1
|
Kiss MG, Cohen O, McAlpine CS, Swirski FK. Influence of sleep on physiological systems in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1284-1300. [PMID: 39528718 PMCID: PMC11567060 DOI: 10.1038/s44161-024-00560-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Sleep is a fundamental requirement of life and is integral to health. Deviation from optimal sleep associates with numerous diseases including those of the cardiovascular system. Studies, spanning animal models to humans, show that insufficient, disrupted or inconsistent sleep contribute to poor cardiovascular health by disrupting body systems. Fundamental experiments have begun to uncover the molecular and cellular links between sleep and heart health while large-scale human studies have associated sleep with cardiovascular outcomes in diverse populations. Here, we review preclinical and clinical findings that demonstrate how sleep influences the autonomic nervous, metabolic and immune systems to affect atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Máté G Kiss
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oren Cohen
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Filip K Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
2
|
Cheung EC, Nilsson A, Venter I, Kowalik G, Ribeiro C, Rodriguez J, Kuraoka K, Russo R, Escobar JB, Alber BR, Mendelowitz D, Kay MW, Schunke KJ. Sex differences in cardiac transcriptomic response to neonatal sleep apnea. Physiol Rep 2024; 12:e16110. [PMID: 38981849 PMCID: PMC11233197 DOI: 10.14814/phy2.16110] [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: 03/25/2024] [Revised: 05/03/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
Pediatric obstructive sleep apnea poses a significant health risk, with potential long-term consequences on cardiovascular health. This study explores the dichotomous nature of neonatal cardiac response to chronic intermittent hypoxia (CIH) between males and females, aiming to fill a critical knowledge gap in the understanding of sex-specific cardiovascular consequences of sleep apnea in early life. Neonates were exposed to CIH until p28 and underwent comprehensive in vivo physiological assessments, including whole-body plethysmography, treadmill stress-tests, and echocardiography. Results indicated that male CIH rats weighed 13.7% less than age-matched control males (p = 0.0365), while females exhibited a mild yet significant increased respiratory drive during sleep (93.94 ± 0.84 vs. 95.31 ± 0.81;p = 0.02). Transcriptomic analysis of left ventricular tissue revealed a substantial sex-based difference in the cardiac response to CIH, with males demonstrating a more pronounced alteration in gene expression compared to females (5986 vs. 3174 genes). The dysregulated miRNAs in males target metabolic genes, potentially predisposing the heart to altered metabolism and substrate utilization. Furthermore, CIH in males was associated with thinner left ventricular walls and dysregulation of genes involved in the cardiac action potential, possibly predisposing males to CIH-related arrhythmia. These findings emphasize the importance of considering sex-specific responses in understanding the cardiovascular implications of pediatric sleep apnea.
Collapse
Affiliation(s)
- Emily C. Cheung
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Anna Nilsson
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Ian Venter
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Grant Kowalik
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Caitlin Ribeiro
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Jeannette Rodriguez
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Kiralee Kuraoka
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Rebekah Russo
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Joan B. Escobar
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Bridget R. Alber
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - David Mendelowitz
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Matthew W. Kay
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Kathryn J. Schunke
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| |
Collapse
|
3
|
Pramanik S, Mondal S, Palui R, Ray S. Type 2 diabetes in children and adolescents: Exploring the disease heterogeneity and research gaps to optimum management. World J Clin Pediatr 2024; 13:91587. [PMID: 38947996 PMCID: PMC11212753 DOI: 10.5409/wjcp.v13.i2.91587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 06/07/2024] Open
Abstract
Over the past 20 years, the incidence and prevalence of type 2 diabetes mellitus (T2DM) in children and adolescents have increased, particularly in racial and ethnic minorities. Despite the rise in T2DM in children and adolescents, the pathophysiology and progression of disease in this population are not clearly understood. Youth-onset T2DM has a more adverse clinical course than is seen in those who develop T2DM in adulthood or those with T1DM. Furthermore, the available therapeutic options are more limited for children and adolescents with T2DM compared to adult patients, mostly due to the challenges of implementing clinical trials. A better understanding of the mechanisms underlying the de-velopment and aggressive disease phenotype of T2DM in youth is important to finding effective prevention and management strategies. This review highlights the key evidence about T2DM in children and adolescents and its current burden and challenges both in clinical care and research activities.
Collapse
Affiliation(s)
- Subhodip Pramanik
- Department of Endocrinology, Neotia Getwel Multi-specialty hospital, Siliguri 734010, West Bengal, India
| | - Sunetra Mondal
- Department of Endocrinology, NRS Medical College and Hospital, Kolkata 700014, West Bengal, India
| | - Rajan Palui
- Department of Endocrinology, The Mission Hospital, Durgapur 713212, West Bengal, India
| | - Sayantan Ray
- Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar, Bhubaneswar 751019, Odisha, India
| |
Collapse
|
4
|
Paul O, Akolia IK, Qin Tao J, Jain N, Louneva N, Montone KT, Fisher AB, Rajapakse CS, Bermudez C, Chatterjee S. Reactive oxygen species in endothelial signaling in COVID-19: Protective role of the novel peptide PIP-2. PLoS One 2024; 19:e0289854. [PMID: 38771750 PMCID: PMC11108150 DOI: 10.1371/journal.pone.0289854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 04/02/2024] [Indexed: 05/23/2024] Open
Abstract
INTRODUCTION Recent research suggests that endothelial activation plays a role in coronavirus disease 2019 (COVID-19) pathogenesis by promoting a pro-inflammatory state. However, the mechanism by which the endothelium is activated in COVID-19 remains unclear. OBJECTIVE To investigate the mechanism by which COVID-19 activates the pulmonary endothelium and drives pro-inflammatory phenotypes. HYPOTHESIS The "inflammatory load or burden" (cytokine storm) of the systemic circulation activates endothelial NADPH oxidase 2 (NOX2) which leads to the production of reactive oxygen species (ROS) by the pulmonary endothelium. Endothelial ROS subsequently activates pro-inflammatory pathways. METHODS The inflammatory burden of COVID-19 on the endothelial network, was recreated in vitro, by exposing human pulmonary microvascular endothelial cells (HPMVEC) to media supplemented with serum from COVID-19 affected individuals (sera were acquired from patients with COVID-19 infection that eventually died. Sera was isolated from blood collected at admission to the Intensive Care Unit of the Hospital of the University of Pennsylvania). Endothelial activation, inflammation and cell death were assessed in HPMVEC treated with serum either from patients with COVID-19 or from healthy individuals. Activation was monitored by measuring NOX2 activation (Rac1 translocation) and ROS production; inflammation (or appearance of a pro-inflammatory phenotype) was monitored by measuring the induction of moieties such as intercellular adhesion molecule (ICAM-1), P-selectin and the NLRP3 inflammasome; cell death was measured via SYTOX™ Green assays. RESULTS Endothelial activation (i.e., NOX2 activation and subsequent ROS production) and cell death were significantly higher in the COVID-19 model than in healthy samples. When HPMVEC were pre-treated with the novel peptide PIP-2, which blocks NOX2 activation (via inhibition of Ca2+-independent phospholipase A2, aiPLA2), significant abrogation of ROS was observed. Endothelial inflammation and cell death were also significantly blunted. CONCLUSIONS The endothelium is activated during COVID-19 via cytokine storm-driven NOX2-ROS activation, which causes a pro-inflammatory phenotype. The concept of endothelial NOX2-ROS production as a unifying pathophysiological axis in COVID-19 raises the possibility of using PIP-2 to maintain vascular health.
