSA. Thus, this review aims to describe the mechanisms involved in the BK channel activation after a hypoxic stimulus and their relationship with disorders like OSA. A deep understanding of the molecular mechanism involved in hypoxic response may help in the therapeutic approaches to treat the pathological processes associated with diseases involving cellular hypoxia.
Collapse
Affiliation(s)
- Sara V Ochoa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Semillero de Investigación, Biofísica y Fisiología de Canales Iónicos, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Liliana Otero
- Center of Dental Research Dentistry Faculty, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Fernando Hinostroza
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile.,Centro de Investigación de Estudios Avanzados del Maule, CIEAM, Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile.,Facultad de Ciencias de la Salud, Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Universidad Católica del Maule, Talca, Chile
| | - Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile
| | - Yolima P Torres
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Semillero de Investigación, Biofísica y Fisiología de Canales Iónicos, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
32
|
Nozawa S, Urushihata K, Machida R, Hanaoka M. The positional characteristics of patients with obstructive sleep apnea: a single institute retrospective study in Japan. Sleep Biol Rhythms 2022; 20:115-121. [PMID: 38469073 PMCID: PMC10899958 DOI: 10.1007/s41105-021-00347-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/05/2021] [Indexed: 10/20/2022]
Abstract
Obstructive sleep apnea (OSA) causes sleep-disordered breathing (SDB) due to upper airway obstruction. The severity of OSA changes with position during sleep. Patients with marked significant improvement in apnea-hypopnea index (AHI) level by sleep position change are defined as ''positional patients'' (PP), while those without improvement are defined as ''non-positional patients'' (NPP). We aimed to verify their clinical characteristics. Between May 2008 and May 2020, 237 patients with OSA were registered retrospectively and classified into two groups: PP (n = 158) and NPP (n = 79). The differences in clinical background and full-night polysomnography (PSG) between the two groups were observed. A logistic regression analysis was conducted to identify the risk factors for severe AHI (≥ 30 events/h) in the PP group. Moreover, confounding factor-adjusted sub-analysis by a propensity score matching method was performed, and the PSG results were compared between the two groups. The PP group was older than the NPP group. Furthermore, the PP group had lower body mass index (BMI) and AHI levels compared with the NPP group. The independent risk factors for severe AHI in the PP group were BMI and being in the supine position during sleep. The PP group had a significantly milder nocturnal hypoxemia despite having no significant difference in AHI levels between the two groups. The characteristics of PP were old age, low BMI, and low AHI associated with milder nocturnal hypoxemia. Moreover, they were less likely to worsen with nocturnal hypoxemia compared with NPP having similar severity.
Collapse
Affiliation(s)
- Shuhei Nozawa
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Kazuhisa Urushihata
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Ryosuke Machida
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Masayuki Hanaoka
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| |
Collapse
|
33
|
Ren R, Zhang Y, Yang L, Somers VK, Covassin N, Tang X. Association Between Arousals During Sleep and Hypertension Among Patients With Obstructive Sleep Apnea. J Am Heart Assoc 2021; 11:e022141. [PMID: 34970921 PMCID: PMC9075207 DOI: 10.1161/jaha.121.022141] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Sleep fragmentation induced by repetitive arousals is a hallmark of obstructive sleep apnea (OSA). Sleep fragmentation has been linked to hypertension in community‐based studies, but it is unclear if this association is manifest in OSA. We aimed to explore whether frequent arousals from sleep modify the relationship between OSA and prevalent hypertension. Methods and Results A total of 10 102 patients with OSA and 1614 primary snorers were included in the study. Hypertension was defined on either direct blood pressure measures or diagnosis by a physician. Spontaneous, respiratory, and movement arousals were derived by polysomnography. Logistic regression models were used to estimate the associations between arousals and prevalent hypertension in patients with OSA and primary snorers. For every 10‐unit increase of total arousal index, odds of hypertension significantly increased in both the total sample (odds ratio [OR], 1.08; 95% CI, 1.03–1.14; P=0.002) and patients with OSA (OR, 1.10; 95% CI, 1.04–1.16; P<0.001), but not in the primary snoring group. Total arousal index was significantly associated with systolic blood pressure and diastolic blood pressure in the total sample (β=0.05 and β=0.06; P<0.001) and in patients with (β=0.05 and β=0.06; P<0.01), but not in primary snorers. In addition, a greater influence of respiratory events with arousals than respiratory events without arousals on blood pressure in OSA was also noted. Results were independent of confounders, including apnea‐hypopnea index and nocturnal hypoxemia. Conclusions We conclude that repetitive arousals from sleep are independently associated with prevalent hypertension in patients with OSA.