Collapse
Affiliation(s)
- Oindrila Paul
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| | - Isha K. Akolia
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| | - Jian Qin Tao
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| | - Nikita Jain
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| | - Natalia Louneva
- Peroxitech Inc., Philadelphia, Pennsylvania, United States of America
| | - Kathleen T. Montone
- Department of Pathology, Philadelphia, Pennsylvania, United States of America
| | - Aron B. Fisher
- Peroxitech Inc., Philadelphia, Pennsylvania, United States of America
| | - Chamith S. Rajapakse
- Department of Radiology, Philadelphia, Pennsylvania, United States of America
- Department of Orthopedic Surgery, Philadelphia, Pennsylvania, United States of America
| | - Christian Bermudez
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Shampa Chatterjee
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
5
|
Weng L, Luo X, Luo Y, Zhang Q, Yao K, Tan J, Yin Y. Association Between Sleep Apnea Syndrome and Osteoarthritis: Insights from Bidirectional Mendelian Randomization and Bioinformatics Analysis. Nat Sci Sleep 2024; 16:473-487. [PMID: 38737460 PMCID: PMC11088414 DOI: 10.2147/nss.s461010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/26/2024] [Indexed: 05/14/2024] Open
Abstract
Background Sleep apnea syndrome(SAS) and osteoarthritis (OA) are two prevalent diseases that often coexist, but the causal relationship between them remains unclear. In light of this, our team utilizes Mendelian Randomization and bioinformatics analysis methods to investigate the potential association between the two diseases. Methods In this study, we utilized GWAS data pertaining to SAS and OA to assess the causal relationship between the two diseases through Mendelian randomization (MR) analysis. We then employed transcriptomic data to perform differential gene identification, WGCNA, shared gene determination, functional enrichment analysis, and colocalization analysis, all designed to further elucidate the mechanisms underlying the association between the two diseases. In the end, we utilized Mendelian randomization (MR) analysis again to delve deeper into the relationship between the two diseases and immune cells. Results Our research findings indicate that SAS is a risk factor for OA (p = 0.000004), knee OA (p = 0.0000001) and hip OA(p = 0.001). Furthermore, OA (p = 0.000195), knee OA (p = 0.001) are significant risk factors for SAS. However, there is no clear evidence that hip OA (p = 0.892) is a risk factor for SAS. Interestingly, the genes shared between OA and SAS are significantly enriched in leukocyte migration, leukocyte chemotaxis. Moreover, colocalization analysis suggests that the genes JUNB, COL8A1, FOSB, and IER2 may be key genes associated with both diseases. Furthermore, 57 immune cell phenotypes are associated with SAS, 95 with OA, and 6 shared between both diseases. Conclusion This research confirmed the bidirectional causal relationship between SAS and OA. Notably, the 4 genes (JUNB, COL8A1, FOSB, IER2) and 6 immune phenotypes are crucial for both diseases, these provide hopeful targets for future interventions against these two diseases.
Collapse
Affiliation(s)
- Lian Weng
- Department of orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Sichuan Provincial Laboratory of Orthopedic Engineering, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Department of Clinical Medicine, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xiongjunjie Luo
- Department of orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Sichuan Provincial Laboratory of Orthopedic Engineering, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Department of Clinical Medicine, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yuxi Luo
- Department of orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Sichuan Provincial Laboratory of Orthopedic Engineering, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Department of Clinical Medicine, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Qian Zhang
- Department of orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Sichuan Provincial Laboratory of Orthopedic Engineering, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Department of Clinical Medicine, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Kaitao Yao
- Department of orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Sichuan Provincial Laboratory of Orthopedic Engineering, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Department of Clinical Medicine, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Junjie Tan
- Department of orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Sichuan Provincial Laboratory of Orthopedic Engineering, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Department of Clinical Medicine, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yiran Yin
- Department of orthopedics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Sichuan Provincial Laboratory of Orthopedic Engineering, Luzhou, Sichuan Province, 646000, People’s Republic of China
- Department of Clinical Medicine, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
6
|
White B, Ng SM, Agwu JC, Barrett TG, Birchmore N, Kershaw M, Drew J, Kavvoura F, Law J, Moudiotis C, Procter E, Paul P, Regan F, Reilly P, Sachdev P, Sakremath R, Semple C, Sharples K, Skae M, Timmis A, Williams E, Wright N, Soni A. A practical evidence-based approach to management of type 2 diabetes in children and young people (CYP): UK consensus. BMC Med 2024; 22:144. [PMID: 38561783 PMCID: PMC10986054 DOI: 10.1186/s12916-024-03349-4] [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: 07/18/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Type 2 diabetes in young people is an aggressive disease with a greater risk of complications leading to increased morbidity and mortality during the most productive years of life. Prevalence in the UK and globally is rising yet experience in managing this condition is limited. There are no consensus guidelines in the UK for the assessment and management of paediatric type 2 diabetes. METHODS Multidisciplinary professionals from The Association of Children's Diabetes Clinicians (ACDC) and the National Type 2 Diabetes Working Group reviewed the evidence base and made recommendations using the Grading Of Recommendations, Assessment, Development and Evaluation (GRADE) methodology. RESULTS AND DISCUSSION Young people with type 2 diabetes should be managed within a paediatric diabetes team with close working with adult diabetes specialists, primary care and other paediatric specialties. Diagnosis of diabetes type can be challenging with many overlapping features. Diabetes antibodies may be needed to aid diagnosis. Co-morbidities and complications are frequently present at diagnosis and should be managed holistically. Lifestyle change and metformin are the mainstay of early treatment, with some needing additional basal insulin. GLP1 agonists should be used as second-line agents once early ketosis and symptoms are controlled. Glycaemic control improves microvascular but not cardiovascular risk. Reduction in excess adiposity, smoking prevention, increased physical activity and reduction of hypertension and dyslipidaemia are essential to reduce major adverse cardiovascular events. CONCLUSIONS This evidence-based guideline aims to provide a practical approach in managing this condition in the UK.
Collapse
Affiliation(s)
- Billy White
- University College London Hospitals NHS Foundation Trust, London, UK
| | - S M Ng
- Mersey And West Lancashire Teaching Hospitals NHS Trust, Ormskirk, UK
| | - J C Agwu
- Wye Valley NHS Trust, Hereford, UK
| | - T G Barrett
- Birmingham Women's And Children NHS Foundation Trust, Birmingham, UK
| | - N Birchmore
- Great Ormond Street Hospital For Children, NHS Foundation Trust, London, UK
| | - M Kershaw
- Birmingham Women's And Children NHS Foundation Trust, Birmingham, UK
| | - J Drew
- Nottingham University Hospitals NHS Foundation Trust, Nottingham, UK
| | - F Kavvoura
- Royal Berkshire NHS Foundation Trust, Reading, UK
| | - J Law
- Nottingham University Hospitals NHS Foundation Trust, Nottingham, UK
| | - C Moudiotis
- Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - E Procter
- Nottingham University Hospitals NHS Foundation Trust, Nottingham, UK
| | - P Paul
- Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - F Regan
- Guy's and St Thomas's NHS Foundation Trust, London, UK
| | - P Reilly
- Torbay and South Devon NHS Foundation Trust, Torquay, UK
| | - P Sachdev
- Nottingham University Hospitals NHS Foundation Trust, Nottingham, UK
| | - R Sakremath
- Shrewsbury and Telford Hospital NHS Trust, Shrewsbury, UK
| | - C Semple
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | | | - M Skae
- Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - A Timmis
- Countess of Chester Hospital NHS Foundation Trust, Chester, UK
| | - E Williams
- Hampshire Hospitals NHS Foundation Trust, Winchester, UK
| | - N Wright
- Sheffield Children's Hospital NHS Foundation Trust, Sheffield, S102TH, UK
| | - A Soni
- Sheffield Children's Hospital NHS Foundation Trust, Sheffield, S102TH, UK.
| |
Collapse
|
7
|
Miller MA, Howarth NE. Sleep and cardiovascular disease. Emerg Top Life Sci 2023; 7:457-466. [PMID: 38084859 PMCID: PMC10754327 DOI: 10.1042/etls20230111] [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: 09/18/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
This review centres around the recent evidence in examining the intersection of sleep and cardiovascular disease (CVD). Sleep in this review will be further subdivided to consider both sleep quantity and quality along and will also consider some of the more common sleep disorders, such as insomnia and obstructive sleep apnoea, in the context of CVD. Sleep disorders have been further explored in several specific populations which are both at risk of sleep disorders and CVD. Secondly, the review will present some of the risk factors for CVD that are affected by sleep and sleep disorders which include hypertension, diabetes, and obesity. It will also examine the potential underlying mechanisms including inflammation, appetite control, endocrine, and genetic processes that are affected by sleep and sleep disorders leading to increased risk of CVD development. In addition, we will consider the observed bi-directional relationships between sleep and cardiovascular risk factors. For example, obesity, a risk factor for CVD can be affected by sleep, but in turn can increase the risk of certain sleep disorder development which disrupts sleep, leading to further risk of obesity development and increased CVD risk. Finally, the review will explore emerging evidence around lifestyle interventions that have included a sleep component and how it impacts the management of CVD risk factor. The need for increased awareness of the health effects of poor sleep and sleep disorders will be discussed alongside the need for policy intervention to improve sleep to facilitate better health and well-being.
Collapse
Affiliation(s)
- Michelle A. Miller
- Division of Health Sciences (Mental Health and Wellbeing), Warwick Medical School, University of Warwick, Gibbet Hill, Coventry CV4 7AL, U.K
| | - Nathan E. Howarth
- Division of Health Sciences (Mental Health and Wellbeing), Warwick Medical School, University of Warwick, Gibbet Hill, Coventry CV4 7AL, U.K
| |
Collapse
|
8
|
Figtree GA, Vernon ST, Harmer JA, Gray MP, Arnott C, Bachour E, Barsha G, Brieger D, Brown A, Celermajer DS, Channon KM, Chew NWS, Chong JJH, Chow CK, Cistulli PA, Ellinor PT, Grieve SM, Guzik TJ, Hagström E, Jenkins A, Jennings G, Keech AC, Kott KA, Kritharides L, Mamas MA, Mehran R, Meikle PJ, Natarajan P, Negishi K, O'Sullivan J, Patel S, Psaltis PJ, Redfern J, Steg PG, Sullivan DR, Sundström J, Vogel B, Wilson A, Wong D, Bhatt DL, Kovacic JC, Nicholls SJ. Clinical Pathway for Coronary Atherosclerosis in Patients Without Conventional Modifiable Risk Factors: JACC State-of-the-Art Review. J Am Coll Cardiol 2023; 82:1343-1359. [PMID: 37730292 PMCID: PMC10522922 DOI: 10.1016/j.jacc.2023.06.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 06/28/2023] [Indexed: 09/22/2023]
Abstract
Reducing the incidence and prevalence of standard modifiable cardiovascular risk factors (SMuRFs) is critical to tackling the global burden of coronary artery disease (CAD). However, a substantial number of individuals develop coronary atherosclerosis despite no SMuRFs. SMuRFless patients presenting with myocardial infarction have been observed to have an unexpected higher early mortality compared to their counterparts with at least 1 SMuRF. Evidence for optimal management of these patients is lacking. We assembled an international, multidisciplinary team to develop an evidence-based clinical pathway for SMuRFless CAD patients. A modified Delphi method was applied. The resulting pathway confirms underlying atherosclerosis and true SMuRFless status, ensures evidence-based secondary prevention, and considers additional tests and interventions for less typical contributors. This dedicated pathway for a previously overlooked CAD population, with an accompanying registry, aims to improve outcomes through enhanced adherence to evidence-based secondary prevention and additional diagnosis of modifiable risk factors observed.
Collapse
Affiliation(s)
- Gemma A Figtree
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Cardiovascular Discovery Group, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia; Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia.
| | - Stephen T Vernon
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Cardiovascular Discovery Group, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia; Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Jason A Harmer
- Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia; The George Institute for Global Health, Faculty of Medicine, UNSW, Sydney, New South Wales, Australia
| | - Michael P Gray
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Cardiovascular Discovery Group, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia
| | - Clare Arnott
- The George Institute for Global Health, Faculty of Medicine, UNSW, Sydney, New South Wales, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Eric Bachour
- Consumer Representative, Agile Group Switzerland AG, Zug, Switzerland
| | - Giannie Barsha
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Cardiovascular Discovery Group, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia
| | - David Brieger
- Department of Cardiology, Concord Repatriation General Hospital, Concord, New South Wales, Australia
| | - Alex Brown
- National Centre for Indigenous Genomics, Australian National University, Canberra, Australian Capitol Territory, Australia; Telethon Kids Institute, Nedlands, Western Australia, Australia
| | - David S Celermajer
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Keith M Channon
- British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas W S Chew
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore
| | - James J H Chong
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia; Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Clara K Chow
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Peter A Cistulli
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia; Department of Respiratory & Sleep Medicine, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stuart M Grieve
- Department of Radiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia; Imaging and Phenotyping Laboratory, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Tomasz J Guzik
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom; Department of Internal Medicine and Omicron Medical Genomics Laboratory, Jagiellonian University Medical College, Krakow, Poland
| | - Emil Hagström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Alicia Jenkins
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia; Diabetes and Vascular Medicine, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Garry Jennings
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Anthony C Keech
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Katharine A Kott
- Cardiovascular Discovery Group, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia; Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Leonard Kritharides
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Concord Repatriation General Hospital, Concord, New South Wales, Australia; The ANZAC Research Institute, Concord Repatriation General Hospital, Concord, New South Wales, Australia
| | - Mamas A Mamas
- Keele Cardiovascular Research Group, Centre for Prognostic Research, Keele University, Keele, United Kingdom; Department of Cardiology, Royal Stoke University Hospital, Stoke-on-Trent, United Kingdom
| | - Roxana Mehran
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Vicotria, Australia
| | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Kazuaki Negishi
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia; Department of Cardiology, Nepean Hospital, Kingswood, New South Wales, Australia
| | - John O'Sullivan
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia; Precision Cardiovascular Laboratory, University of Sydney, Camperdown, New South Wales, Australia; Heart Research Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Sanjay Patel
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia; Heart Research Institute, University of Sydney, Camperdown, New South Wales, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, SAHMRI, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Cardiology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - Julie Redfern
- The George Institute for Global Health, Faculty of Medicine, UNSW, Sydney, New South Wales, Australia; Sydney School of Health Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Philippe G Steg
- Université de Paris, Assistance Publique-Hôpitaux de Paris, French Alliance for Cardiovascular Trials and INSERM Unité 1148, Paris, France
| | - David R Sullivan
- Department of Chemical Pathology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Johan Sundström
- The George Institute for Global Health, Faculty of Medicine, UNSW, Sydney, New South Wales, Australia; Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Birgit Vogel
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrew Wilson
- Menzies Centre for Health Policy and Economics, Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Dennis Wong
- Monash Cardiovascular Research Centre, Monash University, Clayton, Victoria, Australia; MonashHeart, Monash Health, Clayton, Victoria, Australia
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, New York, New York, USA
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
9
|
Chung J, Goodman M, Huang T, Wallace ML, Lutsey PL, Chen JT, Castro-Diehl C, Bertisch S, Redline S. Multi-dimensional sleep and mortality: The Multi-Ethnic Study of Atherosclerosis. Sleep 2023; 46:zsad048. [PMID: 37523657 PMCID: PMC10848217 DOI: 10.1093/sleep/zsad048] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/30/2023] [Indexed: 08/02/2023] Open
Abstract
STUDY OBJECTIVES Multiple sleep characteristics are informative of health, sleep characteristics cluster, and sleep health can be described as a composite of positive sleep attributes. We assessed the association between a sleep score reflecting multiple sleep dimensions, and mortality. We tested the hypothesis that more favorable sleep (higher sleep scores) is associated with lower mortality. METHODS The Multi-Ethnic Study of Atherosclerosis (MESA) is a racially and ethnically-diverse multi-site, prospective cohort study of US adults. Sleep was measured using unattended polysomnography, 7-day wrist actigraphy, and validated questionnaires (2010-2013). 1726 participants were followed for a median of 6.9 years (Q1-Q3, 6.4-7.4 years) until death (171 deaths) or last contact. Survival models were used to estimate the association between the exposure of sleep scores and the outcome of all-cause mortality, adjusting for socio-demographics, lifestyle, and medical comorbidities; follow-up analyses examined associations between individual metrics and mortality. The exposure, a sleep score, was constructed by an empirically-based Principal Components Analysis on 13 sleep metrics, selected a priori. RESULTS After adjusting for multiple confounders, a 1 standard deviation (sd) higher sleep score was associated with 25% lower hazard of mortality (Hazard Ratio [HR]: 0.75; 95% Confidence interval: [0.65, 0.87]). The largest drivers of this association were: night-to-night sleep regularity, total sleep time, and the Apnea-Hypopnea Index. CONCLUSION More favorable sleep across multiple characteristics, operationalized by a sleep score, is associated with lower risk of death in a diverse US cohort of adults. Results suggest that interventions that address multiple dimensions may provide novel approaches for improving health.