Collapse
Affiliation(s)
- Rong Ren
- Department of Respiratory and Critical Care Medicine Sleep Medicine Center Translational Neuroscience Center State Key Laboratory West China HospitalSichuan University Chengdu China
| | - Ye Zhang
- Department of Respiratory and Critical Care Medicine Sleep Medicine Center Translational Neuroscience Center State Key Laboratory West China HospitalSichuan University Chengdu China
| | - Linghui Yang
- Department of Respiratory and Critical Care Medicine Sleep Medicine Center Translational Neuroscience Center State Key Laboratory West China HospitalSichuan University Chengdu China
| | - Virend K Somers
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN
| | - Naima Covassin
- Department of Cardiovascular Medicine Mayo Clinic Rochester MN
| | - Xiangdong Tang
- Department of Respiratory and Critical Care Medicine Sleep Medicine Center Translational Neuroscience Center State Key Laboratory West China HospitalSichuan University Chengdu China
| |
Collapse
|
34
|
Chen XY, Wang JQ, Cheng SJ, Wang Y, Deng MY, Yu T, Wang HY, Zhou WJ. Diazoxide Post-conditioning Activates the HIF-1/HRE Pathway to Induce Myocardial Protection in Hypoxic/Reoxygenated Cardiomyocytes. Front Cardiovasc Med 2021; 8:711465. [PMID: 34938777 PMCID: PMC8687117 DOI: 10.3389/fcvm.2021.711465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
Background: Previous studies have shown that diazoxide can protect against myocardial ischemia-reperfusion injury (MIRI). The intranuclear hypoxia-inducible factor-1 (HIF-1)/hypoxia-response element (HRE) pathway has been shown to withstand cellular damage caused by MIRI. It remains unclear whether diazoxide post-conditioning is correlated with the HIF-1/HRE pathway in protective effect on cardiomyocytes. Methods: An isolated cardiomyocyte model of hypoxia-reoxygenation injury was established. Prior to reoxygenation, cardiomyocytes underwent post-conditioning treatment by diazoxide, and 5-hydroxydecanoate (5-HD), N-(2-mercaptopropionyl)-glycine (MPG), or dimethyloxallyl glycine (DMOG) followed by diazoxide. At the end of reoxygenation, ultrastructural morphology; mitochondrial membrane potential; interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), reactive oxygen species (ROS), and HIF-1α levels; and downstream gene mRNA and protein levels were analyzed to elucidate the protective mechanism of diazoxide post-conditioning. Results: Diazoxide post-conditioning enabled activation of the HIF-1/HRE pathway to induce myocardial protection. When the mitoKATP channel was inhibited and ROS cleared, the diazoxide effect was eliminated. DMOG was able to reverse the effect of ROS absence to restore the diazoxide effect. MitoKATP and ROS in the early reoxygenation phase were key to activation of the HIF-1/HRE pathway. Conclusion: Diazoxide post-conditioning promotes opening of the mitoKATP channel to generate a moderate ROS level that activates the HIF-1/HRE pathway and subsequently induces myocardial protection.
Collapse
Affiliation(s)
- Xi-Yuan Chen
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
- Department of Anesthesiology, The Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Jia-Qi Wang
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Si-Jing Cheng
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Yan Wang
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Meng-Yuan Deng
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Tian Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Hai-Ying Wang
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Wen-Jing Zhou
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Guizhou, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| |
Collapse
|
35
|
Lai S, Chen L, Zhan P, Lin G, Lin H, Huang H, Chen Q. Circular RNA Expression Profiles and Bioinformatic Analysis in Mouse Models of Obstructive Sleep Apnea-Induced Cardiac Injury: Novel Insights Into Pathogenesis. Front Cell Dev Biol 2021; 9:767283. [PMID: 34820383 PMCID: PMC8606653 DOI: 10.3389/fcell.2021.767283] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 01/22/2023] Open
Abstract
Circular RNAs (circRNAs) participate in the development of various kinds of diseases. However, the function and roles of circRNAs in obstructive sleep apnea (OSA)-induced cardiovascular disease remain poorly understood. Therefore, we sought to explore the circRNA expression profiles and predict their functions in OSA-induced cardiac injury with the use of bioinformatics analysis. The model of OSA was established in mouse treated by chronic intermittent hypoxia (CIH) exposure. Then, we screened the circRNA profile using circRNA microarray. By comparing circRNA expression in three matched pairs of CIH-treated cardiac tissues and controls, differentially expressed circRNAs were identified in the CIH groups. Comparison of the selected circRNAs expression levels was performed between qRT-PCR and microarray. Meanwhile, we employed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses to predict the functions of these selected circRNAs. Finally, we constructed a circRNA-miRNA-mRNA network based on the target prediction. It was found that a total of 124 circRNAs were differentially expressed in CIH-treated cardiac tissues (p ≤ 0.05, fold-change ≥ 1.5). Among them, 23 circRNAs were significantly down-regulated, and the other 101 were up-regulated. Then, ten circRNAs were randomly selected to validate the reliability of the microarray results by using qRT-PCR. Next, we conducted the GO and KEGG pathway analysis to explore the parental genes functions of differentially expressed circRNA. Finally, two significantly differentially expressed circRNAs (mmu_circRNA_014309 and mmu_circRNA_21856) were further selected to create a circRNA-miRNA-mRNA regulation network. Our study did first reveal that the differentially expressed circRNAs played a vital role in the pathogenesis of OSA-induced cardiac damage. Thus, our findings bring us closer to unraveling the pathophysiologic mechanisms and eliciting novel therapeutic targets for the treatment of OSA-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Suxian Lai
- Department of Neonatology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, China
| | - Lijun Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Pingyun Zhan
- Department of Cardiology, Haidu Hospital, Quanzhou, China
| | - Guofu Lin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hai Lin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Huibin Huang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qingshi Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
36
|
Li X, Hu R, Ren X, He J. Interleukin-8 concentrations in obstructive sleep apnea syndrome: a systematic review and meta-analysis. Bioengineered 2021; 12:10666-10681. [PMID: 34747311 PMCID: PMC8809978 DOI: 10.1080/21655979.2021.2001203] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Interleukin (IL)-8 has been shown to play an important role in obstructive sleep apnea syndrome (OSAS). However, its role in OSAS development is still controversial. This meta-analysis was to explore the correlation between interleukin (IL)-8 concentration and OSAS. Database (from the inception to July 2021) searches on PubMed, Web of Science, Medline, EMBASE, and Cochrane Library were conducted for studies analyzing the correlation between IL-8 concentration and OSAS, regardless of the language of publication. Standardized mean difference (SMD) and 95% confidence intervals (CI) were used to analyze any prospective association between IL-8 concentration and OSAS. A total of 25 eligible studies, including 2301 participants and 1123 controls, were included in this meta-analysis. The included studies evaluating the association between serum IL-8 concentration and OSAS indicated that adults and children with OSAS had elevated serum concentrations of IL-8 compared with controls (SMD = 0.997, 95% CI = 0.437–1.517, P < 0.001; SMD = 0.431, 95% CI = 0.104–0.759, P = 0.01). Categorization of the study population into subgroups according to body mass index, apnea–hypopnea index (AHI), ethnicity, and sample size also showed that individuals with OSAS had elevated serum concentrations of IL-8 compared with controls. Additionally, the results demonstrated that the higher the AHI, higher was the IL-8 concentration. Similar results were observed in the literature on the association between plasma IL-8 concentration and OSAS. This meta-analysis verified that compared with controls, children and adults with OSAS have significantly elevated IL-8 concentrations.
Collapse
Affiliation(s)
- Xiaoyan Li
- Clinical Medical College of Chengdu Medical College, Chengdu, Sichuan, 610500, China.,Department of Endocrinology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Rong Hu
- Clinical Medical College of Chengdu Medical College, Chengdu, Sichuan, 610500, China.,Medical department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Xinyi Ren
- Clinical Medical College of Chengdu Medical College, Chengdu, Sichuan, 610500, China.,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Jie He
- Clinical Medical College of Chengdu Medical College, Chengdu, Sichuan, 610500, China.,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
| |
Collapse
|
37
|
Xiong M, Zhao Y, Mo H, Yang H, Yue F, Hu K. Intermittent hypoxia increases ROS/HIF-1α 'related oxidative stress and inflammation and worsens bleomycin-induced pulmonary fibrosis in adult male C57BL/6J mice. Int Immunopharmacol 2021; 100:108165. [PMID: 34560512 DOI: 10.1016/j.intimp.2021.108165] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/05/2021] [Accepted: 09/14/2021] [Indexed: 10/20/2022]
Abstract
Obstructive sleep apnea (OSA) has been increasingly recognized as a risk factor for idiopathic pulmonary fibrosis (IPF). The intermittent hypoxia (IH) and re-oxygenation of OSA contribute to poor outcomes of IPF, however, the potential mechanism remains unknown. Here, C57BL/6J mice were administered intratracheal injection of Bleomycin (BLM) or saline and then exposed to IH (alternating cycles of FiO2 21% for 60S and FiO2 10% for 30 s, 40 cycles/hour, 8 h/day) to mimic OSA or intermittent air (IA) for 4 days, 8 days or 21 days. This study found that pulmonary fibrosis in BLM + IH treated mice was more severe than that in BLM + IA group at day 8 and 21, but not observed at day 4. Besides, the expression of reactive oxygen species (ROS) and hypoxia inducible factor-1α (HIF-1α),which are related to hypoxia reduced oxidative stress and inflammation, were higher in BLM + IH treated mice than BLM + IA mice, and IH increased these indexes in BLM treated mice from day 4 to day 21. Interestingly, a positive linear correlation between the HIF-1α expression and hydroxyproline (HYP) content was observed. We further found some inflammatory cells in bronchoalveolar lavage fluid were increased significantly from day 4 to 21, and there was a positive correlation between inflammation and ROS expression. Our results demonstrated that IH aggravated BLM-induced pulmonary fibrosis, and ROS/HIF-1α related oxidative stress and inflammation involved. The increase of ROS/HIF-1α related oxidative stress and inflammation may be a potential mechanism of moderate-to-severe OSA in potentiating pulmonary fibrosis of IPF, which warrants further study.