Collapse
Affiliation(s)
- Joon Chung
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Matthew Goodman
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tianyi Huang
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Pamela L Lutsey
- Division of Epidemiology and Community Health, School of Public Health, the University of Minnesota, Minneapolis, MN, USA
| | - Jarvis T Chen
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Suzanne Bertisch
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Sanchez-Azofra A, Gu W, Masso-Silva JA, Sanz-Rubio D, Marin-Oto M, Cubero P, Gil AV, Moya EA, Barnes LA, Mesarwi OA, Marin T, Simonson TS, Crotty Alexander LE, Marin JM, Malhotra A. Inflammation biomarkers in OSA, chronic obstructive pulmonary disease, and chronic obstructive pulmonary disease/OSA overlap syndrome. J Clin Sleep Med 2023; 19:1447-1456. [PMID: 37082823 PMCID: PMC10394367 DOI: 10.5664/jcsm.10600] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/22/2023]
Abstract
STUDY OBJECTIVES The coexistence of obstructive sleep apnea (OSA) and chronic obstructive pulmonary disease (COPD) in a single individual, also known as overlap syndrome (OVS), is associated with higher cardiovascular risk and mortality than either OSA or COPD alone. However, the underlying mechanisms remain unclear. We hypothesized that patients with OVS have elevated systemic inflammatory biomarkers relative to patients with either disease alone, which could explain greater cardiovascular risk observed in OVS. METHODS We included 255 participants in the study, 55 with COPD alone, 100 with OSA alone, 50 with OVS, and 50 healthy controls. All participants underwent a home sleep study, spirometry, and a blood draw for high-sensitivity C-reactive protein and total blood count analysis. In a randomly selected subset of 186 participants, inflammatory protein profiling was performed using Bio-Rad Bio-Plex Pro Human Cytokine 27-Plex Assays. Biomarker level differences across groups were identified using a mixed linear model. RESULTS Levels of interleukin 6 (IL-6), high-sensitivity C-reactive protein (hs-CRP), and granulocyte colony stimulating factor (G-CSF) were higher in participants with OVS and COPD compared with healthy controls and participants with OSA. Furthermore, participants with OVS had higher circulating levels of leukocytes and neutrophils than those with COPD, OSA, and controls. CONCLUSIONS COPD and OVS are associated with higher systemic inflammation relative to OSA and healthy controls. This work proposes the potential utilization of interleukin 6, granulocyte colony stimulating factor, and high-sensitivity C-reactive protein as screening biomarkers for COPD in patients with OSA. Inflammatory pathways may not fully explain the higher cardiovascular risk observed in OVS, indicating the need for further investigation. CITATION Sanchez-Azofra A, Gu W, Masso-Silva JA, et al. Inflammation biomarkers in OSA, chronic obstructive pulmonary disease, and chronic obstructive pulmonary disease/OSA overlap syndrome. J Clin Sleep Med. 2023;19(8):1447-1456.
Collapse
Affiliation(s)
- Ana Sanchez-Azofra
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
- Division of Pulmonary and Sleep Medicine. Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Madrid, España
| | - Wanjun Gu
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
- Herbert Wertheim School of Public Health and Longevity Sciences, University of California, San Diego, La Jolla, California
| | - Jorge A. Masso-Silva
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
- Section of Pulmonary and Critical Care, VA San Diego, La Jolla, California
| | - David Sanz-Rubio
- Translational Research Unit, IIS Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Marta Marin-Oto
- Translational Research Unit, IIS Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Pablo Cubero
- Translational Research Unit, IIS Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Ana V. Gil
- Translational Research Unit, IIS Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Esteban A. Moya
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
| | - Laura A. Barnes
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
| | - Omar A. Mesarwi
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
| | - Traci Marin
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
- Health Sciences, Department of Respiratory Therapy, Victor Valley College, Victorville, California
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
- Center for Physiological Genomics of Low Oxygen, University of California, La Jolla, California
| | - Laura E. Crotty Alexander
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
- Section of Pulmonary and Critical Care, VA San Diego, La Jolla, California
| | - Jose M. Marin
- Translational Research Unit, IIS Aragón, Hospital Universitario Miguel Servet, Zaragoza, Spain
- CIBERES Instituto Salud Carlos III, and Department of Medicine, University of Zaragoza School of Medicine, Zaragoza, Spain
| | - Atul Malhotra
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, La Jolla, California
- Center for Physiological Genomics of Low Oxygen, University of California, La Jolla, California
| |
Collapse
|
11
|
Erdei E, Zhou X, Shuey C, Ass'ad N, Page K, Gore B, Zhu C, Kanda D, Luo L, Sood A, Zychowski KE. Serum autoantibodies and exploratory molecular pathways in rural miners: A pilot study. J Transl Autoimmun 2023; 6:100197. [PMID: 36942097 PMCID: PMC10023988 DOI: 10.1016/j.jtauto.2023.100197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Introduction The Southwestern United States (SWUS) has an extensive history of coal and metal mining, including uranium (U) mining. Lung diseases, including but not limited to, lung cancer and pulmonary fibrosis, have been studied extensively in miners due to occupational, dust-related exposures. However, high-throughput autoimmune biomarkers are largely understudied in miners, despite the fact that ore miners, such as U-miners, are at an increased risk for the development of autoimmune diseases such as systemic sclerosis and systemic lupus erythematosus (SLE). Additionally, there are current gaps in knowledge regarding which signaling pathways may play a role in occupational exposure-associated autoimmunity. Methods Most current and former miners in the SWUS live close to their previous workplaces, in remote areas, with limited access to healthcare. In this pilot study, by leveraging a mobile clinical platform for patient care and clinical outreach, we recruited 44 miners who self-identified as either U (n = 10) or non-U miners (n = 34) and received health screenings. Serum IgG and IgM autoantibodies against 128 antigens were assessed using a high-throughput molecular technique, as a preliminary health screening opportunity. Results Even when adjusting for age as a covariate, there was a significant (p < 0.05) association between self-reported U-mining exposure and biomarkers including IgM alpha-actinin, histones H2B, and H4, myeloperoxidase (MPO) and myelin basic protein. However, adjusting for age did not result in significant associations for IgG autoantibody production in U-miners. Bioinformatic pathway analysis revealed several altered signaling pathways between IgM and IgG autoantibodies among both U and non-U miners. Conclusions Further research is warranted regarding the mechanistic connection between U-exposure and autoantibody development, especially regarding histone-related alterations and IgM autoantibody production.
Collapse
Affiliation(s)
- Esther Erdei
- College of Pharmacy, University of New Mexico- Health Sciences Center, 905 Vassar Drive NE, Albuquerque, NM, 87106, USA
| | - Xixi Zhou
- College of Pharmacy, University of New Mexico- Health Sciences Center, 905 Vassar Drive NE, Albuquerque, NM, 87106, USA
| | - Chris Shuey
- Southwest Research and Information Center, 105 Stanford Drive SE, Albuquerque, NM, 87106, USA
| | - Nour Ass'ad
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Kimberly Page
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Bobbi Gore
- Miners' Colfax Medical Center, 203 Hospital Drive, Raton, NM, 87740, USA
| | - Chengsong Zhu
- Department of Immunology and Microarray Core, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Deborah Kanda
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87106, USA
| | - Li Luo
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, 87106, USA
| | - Akshay Sood
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
- Miners' Colfax Medical Center, 203 Hospital Drive, Raton, NM, 87740, USA
| | - Katherine E. Zychowski
- College of Nursing, University of New Mexico- Health Sciences Center, 2502 Marble Ave NE, Albuquerque, NM, 87131, USA
| |
Collapse
|
12
|
Young TL, Scieszka D, Begay JG, Lucas SN, Herbert G, Zychowski K, Hunter R, Salazar R, Ottens AK, Erdely A, Gu H, Campen MJ. Aging influence on pulmonary and systemic inflammation and neural metabolomics arising from pulmonary multi-walled carbon nanotube exposure in apolipoprotein E-deficient and C57BL/6 female mice. Inhal Toxicol 2023; 35:86-100. [PMID: 35037817 PMCID: PMC10037439 DOI: 10.1080/08958378.2022.2026538] [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: 09/28/2021] [Accepted: 01/03/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Environmental exposures exacerbate age-related pathologies, such as cardiovascular and neurodegenerative diseases. Nanoparticulates, and specifically carbon nanomaterials, are a fast-growing contributor to the category of inhalable pollutants, whose risks to health are only now being unraveled. The current study assessed the exacerbating effect of age on multiwalled-carbon nanotube (MWCNT) exposure in young and old C57BL/6 and ApoE-/- mice. MATERIALS AND METHODS Female C57BL/6 and apolipoprotein E-deficient (ApoE-/-) mice, aged 8 weeks and 15 months, were exposed to 0 or 40 µg MWCNT via oropharyngeal aspiration. Pulmonary inflammation, inflammatory bioactivity of serum, and neurometabolic changes were assessed at 24 h post-exposure. RESULTS Pulmonary neutrophil infiltration was induced by MWCNT in bronchoalveolar lavage fluid in both C57BL/6 and ApoE-/-. Macrophage counts decreased with MWCNT exposure in ApoE-/- mice but were unaffected by exposure in C57BL/6 mice. Older mice appeared to have greater MWCNT-induced total protein in lavage fluid. BALF cytokines and chemokines were elevated with MWCNT exposure, but CCL2, CXCL1, and CXCL10 showed reduced responses to MWCNT in older mice. However, no significant serum inflammatory bioactivity was detected. Cerebellar metabolic changes in response to MWCNT were modest, but age and strain significantly influenced metabolite profiles assessed. ApoE-/- mice and older mice exhibited less robust metabolite changes in response to exposure, suggesting a reduced health reserve. CONCLUSIONS Age influences the pulmonary and neurological responses to short-term MWCNT exposure. However, with only the model of moderate aging (15 months) in this study, the responses appeared modest compared to inhaled toxicant impacts in more advanced aging models.