Collapse
Affiliation(s)
- Mengqing Xiong
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Yang Zhao
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Huaheng Mo
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Haizhen Yang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Fang Yue
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
38
|
阿布利克木·依明, 阿不拉江·托合提, 艾力根·阿不都热依木, 唐 亮. [Analysis of propensity score matching between inflammatory factor levels and gene polymorphisms and susceptibility to obstructive sleep apnea]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2021; 35:728-732. [PMID: 34304535 PMCID: PMC10127807 DOI: 10.13201/j.issn.2096-7993.2021.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 11/12/2022]
Abstract
Objective:To explore the correlation between the levels of inflammatory factors and their gene polymorphisms and the susceptibility to obstructive sleep apnea(OSA). Methods:Seventy-nine patients who were diagnosed with OSA in People's Hospital of Xinjiang Uygur Autonomous Region from October 2018 to December 2020 were selected as the study group, and 104 healthy adults who received physical examination during the same period were selected as the control group. After collecting the clinical data of the two groups of patients, the two groups of patients were matched by propensity scores. Then, the serum IL-6, IL-8 and TNF-α levels and the peripheral blood IL-6, IL-8 and TNF-α gene polymorphism analysis and comparison were performed on the included matched patients. Results:Before matching, the age, proportion of males, BMI, neck circumference, smoking history, SBP and DBP of the study group were higher than those of the control group, and the difference was statistically significant(P<0.05). After the two groups of patients were matched by propensity scores, a total of 67 patients were successfully matched. The covariates between the two groups were all balanced after matching, and the balance of the PSM covariates was significantly improved(P>0.05). After matching, the fasting blood glucose, C-reactive protein, IL-6, IL-8, TNF-α, AHI and Ts90% of the study group were significantly higher than those of the control group, and the difference was statistically significant(P<0.05). Hardy-Weinberg balance verifies that the gene frequencies of each group are in accordance with the genetic balance rule(P>0.05); after matching, the distribution of IL-6(rs1800795) and TNF-α(rs1800629) genotypes between the two groups is compared, and the difference is statistically significant scientific significance; the frequency of IL-6(rs1800795) allele C and the frequency of TNF-α(rs1800629) allele A in the study group were significantly higher than those in the control group, and the difference was statistically significant(P<0.05). Conclusion:The levels of inflammatory factors TNF-α and IL-6 and their gene polymorphisms are significantly related to the susceptibility to OSA.
Collapse
Affiliation(s)
- 阿布利克木·依明
- 新疆维吾尔自治区人民医院耳鼻喉诊疗中心(乌鲁木齐,830000)Otorhinolaryngology Clinic, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - 阿不拉江·托合提
- 新疆维吾尔自治区人民医院耳鼻喉诊疗中心(乌鲁木齐,830000)Otorhinolaryngology Clinic, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - 艾力根·阿不都热依木
- 新疆维吾尔自治区人民医院耳鼻喉诊疗中心(乌鲁木齐,830000)Otorhinolaryngology Clinic, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - 亮 唐
- 新疆维吾尔自治区人民医院耳鼻喉诊疗中心(乌鲁木齐,830000)Otorhinolaryngology Clinic, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| |
Collapse
|
39
|
Kao TI, Chen PJ, Wang YH, Tseng HH, Chang SH, Wu TS, Yang SH, Lee YT, Hwang TL. Bletinib ameliorates neutrophilic inflammation and lung injury by inhibiting Src family kinase phosphorylation and activity. Br J Pharmacol 2021; 178:4069-4084. [PMID: 34131920 PMCID: PMC8518616 DOI: 10.1111/bph.15597] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/07/2021] [Accepted: 04/29/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil overactivation is crucial in the pathogenesis of acute lung injury (ALI). Bletinib (3,3'-dihydroxy-2',6'-bis(p-hydroxybenzyl)-5-methoxybibenzyl), a natural bibenzyl, extracted from the Bletilla plant, exhibits anti-inflammatory, antibacterial, and antimitotic effects. In this study, we evaluated the therapeutic effects of bletinib in human neutrophilic inflammation and LPS-mediated ALI in mice. EXPERIMENTAL APPROACH In human neutrophils activated with the formyl peptide (fMLP), we assessed integrin expression, superoxide anion production, degranulation, neutrophil extracellular trap (NET) formation, and adhesion through flow cytometry, spectrophotometry, and immunofluorescence microscopy. Immunoblotting was used to measure phosphorylation of Src family kinases (SFKs) and downstream proteins. Finally, a LPS-induced ALI model in male BALB/c mice was used to investigate the potential therapeutic effects of bletinib treatment. KEY RESULTS In activated human neutrophils, bletinib reduced degranulation, respiratory burst, NET formation, adhesion, migration, and integrin expression; suppressed the enzymic activity of SFKs, including Src, Lyn, Fgr, and Hck; and inhibited the phosphorylation of SFKs as well as Vav and Bruton's tyrosine kinase (Btk). In mice with ALI, the pulmonary sections demonstrated considerable amelioration of prominent inflammatory changes, such as haemorrhage, pulmonary oedema, and neutrophil infiltration, after bletinib treatment. CONCLUSION AND IMPLICATIONS Bletinib regulates neutrophilic inflammation by inhibiting the SFK-Btk-Vav pathway. Bletinib ameliorates LPS-induced ALI in mice. Further biochemical optimisation of bletinib may be a promising strategy for the development of novel therapeutic agents for inflammatory diseases.
Collapse
Affiliation(s)
- Ting-I Kao
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Jen Chen
- Department of Cosmetic Science, Providence University, Taichung, Taiwan
| | - Yi-Hsuan Wang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Hui Tseng
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Hsin Chang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tian-Shung Wu
- Department of Chemistry, National Cheng Kung University, Tainan, Taiwan
| | - Sien-Hung Yang
- Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Yen-Tung Lee
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Cosmetic Science, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Chinese Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Biomedical Sciences and Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
40
|
Mallet RT, Burtscher J, Richalet JP, Millet GP, Burtscher M. Impact of High Altitude on Cardiovascular Health: Current Perspectives. Vasc Health Risk Manag 2021; 17:317-335. [PMID: 34135590 PMCID: PMC8197622 DOI: 10.2147/vhrm.s294121] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Globally, about 400 million people reside at terrestrial altitudes above 1500 m, and more than 100 million lowlanders visit mountainous areas above 2500 m annually. The interactions between the low barometric pressure and partial pressure of O2, climate, individual genetic, lifestyle and socio-economic factors, as well as adaptation and acclimatization processes at high elevations are extremely complex. It is challenging to decipher the effects of these myriad factors on the cardiovascular health in high altitude residents, and even more so in those ascending to high altitudes with or without preexisting diseases. This review aims to interpret epidemiological observations in high-altitude populations; present and discuss cardiovascular responses to acute and subacute high-altitude exposure in general and more specifically in people with preexisting cardiovascular diseases; the relations between cardiovascular pathologies and neurodegenerative diseases at altitude; the effects of high-altitude exercise; and the putative cardioprotective mechanisms of hypobaric hypoxia.
Collapse
Affiliation(s)
- Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
- Institute of Sport Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
| | - Jean-Paul Richalet
- Laboratoire Hypoxie & Poumon, UMR Inserm U1272, Université Sorbonne Paris Nord 13, Bobigny Cedex, F-93017, France
| | - Gregoire P Millet
- Department of Biomedical Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
- Institute of Sport Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, A-6020, Austria
- Austrian Society for Alpine and High-Altitude Medicine, Mieming, Austria
| |
Collapse
|
41
|
Li Z, Zhao H, Wang J. Metabolism and Chronic Inflammation: The Links Between Chronic Heart Failure and Comorbidities. Front Cardiovasc Med 2021; 8:650278. [PMID: 34026868 PMCID: PMC8131678 DOI: 10.3389/fcvm.2021.650278] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) patients often suffer from multiple comorbidities, such as diabetes, atrial fibrillation, depression, chronic obstructive pulmonary disease, and chronic kidney disease. The coexistance of comorbidities usually leads to multi morbidity and poor prognosis. Treatments for HF patients with multi morbidity are still an unmet clinical need, and finding an effective therapy strategy is of great value. HF can lead to comorbidity, and in return, comorbidity may promote the progression of HF, creating a vicious cycle. This reciprocal correlation indicates there may be some common causes and biological mechanisms. Metabolism remodeling and chronic inflammation play a vital role in the pathophysiological processes of HF and comorbidities, indicating metabolism and inflammation may be the links between HF and comorbidities. In this review, we comprehensively discuss the major underlying mechanisms and therapeutic implications for comorbidities of HF. We first summarize the potential role of metabolism and inflammation in HF. Then, we give an overview of the linkage between common comorbidities and HF, from the perspective of epidemiological evidence to the underlying metabolism and inflammation mechanisms. Moreover, with the help of bioinformatics, we summarize the shared risk factors, signal pathways, and therapeutic targets between HF and comorbidities. Metabolic syndrome, aging, deleterious lifestyles (sedentary behavior, poor dietary patterns, smoking, etc.), and other risk factors common to HF and comorbidities are all associated with common mechanisms. Impaired mitochondrial biogenesis, autophagy, insulin resistance, and oxidative stress, are among the major mechanisms of both HF and comorbidities. Gene enrichment analysis showed the PI3K/AKT pathway may probably play a central role in multi morbidity. Additionally, drug targets common to HF and several common comorbidities were found by network analysis. Such analysis has already been instrumental in drug repurposing to treat HF and comorbidity. And the result suggests sodium-glucose transporter-2 (SGLT-2) inhibitors, IL-1β inhibitors, and metformin may be promising drugs for repurposing to treat multi morbidity. We propose that targeting the metabolic and inflammatory pathways that are common to HF and comorbidities may provide a promising therapeutic strategy.
Collapse
Affiliation(s)
- Zhiwei Li
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Hongmei Zhao
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
42
|
Basmadjian OM, Occhieppo VB, Marchese NA, Silvero C MJ, Becerra MC, Baiardi G, Bregonzio C. Amphetamine Induces Oxidative Stress, Glial Activation and Transient Angiogenesis in Prefrontal Cortex via AT 1-R. Front Pharmacol 2021; 12:647747. [PMID: 34012397 PMCID: PMC8126693 DOI: 10.3389/fphar.2021.647747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/30/2021] [Indexed: 01/21/2023] Open
Abstract
Background: Amphetamine (AMPH) alters neurons, glia and microvessels, which affects neurovascular unit coupling, leading to disruption in brain functions such as attention and working memory. Oxidative stress plays a crucial role in these alterations. The angiotensin type I receptors (AT1-R) mediate deleterious effects, such as oxidative/inflammatory responses, endothelial dysfunction, neuronal oxidative damage, alterations that overlap with those observed from AMPH exposure. Aims: The aim of this study was to evaluate the AT1-R role in AMPH-induced oxidative stress and glial and vascular alterations in the prefrontal cortex (PFC). Furthermore, we aimed to evaluate the involvement of AT1-R in the AMPH-induced short-term memory and working memory deficit. Methods: Male Wistar rats were repeatedly administered with the AT1-R blocker candesartan (CAND) and AMPH. Acute oxidative stress in the PFC was evaluated immediately after the last AMPH administration by determining lipid and protein peroxidation. After 21 off-drug days, long-lasting alterations in the glia, microvessel architecture and to cognitive tasks were evaluated by GFAP, CD11b and von Willebrand immunostaining and by short-term and working memory assessment. Results: AMPH induced acute oxidative stress, long-lasting glial reactivity in the PFC and a working memory deficit that were prevented by AT1-R blockade pretreatment. Moreover, AMPH induces transient angiogenesis in PFC via AT1-R. AMPH did not affect short-term memory. Conclusion: Our results support the protective role of AT1-R blockade in AMPH-induced oxidative stress, transient angiogenesis and long-lasting glial activation, preserving working memory performance.