Collapse
Affiliation(s)
- Tamara L. Young
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - David Scieszka
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Jessica G. Begay
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Selita N. Lucas
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | | | - Russell Hunter
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Raul Salazar
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Andrew K. Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, Richmond, VA 23298
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, US 85004
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
13
|
Janmohammadi P, Raeisi T, Zarei M, Nejad MM, Karimi R, Mirali Z, Zafary R, Alizadeh S. Adipocytokines in obstructive sleep apnea: A systematic review and meta-analysis. Respir Med 2023; 208:107122. [PMID: 36682601 DOI: 10.1016/j.rmed.2023.107122] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/23/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND AIM Adipocytokines play an important role in obstructive sleep apnea (OSA) by mediating inflammatory responses. Previous studies have reported that OSA is related to a change in the serum levels of adipocytokines; however, the results are still controversial. This meta-analysis aimed to assess the relationship between OSA and circulating level of adipocytokines in adults and children. METHODS A comprehensive search was conducted in databases of Medline/PubMed and Scopus for pertinent articles published since their inception to July 2022. Weighted mean differences (WMDs) and 95% confidence intervals (CIs) were used to assess the strength of the relationship between the concentrations of adipocytokines with OSA. RESULTS In the overall analysis, contrary to IL-10, which showed a significant reduction, IL-1β, IL-4, IL-8, IL-17, and IFN- gamma showed higher levels in OSA patients in comparison with control groups (p <0.05). For adults, IL-1β, IL-8, IL-17, IL-18, vaspin, visfatin, and chemerin were linked to a greater serum levels in patients with OSA, while, IL-5 and IL-10 were detected significantly lower in adults with OSA in comparison with healthy adults (p <0.05). In children with OSA, the serum levels of IL-4, IL-8, IL-12, IL-17, IL-23, and IFN-gamma were significantly higher than healthy children (p <0.05). CONCLUSION The levels of inflammatory markers were found to be higher in OSA patients compared with control individuals, suggesting that adipocytokines may contribute to the pathology of OSA.
Collapse
Affiliation(s)
- Parisa Janmohammadi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Tahereh Raeisi
- Department of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahtab Zarei
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Mofidi Nejad
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Roya Karimi
- Department of Preventive Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Zahra Mirali
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Reza Zafary
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shahab Alizadeh
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Proteomic profiling for prediction of recurrent cardiovascular event in patients with acute coronary syndrome and obstructive sleep apnea: A post-hoc analysis from the ISAACC study. Biomed Pharmacother 2023; 158:114125. [PMID: 36549084 DOI: 10.1016/j.biopha.2022.114125] [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: 09/21/2022] [Revised: 11/28/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is associated with a recurrent cardiovascular event (CVE) risk in patients with a first acute coronary syndrome (ACS). However, the pathological pathways by which OSA promotes this deleterious role are unknown. We aim to explore the proteomic profile associated with OSA that promote the recurrent CVE risk in severe OSA patients with ACS without previous cardiovascular diseases. METHODS This post-hoc analysis from the ISAACC study (NCT01335087) included 86 patients admitted for ACS. Patients underwent respiratory polygraphy for the first 24-72 h to OSA diagnosis. We analyzed of 276 cardiovascular and inflammatory related proteins in baseline fasting plasma samples using proximity expression assay technology (Olink®, Sweden). Protein levels were compared between severe OSA patients with/without recurrent CVEs during follow-up. Random forest was conducted to select relevant proteins and generate a predictive model of recurrent CVE. RESULTS We included 86 patients (median age: 61 years, median BMI: 29.4 kg/m2 and 86 % males) admitted for ACS with severe OSA (56 without recurrent CVE/30 with recurrent CVE). The plasma levels of 38 proteins were differentially expressed between groups. Additionally, 12 proteins had a significant association with respiratory polygraphy parameters. Three proteins discriminate with an AUC of 0.81 (95 % CI of 0.71-0.9) between severe OSA patients with and without recurrent CVE. These proteins were implicated in cell proliferation, communication and apoptosis, and regulation/response to the inflammatory and immune systems. CONCLUSION In ACS patients with severe OSA, a proteomic profile was associated with recurrent CVEs. This proteomic profile was correlated with specific OSA parameters from respiratory polygraphy. Proteomic profiling may provide an new direction for patient risk stratification and clinical management.
Collapse
|
15
|
The reproducibility of clinical OSA subtypes: a population-based longitudinal study. Sleep Breath 2022; 26:1253-1263. [DOI: 10.1007/s11325-021-02470-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022]
|
16
|
Yeo BSY, Koh JH, Tan BKJ, Ding Y, Teo YH, Alkan U, See A, Loh S, Toh ST. Improved Inflammatory and Cardiometabolic Profile After Soft-Tissue Sleep Surgery for Obstructive Sleep Apnea: A Systematic Review and Meta-analysis. JAMA Otolaryngol Head Neck Surg 2022; 148:862-869. [PMID: 35951318 PMCID: PMC9372898 DOI: 10.1001/jamaoto.2022.2285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/23/2022] [Indexed: 11/14/2022]
Abstract
Importance Obstructive sleep apnea (OSA) is associated with a rise in serum inflammatory markers, which may be attenuated by sleep surgery. Objective To evaluate whether sleep surgery was associated with improved levels of proinflammatory markers in adults with OSA. Data Sources Two authors independently searched Cochrane, Embase, and PubMed databases from inception through June 14, 2022. Study Selection Two authors searched the Cochrane, Embase, and PubMed databases for studies comparing preoperative and postoperative levels of serum biomarkers in patients undergoing sleep surgery. Data Extraction and Synthesis Data were extracted from included articles into a structured proforma. Meta-analyses of the standardized mean difference (SMD) were conducted in random-effects models. To ensure relevance to clinicians and patients, the probability of benefit and number needed to treat were calculated for outcomes that demonstrated a statistically significant effect after sleep surgery. Main Outcomes and Measures The primary outcome was the preoperative and postoperative levels of serum biomarkers in patients undergoing sleep surgery, including C-reactive protein (CRP), glucose, low-density lipoprotein (LDL), high-density lipoprotein (HDL), and interleukin-6 (IL-6). Data analysis was performed from April to May 2022. Results Of the 3218 studies screened, 26 studies with 1187 patients (mean [SD] age, 42.8 [11.1] years; 932 [78.5%] men and 255 [21.5%] women) were included. Soft-tissue sleep surgery was associated with a large decrease in CRP (SMD, -0.377; 95% CI, -0.617 to -0.137), total cholesterol (SMD, -0.267; 95% CI, -0.417 to -0.116), LDL (SMD, -0.201; 95% CI, -0.344 to -0.058), IL-6 (SMD, -1.086; 95% CI, -1.952 to -0.221), tumor necrosis factor-α (SMD, -0.822; 95% CI, -1.617 to -0.027), triglyceride (SMD, -0.186; 95% CI, -0.301 to -0.071), and leptin (SMD, -0.519; 95% CI, -0.954 to -0.083) in patients with OSA. Meta-regression highlighted that increased age, higher preoperative score for cumulative sleep time percentage with oxyhemoglobin saturation less than 90% (CT90), and greater change in CT90 postoperatively were associated with a greater decrease in serum CRP levels after soft-tissue sleep surgery. A greater reduction in apnea hypopnea index (AHI) was strongly associated with a greater reduction in total cholesterol and LDL. A greater reduction in body mass index and AHI were also associated with a greater increase in HDL. Conclusions and Relevance The findings of this systematic review and meta-analysis of 26 studies suggest that sleep surgery is associated with decreased levels of CRP, total cholesterol, LDL, triglyceride, IL-6, leptin, and TNF-α, which may improve the inflammatory and cardiometabolic profile of patients who undergo sleep surgery.