Collapse
Affiliation(s)
- Osvaldo M Basmadjian
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria B Occhieppo
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Natalia A Marchese
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M Jazmin Silvero C
- Instituto Multidisciplinario de Biología Vegetal (IMBIV-CONICET) Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Cecilia Becerra
- Instituto Multidisciplinario de Biología Vegetal (IMBIV-CONICET) Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gustavo Baiardi
- Laboratorio de Neurofarmacología, (IIBYT-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina.,Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Córdoba, Argentina
| | - Claudia Bregonzio
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
43
|
Chen J, Lin S, Zeng Y. An Update on Obstructive Sleep Apnea for Atherosclerosis: Mechanism, Diagnosis, and Treatment. Front Cardiovasc Med 2021; 8:647071. [PMID: 33898538 PMCID: PMC8060459 DOI: 10.3389/fcvm.2021.647071] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
The occurrence and development of atherosclerosis could be influenced by intermittent hypoxia. Obstructive sleep apnea (OSA), characterized by intermittent hypoxia, is world-wide prevalence with increasing morbidity and mortality rates. Researches remain focused on the study of its mechanism and improvement of diagnosis and treatment. However, the underlying mechanism is complex, and the best practice for OSA diagnosis and treatment considering atherosclerosis and related cardiovascular diseases is still debatable. In this review, we provided an update on research in OSA in the last 5 years with regard to atherosclerosis. The processes of inflammation, oxidative stress, autonomic nervous system activation, vascular dysfunction, platelet activation, metabolite dysfunction, small molecule RNA regulation, and the cardioprotective occurrence was discussed. Additionally, improved diagnosis such as, the utilized of portable device, and treatment especially with inconsistent results in continuous positive airway pressure and mandibular advancement devices were illustrated in detail. Therefore, further fundamental and clinical research should be carried out for a better understanding the deep interaction between OSA and atherosclerosis, as well as the suggestion of newer diagnostic and treatment options.
Collapse
Affiliation(s)
- Jin Chen
- Clinical Center for Molecular Diagnosis and Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yiming Zeng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
44
|
Huang X, Zhao X, Zhu K, Ding S, Shao B. Sodium dehydroacetate exposure decreases locomotor persistence and hypoxia tolerance in zebrafish. ENVIRONMENTAL RESEARCH 2021; 195:110276. [PMID: 33131684 DOI: 10.1016/j.envres.2020.110276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/13/2020] [Accepted: 09/27/2020] [Indexed: 06/11/2023]
Abstract
Environmental exposure to sodium dehydroacetate (DHA-S) is inevitable as DHA-S is a high-volume preservative widely used in cosmetics, processed foods and personal care products. DHA-S is absorbed rapidly when administered orally or on the skin and generally considered to be safe and well tolerated. However, DHA-S has recently been reported to induce weight loss and allergic contact dermatitis, yet little is known about how DHA-S affect the related biological processes. Here, we characterize the biological effects of DHA-S on zebrafish model by directly waterborne exposure. Zebrafish is susceptible to DHA-S exposure at early developmental stage. DHA-S decreased the hatch rate and locomotor persistence of zebrafish, and eventually induced lethality during the continuous exposure at relatively low concentrations of commonly addition. Acute DHA-S exposure decreased respiration capacity in larval zebrafish, promoted the expression of HIF-1α (hypoxia-inducible factor-1α) and caused rapid adult zebrafish death in 30 h. We further demonstrated that DHA-S inhibited the activity of succinate dehydrogenase (SDH) inducing respiratory chain interruption, energy deficiency and organic acids accumulation. These results suggest that the approved DHA-S may pose serious environmental/ecological pressures on the aquatic animal's migration.
Collapse
Affiliation(s)
- Xiaoyong Huang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China; Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Centers for Disease Control and Preventative Medical Research, Beijing, 100013, China
| | - Xiaole Zhao
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Kui Zhu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Shuangyang Ding
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China.
| | - Bing Shao
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Centers for Disease Control and Preventative Medical Research, Beijing, 100013, China.
| |
Collapse
|
45
|
Pourbagher-Shahri AM, Farkhondeh T, Ashrafizadeh M, Talebi M, Samargahndian S. Curcumin and cardiovascular diseases: Focus on cellular targets and cascades. Biomed Pharmacother 2021; 136:111214. [PMID: 33450488 DOI: 10.1016/j.biopha.2020.111214] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are one of the leading causes of the most considerable mortality globally, and it has been tried to find the molecular mechanisms and design new drugs that triggered the molecular target. Curcumin is the main ingredient of Curcuma longa (turmeric) that has been used in traditional medicine for treating several diseases for years. Numerous investigations have indicated the beneficial effect of Curcumin in modulating multiple signaling pathways involved in oxidative stress, inflammation, apoptosis, and proliferation. The cardiovascular protective effects of Curcumin against CVDs have been indicated in several studies. In the current review study, we provided novel information on Curcumin's protective effects against various CVDs and potential molecular signaling targets of Curcumin. Nonetheless, more studies should be performed to discover the exact molecular target of Curcumin against CVDs.