Collapse
Affiliation(s)
- Brian Sheng Yep Yeo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jin Hean Koh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Yichen Ding
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yao Hao Teo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Uri Alkan
- Department of Otorhinolaryngology−Head and Neck Surgery, Rabin Medical Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anna See
- Department of Otorhinolaryngology−Head and Neck Surgery, Singapore General Hospital, Singapore
- SingHealth Duke University−National University of Singapore Sleep Centre, Singapore
| | - Shaun Loh
- Department of Otorhinolaryngology−Head and Neck Surgery, Singapore General Hospital, Singapore
- SingHealth Duke University−National University of Singapore Sleep Centre, Singapore
| | - Song Tar Toh
- Department of Otorhinolaryngology−Head and Neck Surgery, Singapore General Hospital, Singapore
- SingHealth Duke University−National University of Singapore Sleep Centre, Singapore
| |
Collapse
|
17
|
Lopez K, Camacho A, Jacquez Q, Amistadi MK, Medina S, Zychowski K. Lung-Based, Exosome Inhibition Mediates Systemic Impacts Following Particulate Matter Exposure. TOXICS 2022; 10:457. [PMID: 36006136 PMCID: PMC9413489 DOI: 10.3390/toxics10080457] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Particulate matter (PM) exposure is a global health issue that impacts both urban and rural communities. Residential communities in the Southwestern United States have expressed concerns regarding the health impacts of fugitive PM from rural, legacy mine-sites. In addition, the recent literature suggests that exosomes may play a role in driving toxicological phenotypes following inhaled exposures. In this study, we assessed exosome-driven mechanisms and systemic health impacts following inhaled dust exposure, using a rodent model. Using an exosome inhibitor, GW4869 (10 μM), we inhibited exosome generation in the lungs of mice via oropharyngeal aspiration. We then exposed mice to previously characterized inhaled particulate matter (PM) from a legacy mine-site and subsequently assessed downstream behavioral, cellular, and molecular biomarkers in lung, serum, and brain tissue. Results indicated that CCL-2 was significantly upregulated in the lung tissue and downregulated in the brain (p < 0.05) following PM exposure. Additional experiments revealed cerebrovascular barrier integrity deficits and increased glial fibrillary acidic protein (GFAP) staining in the mine-PM exposure group, mechanistically dependent on exosome inhibition. An increased stress and anxiety response, based on the open-field test, was noted in the mine-PM exposure group, and subsequently mitigated with GW4869 intervention. Exosome lipidomics revealed 240 and eight significantly altered positive-ion lipids and negative-ion lipids, respectively, across the three treatment groups. Generally, phosphatidylethanolamine (PE) and phosphatidylcholine (PC) lipids were significantly downregulated in the PM group, compared to FA. In conclusion, these data suggest that systemic, toxic impacts of inhaled PM may be mechanistically dependent on lung-derived, circulating exosomes, thereby driving a systemic, proinflammatory phenotype.
Collapse
Affiliation(s)
- Keegan Lopez
- Department of Biology, College of Arts and Sciences, New Mexico Highlands University, Las Vegas, NM 88901, USA
| | - Alexandra Camacho
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA
| | - Quiteria Jacquez
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA
| | - Mary Kay Amistadi
- Arizona Laboratory for Emerging Contaminants, University of Arizona, Tucson, AZ 85721, USA
| | - Sebastian Medina
- Department of Biology, College of Arts and Sciences, New Mexico Highlands University, Las Vegas, NM 88901, USA
| | - Katherine Zychowski
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
18
|
Zhao F, Meng Y, Wang Y, Fan S, Liu Y, Zhang X, Ran C, Wang H, Lu M. Protective effect of Astragaloside IV on chronic intermittent hypoxia-induced vascular endothelial dysfunction through the calpain-1/SIRT1/AMPK signaling pathway. Front Pharmacol 2022; 13:920977. [PMID: 35983375 PMCID: PMC9381017 DOI: 10.3389/fphar.2022.920977] [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: 04/15/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular endothelial dysfunction (VED) is linked with the pathogenesis of obstructive sleep apnea (OSA) comorbidities, such as cardiovascular disease. Astragaloside IV (As-IV) has exhibited significant improvement for endothelial dysfunction. Nonetheless, the protective mechanism is not clear. Therefore, the present study investigated the potential mechanism of As-IV on VED. Calpain-1 knockout and wild-type C57BL/6 mice exposed to chronic intermittent hypoxia (CIH) were established and treated with As-IV (40, 80 mg/kg) for 4 weeks. Human coronary artery endothelial cells (HCAECs) subjected to CIH exposure were pretreated with As-IV, MDL-28170 (calpain-1 inhibitor) and SRT1720 (SIRT1 activator) for 48 h in vitro. The endothelial function, inflammation, oxidative stress and mitochondrial function were measured to evaluate VED. Our data revealed that As-IV treatment ameliorated CIH-induced endothelial-dependent vasomotion and augmented nitric oxide (NO) production. As-IV administration suppressed the secretion of inflammation, oxidative stress and mitochondrial dysfunction. As-IV treatment reduced the expression of calpain-1 and restored the downregulated expression of SIRT1 and Thr172 AMPK and Ser1177 eNOS phosphorylation. The effects of calpain-1 knockout and SRT1720 were similar to the effect of As-IV on VED. These findings demonstrated that As-IV ameliorated VED induced by chronic intermittent hypoxia via the calpain-1/SIRT1/AMPK signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Meili Lu
- *Correspondence: Hongxin Wang, ; Meili Lu,
| |
Collapse
|
19
|
Harki O, Bouyon S, Sallé M, Arco-Hierves A, Lemarié E, Demory A, Chirica C, Vilgrain I, Pépin JL, Faury G, Briançon-Marjollet A. Inhibition of Vascular Endothelial Cadherin Cleavage Prevents Elastic Fiber Alterations and Atherosclerosis Induced by Intermittent Hypoxia in the Mouse Aorta. Int J Mol Sci 2022; 23:ijms23137012. [PMID: 35806017 PMCID: PMC9266969 DOI: 10.3390/ijms23137012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Intermittent hypoxia (IH), the major feature of obstructive sleep apnea syndrome (OSAS), induces atherosclerosis and elastic fiber alterations. VE-cadherin cleavage is increased in OSAS patients and in an IH-cellular model. It is mediated by HIF-1 and Src-tyr-kinases pathways and results in endothelial hyperpermeability. Our aim was to determine whether blocking VE-cadherin cleavage in vivo could be an efficient strategy to inhibit deleterious IH-induced vascular remodeling, elastic fiber defects and atherogenesis. VE-cadherin regulation, aortic remodeling and atherosclerosis were studied in IH-exposed C57Bl/6J or ApoE-/-mice treated or not with Src-tyr-kinases inhibitors (Saracatinib/Pazopanib) or a HIF-1 inhibitor (Acriflavine). Human aortic endothelial cells were exposed to IH and treated with the same inhibitors. LDL and the monocytes transendothelium passage were measured. In vitro, IH increased transendothelium LDL and monocytes passage, and the tested inhibitors prevented these effects. In mice, IH decreased VE-cadherin expression and increased plasmatic sVE level, intima-media thickness, elastic fiber alterations and atherosclerosis, while the inhibitors prevented these in vivo effects. In vivo inhibition of HIF-1 and Src tyr kinase pathways were associated with the prevention of IH-induced elastic fiber/lamella degradation and atherogenesis, which suggests that VE-cadherin could be an important target to limit atherogenesis and progression of arterial stiffness in OSAS.
Collapse
Affiliation(s)
- Olfa Harki
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Sophie Bouyon
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Marine Sallé
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Alejandro Arco-Hierves
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Emeline Lemarié
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Alexandra Demory
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Carole Chirica
- Unité Biochimie Immunoanalyse, Service de Biochimie SB2TE, CHU Grenoble Alpes, 38000 Grenoble, France;
| | - Isabelle Vilgrain
- Université Grenoble Alpes, INSERM U1292, CEA, 38042 Grenoble, France;
| | - Jean-Louis Pépin
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| | - Gilles Faury
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
- Correspondence:
| | - Anne Briançon-Marjollet
- Université Grenoble Alpes, INSERM U1300, CHU Grenoble Alpes, Laboratoire HP2, 38042 Grenoble, France; (O.H.); (S.B.); (M.S.); (A.A.-H.); (E.L.); (A.D.); (J.-L.P.); (A.B.-M.)
| |
Collapse
|
20
|
Cheng W, Pang H, Campen MJ, Zhang J, Li Y, Gao J, Ren D, Ji X, Rothman N, Lan Q, Zheng Y, Leng S, Hu Z, Tang J. Circulatory metabolites trigger ex vivo arterial endothelial cell dysfunction in population chronically exposed to diesel exhaust. Part Fibre Toxicol 2022; 19:20. [PMID: 35313899 PMCID: PMC8939222 DOI: 10.1186/s12989-022-00463-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/15/2022] [Indexed: 11/27/2022] Open
Abstract
Background Chronic exposure to diesel exhaust has a causal link to cardiovascular diseases in various environmental and occupational settings. Arterial endothelial cell function plays an important role in ensuring proper maintenance of cardiovascular homeostasis and the endothelial cell dysfunction by circulatory inflammation is a hallmark in cardiovascular diseases. Acute exposure to diesel exhaust in controlled exposure studies leads to artery endothelial cells dysfunction in previous study, however the effect of chronic exposure remains unknown. Results We applied an ex vivo endothelial biosensor assay for serum samples from 133 diesel engine testers (DETs) and 126 non-DETs with the aim of identifying evidence of increased risk for cardiovascular diseases. Environmental monitoring suggested that DETs were exposed to high levels of diesel exhaust aerosol (282.3 μg/m3 PM2.5 and 135.2 μg/m3 elemental carbon). Surprisingly, chronic diesel exhaust exposure was associated with a pro-inflammatory phenotype in the ex vivo endothelial cell model, in a dose-dependent manner with CCL5 and VCAM as most affected genes. This dysfunction was not mediated by reduction in circulatory pro-inflammatory factors but significantly associated with a reduction in circulatory metabolites cGMP and an increase in primary DNA damage in leucocyte in a dose-dependent manner, which also explained a large magnitude of association between diesel exhaust exposure and ex vivo endothelial biosensor response. Exogenous cGMP addition experiment further confirmed the induction of ex vivo biosensor gene expressions in endothelial cells treated with physiologically relevant levels of metabolites cGMP. Conclusion Serum-borne bioactivity caused the arterial endothelial cell dysfunction may attribute to the circulatory metabolites based on the ex vivo biosensor assay. The reduced cGMP and increased polycyclic aromatic hydrocarbons metabolites-induced cyto/geno-toxic play important role in the endothelial cell dysfunction of workers chronic exposure to diesel exhaust. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-022-00463-0.
Collapse
Affiliation(s)
- Wenting Cheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, Shandong, China
| | - Huanhuan Pang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Jianzhong Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, Shandong, China
| | - Yanting Li
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, Shandong, China
| | - Jinling Gao
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, Shandong, China
| | - Dunqiang Ren
- Department of Respiratory Medicine, Affiliated Hospital of Medical College of Qingdao University, Qingdao University, Qingdao, 266021, Shandong, China
| | - Xiaoya Ji
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, Shandong, China
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, Shandong, China
| | - Shuguang Leng
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA. .,Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA.
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Jinglong Tang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, Shandong, China.
| |
Collapse
|
21
|
Ass’ad NA, Shore X, Myers O, Camacho AR, Jacquez Q, Pollard C, Cook LS, Leng S, Page K, Sood A, Zychowski KE. VCAM-1 Is Upregulated in Uranium Miners Compared to Other Miners. Life (Basel) 2021; 11:1223. [PMID: 34833099 PMCID: PMC8621685 DOI: 10.3390/life11111223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 12/01/2022] Open
Abstract
The United States has a rich history of mining including uranium (U)-mining, coal mining, and other metal mining. Cardiovascular diseases (CVD) are largely understudied in miners and recent literature suggests that when compared to non-U miners, U-miners are more likely to report CVD. However, the molecular basis for this phenomenon is currently unknown. In this pilot study, a New Mexico (NM)-based occupational cohort of current and former miners (n = 44) were recruited via a mobile screening clinic for miners. Serum- and endothelial-based endpoints were used to assess circulating inflammatory potential relevant to CVD. Non-U miners reported significantly fewer pack years of smoking than U-miners. Circulating biomarkers of interest revealed that U-miners had significantly greater serum amyloid A (SAA), soluble intercellular adhesion molecule 1 (ICAM-1, ng/mL), soluble vascular cell adhesion molecule 1 (VCAM-1, ng/mL), and VCAM-1 mRNA expression, as determined by the serum cumulative inflammatory potential (SCIP) assay, an endothelial-based assay. Even after adjusting for various covariates, including age, multivariable analysis determined that U-miners had significantly upregulated VCAM-1 mRNA. In conclusion, VCAM-1 may be an important biomarker and possible contributor of CVD in U-miners. Further research to explore this mechanism may be warranted.
Collapse
Affiliation(s)
- Nour A. Ass’ad
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (N.A.A.); (L.S.C.); (S.L.); (K.P.); (A.S.)
| | - Xin Shore
- Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (X.S.); (O.M.)
| | - Orrin Myers
- Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (X.S.); (O.M.)
| | - Alexandra R. Camacho
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA; (A.R.C.); (Q.J.)
| | - Quiteria Jacquez
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA; (A.R.C.); (Q.J.)
| | - Charles Pollard
- Miners’ Colfax Medical Center, 203 Hospital Drive, Raton, NM 87740, USA;
| | - Linda S. Cook
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (N.A.A.); (L.S.C.); (S.L.); (K.P.); (A.S.)
- Department of Epidemiology, School of Public Health, University of Colorado-Anschutz, Arora, CO 80045, USA
| | - Shuguang Leng
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (N.A.A.); (L.S.C.); (S.L.); (K.P.); (A.S.)
| | - Kimberly Page
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (N.A.A.); (L.S.C.); (S.L.); (K.P.); (A.S.)
| | - Akshay Sood
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (N.A.A.); (L.S.C.); (S.L.); (K.P.); (A.S.)
- Miners’ Colfax Medical Center, 203 Hospital Drive, Raton, NM 87740, USA;
| | - Katherine E. Zychowski
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, NM 87131, USA; (A.R.C.); (Q.J.)
| |
Collapse
|
22
|
Mostovenko E, Dahm MM, Schubauer-Berigan MK, Eye T, Erdely A, Young TL, Campen MJ, Ottens AK. Serum peptidome: diagnostic window into pathogenic processes following occupational exposure to carbon nanomaterials. Part Fibre Toxicol 2021; 18:39. [PMID: 34711247 PMCID: PMC8555107 DOI: 10.1186/s12989-021-00431-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Growing industrial use of carbon nanotubes and nanofibers (CNT/F) warrants consideration of human health outcomes. CNT/F produces pulmonary, cardiovascular, and other toxic effects in animals along with a significant release of bioactive peptides into the circulation, the augmented serum peptidome. While epidemiology among CNT/F workers reports on few acute symptoms, there remains concern over sub-clinical CNT/F effects that may prime for chronic disease, necessitating sensitive health outcome diagnostic markers for longitudinal follow-up. METHODS Here, the serum peptidome was assessed for its biomarker potential in detecting sub-symptomatic pathobiology among CNT/F workers using label-free data-independent mass spectrometry. Studies employed a stratified design between High (> 0.5 µg/m3) and Low (< 0.1 µg/m3) inhalable CNT/F exposures in the industrial setting. Peptide biomarker model building and refinement employed linear regression and partial least squared discriminant analyses. Top-ranked peptides were then sequence identified and evaluated for pathological-relevance. RESULTS In total, 41 peptides were found to be highly discriminatory after model building with a strong linear correlation to personal CNT/F exposure. The top-five peptide model offered ideal prediction with high accuracy (Q2 = 0.99916). Unsupervised validation affirmed 43.5% of the serum peptidomic variance was attributable to CNT/F exposure. Peptide sequence identification reveals a predominant association with vascular pathology. ARHGAP21, ADAM15 and PLPP3 peptides suggest heightened cardiovasculature permeability and F13A1, FBN1 and VWDE peptides infer a pro-thrombotic state among High CNT/F workers. CONCLUSIONS The serum peptidome affords a diagnostic window into sub-symptomatic pathology among CNT/F exposed workers for longitudinal monitoring of systemic health risks.