Collapse
Affiliation(s)
| | - Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), Birjand, Iran; Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Marjan Talebi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, 19968 35115, Iran
| | - Saeed Samargahndian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
46
|
Oxidative stress assessment and its relationship with the prevalence of atherogenic risk in patients with type 2 diabetes. J Diabetes Metab Disord 2021; 20:583-590. [PMID: 34178854 DOI: 10.1007/s40200-021-00785-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/20/2021] [Indexed: 12/27/2022]
Abstract
Objectives During diabetes, prolonged hyperglycemia is characterized by the generation of free radicals via multiple mechanisms leading to various diabetic complications including cardiovascular diseases. This study aims to determine the relationship between a deregulation of the oxidative state in type 2 diabetes patients and the prevalence of atheroma plate formation. Methods This research was carried out at the Bouguerra Boulaares hospital and Alia Salah hospital in Tebessa, Algeria, on 560 patients with type 2 diabetes (300 women and 260 men), compared with 100 normal subjects (50 women and 50 men). For all subjects the following parameters were estimated: blood pressure, BMI (body mass index), glucose, glycated hemoglobin, total cholesterol, HDL Cholesterol, LDL Cholesterol, Triglycerides, Creatinine, serum redox status indicators (GSH, GPx, GSTs, and MDA) and a complete blood count was performed. Results The findings of this study indicated a slight increase in arterial pressure in 336 diabetic patients (60%) with an HbA1c level between 7 and 9% (210 patients) and > 9% (126 patients); while the flow of the glomerular filtration remained within the norms for all the studied subjects. Patients showed an increase in blood glucose levels, disturbance of the lipid parameters with an increase in lipid peroxidation and a decrease in serum and erythrocyte antioxidant defense. Conclusion It can be concluded that the formation of atheroma plate in diabetics is caused by the oxidation of circulating lipoproteins by free radicals generated following hyperglycemia, which can be avoided by supplementing antioxidant molecules such as antioxidant vitamins, trace elements.
Collapse
|
47
|
Harki O, Tamisier R, Pépin JL, Bailly S, Mahmani A, Gonthier B, Salomon A, Vilgrain I, Faury G, Briançon-Marjollet A. VE-cadherin cleavage in sleep apnoea: new insights into intermittent hypoxia-related endothelial permeability. Eur Respir J 2021; 58:13993003.04518-2020. [PMID: 33737411 DOI: 10.1183/13993003.04518-2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/24/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Obstructive sleep apnoea (OSA) causes intermittent hypoxia that in turn induces endothelial dysfunction and atherosclerosis progression. We hypothesised that VE-cadherin cleavage, detected by its released extracellular fragment solubilised in the blood (sVE), may be an early indicator of emergent abnormal endothelial permeability. Our aim was to assess VE-cadherin cleavage in OSA patients and in in vivo and in vitro intermittent hypoxia models to decipher the cellular mechanisms and consequences. METHODS Sera from seven healthy volunteers exposed to 14 nights of intermittent hypoxia, 43 OSA patients and 31 healthy control subjects were analysed for their sVE content. Human aortic endothelial cells (HAECs) were exposed to 6 h of intermittent hypoxia in vitro, with or without an antioxidant or inhibitors of hypoxia-inducible factor (HIF)-1, tyrosine kinases or vascular endothelial growth factor (VEGF) pathways. VE-cadherin cleavage and phosphorylation were evaluated, and endothelial permeability was assessed by measuring transendothelial electrical resistance (TEER) and fluorescein isothiocyanate (FITC)-dextran flux. RESULTS sVE was significantly elevated in sera from healthy volunteers submitted to intermittent hypoxia and OSA patients before treatment, but conversely decreased in OSA patients after 6 months of continuous positive airway pressure treatment. OSA was the main factor accounting for sVE variations in a multivariate analysis. In in vitro experiments, cleavage and expression of VE-cadherin increased upon HAEC exposure to intermittent hypoxia. TEER decreased and FITC-dextran flux increased. These effects were reversed by all of the pharmacological inhibitors tested. CONCLUSIONS We suggest that in OSA, intermittent hypoxia increases endothelial permeability in OSA by inducing VE-cadherin cleavage through reactive oxygen species production, and activation of HIF-1, VEGF and tyrosine kinase pathways.
Collapse
Affiliation(s)
- Olfa Harki
- Université Grenoble Alpes, INSERM, CHU Grenoble Alpes, Laboratoire HP2, Grenoble, France
| | - Renaud Tamisier
- Université Grenoble Alpes, INSERM, CHU Grenoble Alpes, Laboratoire HP2, Grenoble, France
| | - Jean-Louis Pépin
- Université Grenoble Alpes, INSERM, CHU Grenoble Alpes, Laboratoire HP2, Grenoble, France
| | - Sébastien Bailly
- Université Grenoble Alpes, INSERM, CHU Grenoble Alpes, Laboratoire HP2, Grenoble, France
| | - Anissa Mahmani
- Université Grenoble Alpes, INSERM, CHU Grenoble Alpes, Laboratoire HP2, Grenoble, France
| | - Brigitte Gonthier
- Université Grenoble Alpes, INSERM, CHU Grenoble Alpes, Laboratoire HP2, Grenoble, France
| | - Aude Salomon
- Université Grenoble Alpes, INSERM U1036, CEA, Grenoble, France
| | | | - Gilles Faury
- Université Grenoble Alpes, INSERM, CHU Grenoble Alpes, Laboratoire HP2, Grenoble, France
| | | |
Collapse
|
48
|
Cercy SP. Pericytes and the Neurovascular Unit: The Critical Nexus of Alzheimer Disease Pathogenesis? EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2021. [DOI: 10.14218/erhm.2020.00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
49
|
Zhao X, Yu X, Xin S, Zhang W, Zhang X, Ji L. Correlation between OSAHS and Early Peripheral Atherosclerosis Indices in Patients with Type 2 Diabetes Mellitus in China: A Cross-Sectional Inpatient Study. J Diabetes Res 2021; 2021:6630020. [PMID: 33628835 PMCID: PMC7895581 DOI: 10.1155/2021/6630020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/10/2021] [Accepted: 02/02/2021] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To analyze the differences of early atherosclerosis indices in type 2 diabetes mellitus (T2DM) patients with different degrees of obstructive sleep apnea-hypopnea syndrome (OSAHS) and explore the correlation between them, so as to provide a new clinical basis for the prevention and treatment of early atherosclerosis in patients with T2DM and OSAHS. Methods. A prospective study was conducted in 312 patients with T2DM and snoring who were hospitalized in the Department of Endocrinology, Peking University International Hospital from January 2017 to January 2020. According to the monitoring results, 312 patients were divided into 4 groups including the control group (208 cases), mild OSAHS group (18 cases), moderate OSAHS group (38 cases), and severe OSAHS group (48 cases). Multivariate logistic regression analysis was used to analyze the early atherosclerosis indices including brachial-ankle pulse wave velocity (PWV) and ankle-brachial index (ABI) in patients with T2DM coexistence with different degrees of OSAHS. Results. (1) As the degree of OSAHS increased, ABI decreased gradually and was lower than that in the control group, but PWV increased and was higher than that in the control group (p < 0.05, respectively). (2) The apnea-hypopnea index (AHI) positively correlated with PWV (r = 0.36, p < 0.05) and negatively correlated with ABI (r = -0.37, p < 0.05). (3) Multivariate logistic regression showed that after adjusting for age, gender, duration, BMI, blood pressure, blood glucose, blood lipid, and other factors, OSAHS was a risk factor of lower extremity arterial disease (LEAD) in patients with T2DM. With the increase of degree of OSAHS, the risk of lower extremity atherosclerosis gradually increased. Conclusion. OSAHS is an independent risk factor of LEAD in patients with T2DM, and with the increase of AHI, the ABI and PWV have changed, which provides a new clinical basis for the prevention and the treatment of early atherosclerosis in patients with T2DM and OSAHS.
Collapse
Affiliation(s)
- Xin Zhao
- Endocrinology Department, Peking University International Hospital, Beijing, China
| | - Xiaofeng Yu
- Endocrinology Department, Peking University International Hospital, Beijing, China
| | - Sixu Xin
- Endocrinology Department, Peking University International Hospital, Beijing, China
| | - Wei Zhang
- Sleep Center Department, Peking University International Hospital, Beijing, China
| | - Xiaomei Zhang
- Endocrinology Department, Peking University International Hospital, Beijing, China
| | - Linong Ji
- Endocrinology Department, cc, Beijing, China
| |
Collapse
|
50
|
Liu T, Liao XZ, Zhou MT. Ulinastatin alleviates traumatic brain injury by reducing endothelin-1. Transl Neurosci 2021; 12:1-8. [PMID: 33505713 PMCID: PMC7788573 DOI: 10.1515/tnsci-2021-0001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022] Open
Abstract
Background Brain edema is one of the major causes of fatality and disability associated with injury and neurosurgical procedures. The goal of this study was to evaluate the effect of ulinastatin (UTI), a protease inhibitor, on astrocytes in a rat model of traumatic brain injury (TBI). Methodology A rat model of TBI was established. Animals were randomly divided into 2 groups – one group was treated with normal saline and the second group was treated with UTI (50,000 U/kg). The brain water content and permeability of the blood–brain barrier were assessed in the two groups along with a sham group (no TBI). Expression of the glial fibrillary acidic protein, endthelin-1 (ET-1), vascular endothelial growth factor (VEGF), and matrix metalloproteinase 9 (MMP-9) were measured by immunohistochemistry and western blot. Effect of UTI on ERK and PI3K/AKT signaling pathways was measured by western blot. Results UTI significantly decreased the brain water content and extravasation of the Evans blue dye. This attenuation was associated with decreased activation of the astrocytes and ET-1. UTI treatment decreased ERK and Akt activation and inhibited the expression of pro-inflammatory VEGF and MMP-9. Conclusion UTI can alleviate brain edema resulting from TBI by inhibiting astrocyte activation and ET-1 production.
Collapse
Affiliation(s)
- Ting Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Xing-Zhi Liao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Mai-Tao Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|