Collapse
Affiliation(s)
- Ekaterina Mostovenko
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298, USA
| | - Matthew M Dahm
- Division of Field Studies and Engineering, National Institute for Occupational Safety and Health, 1090 Tusculum Avenue, MS-R12, Cincinnati, OH, 45226, USA
| | - Mary K Schubauer-Berigan
- Division of Field Studies and Engineering, National Institute for Occupational Safety and Health, 1090 Tusculum Avenue, MS-R12, Cincinnati, OH, 45226, USA
- Evidence Synthesis and Classification Section, International Agency for Research On Cancer, 150 Cours Albert Thomas, 69372, Lyon, CEDEX 08, France
| | - Tracy Eye
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1095 Willowdale Road, MS-2015, Morgantown, WV, 26505, USA
| | - Aaron Erdely
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, 1095 Willowdale Road, MS-2015, Morgantown, WV, 26505, USA
| | - Tamara L Young
- Department of Pharmaceutical Sciences, University of New Mexico, MSC09 53601, Albuquerque, NM, 87131, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico, MSC09 53601, Albuquerque, NM, 87131, USA
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298, USA.
| |
Collapse
|
23
|
Young TL, Mostovenko E, Denson JL, Begay JG, Lucas SN, Herbert G, Zychowski K, Hunter R, Salazar R, Wang T, Fraser K, Erdely A, Ottens AK, Campen MJ. Pulmonary delivery of the broad-spectrum matrix metalloproteinase inhibitor marimastat diminishes multiwalled carbon nanotube-induced circulating bioactivity without reducing pulmonary inflammation. Part Fibre Toxicol 2021; 18:34. [PMID: 34496918 PMCID: PMC8424988 DOI: 10.1186/s12989-021-00427-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/27/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Multiwalled carbon nanotubes (MWCNT) are an increasingly utilized engineered nanomaterial that pose the potential for significant risk of exposure-related health outcomes. The mechanism(s) underlying MWCNT-induced toxicity to extrapulmonary sites are still being defined. MWCNT-induced serum-borne bioactivity appears to dysregulate systemic endothelial cell function. The serum compositional changes after MWCNT exposure have been identified as a surge of fragmented endogenous peptides, likely derived from matrix metalloproteinase (MMP) activity. In the present study, we utilize a broad-spectrum MMP inhibitor, Marimastat, along with a previously described oropharyngeal aspiration model of MWCNT administration to investigate the role of MMPs in MWCNT-derived serum peptide generation and endothelial bioactivity. RESULTS C57BL/6 mice were treated with Marimastat or vehicle by oropharyngeal aspiration 1 h prior to MWCNT treatment. Pulmonary neutrophil infiltration and total bronchoalveolar lavage fluid protein increased independent of MMP blockade. The lung cytokine profile similarly increased following MWCNT exposure for major inflammatory markers (IL-1β, IL-6, and TNF-α), with minimal impact from MMP inhibition. However, serum peptidomic analysis revealed differential peptide compositional profiles, with MMP blockade abrogating MWCNT-derived serum peptide fragments. The serum, in turn, exhibited differential potency in terms of inflammatory bioactivity when incubated with primary murine cerebrovascular endothelial cells. Serum from MWCNT-treated mice led to inflammatory responses in endothelial cells that were significantly blunted with serum from Marimastat-treated mice. CONCLUSIONS Thus, MWCNT exposure induced pulmonary inflammation that was largely independent of MMP activity but generated circulating bioactive peptides through predominantly MMP-dependent pathways. This MWCNT-induced lung-derived bioactivity caused pathological consequences of endothelial inflammation and barrier disruption.
Collapse
Affiliation(s)
- Tamara L Young
- Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Ekaterina Mostovenko
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298, USA
| | - Jesse L Denson
- Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Jessica G Begay
- Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Selita N Lucas
- Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Guy Herbert
- Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | | | - Russell Hunter
- Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Raul Salazar
- Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Ting Wang
- Department of Internal Medicine, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Kelly Fraser
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, Richmond, VA, 23298, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, MSC09 5360, 1 University of New Mexico, Albuquerque, NM, 87131-0001, USA.
| |
Collapse
|
24
|
Lv Q, Sun H, Du Z, Jiao X, Yu H, Sun Q, Li F, Wang Y, Li L, Hu C, Qin Y. Increased levels of VCAM-1 is associated with higher occurrence of coronary artery disease in adults with moderate to severe obstructive sleep apnea. Sleep Med 2021; 85:131-137. [PMID: 34325382 DOI: 10.1016/j.sleep.2021.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) leads to important vascular abnormalities, including the endothelial dysfunction and the production of endothelial cell adhesion molecules. The adhesion molecules play an important role in the process of endothelial dysfunction in the pathogenesis of atherosclerosis. We assess the relationship between the levels of adhesion molecules and the presence of coronary artery disease (CAD) in Chinese adults with moderate to severe OSA. METHODS The cross-sectional study included a total of 189 Chinese adults: 90 patients with moderate to severe OSA (apnea-hypopnea index≥15 events/h) alone, 40 patients with moderate to severe OSA and CAD, and 59 controls without OSA or with mild OSA and without CAD. We used high-throughput Multiplex Immunobead Assay technology to simultaneously test plasma levels of vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1). The associations between the levels of circulating adhesion molecules and CAD in moderate to severe OSA patients were evaluated by multivariate logistic regression analysis. RESULTS The circulating VCAM-1 levels were significantly elevated in patients suffering from moderate to severe OSA combined CAD compared with patients having moderate to severe OSA alone [853.28 (564.26) vs. 416.61 (301.69) ng/mL, P < 0.001]. Furthermore, circulating VCAM-1 levels were independently associated with CAD (odds ration = 2.113, 95%CI 1.400-2.766, P < 0.001) and showed higher discriminatory accuracy in assessing the presence of CAD (AUC: 0.899, 95%CI 0.849-0.950, P < 0.001) in moderate to severe OSA patients. However, no significant association was found between circulating ICAM-1 levels and CAD in moderate to severe OSA patients. CONCLUSIONS The circulating VCAM-1 levels were significantly correlated with the presence of CAD in Chinese adults with moderate to severe OSA. The circulating VCAM-1 may function as a novel biomarker for monitoring the development and progression of CAD in patients with moderate to severe OSA.
Collapse
Affiliation(s)
- Qianwen Lv
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Haili Sun
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Zhiyong Du
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Xiaolu Jiao
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Huahui Yu
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Qiuju Sun
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Fan Li
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yu Wang
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Linyi Li
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Chaowei Hu
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yanwen Qin
- Key Laboratory of Upper Airway Dysfunction-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China.
| |
Collapse
|
25
|
Sanz-Rubio D, Khalyfa A, Qiao Z, Ullate J, Marin JM, Kheirandish-Gozal L, Gozal D. Cell-Selective Altered Cargo Properties of Extracellular Vesicles Following In Vitro Exposures to Intermittent Hypoxia. Int J Mol Sci 2021; 22:ijms22115604. [PMID: 34070558 PMCID: PMC8198838 DOI: 10.3390/ijms22115604] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/09/2021] [Accepted: 05/19/2021] [Indexed: 01/09/2023] Open
Abstract
Intermittent hypoxia (IH), a hallmark of obstructive sleep apnea (OSA), is associated with cardiovascular and metabolic dysfunction. However, the mechanisms underlying these morbidities remain poorly delineated. Extracellular vesicles (EVs) mediate intercellular communications, play pivotal roles in a multitude of physiological and pathological processes, and could mediate IH-induced cellular effects. Here, the effects of IH on human primary cells and the release of EVs were examined. Microvascular endothelial cells (HMVEC-d), THP1 monocytes, THP1 macrophages M0, THP1 macrophages M1, THP1 macrophages M2, pre-adipocytes, and differentiated adipocytes (HAd) were exposed to either room air (RA) or IH for 24 h. Secreted EVs were isolated and characterized using transmission electron microscopy, nanoparticle tracking analysis, and Western blotting. The effects of each of the cell-derived EVs on endothelial cell (EC) monolayer barrier integrity, on naïve THP1 macrophage polarity, and on adipocyte insulin sensitivity were also evaluated. IH did not alter EVs cell quantal release, but IH-EVs derived from HMVEC-d (p < 0.01), THP1 M0 (p < 0.01) and HAd (p < 0.05) significantly disrupted HMVEC-d monolayer integrity, particularly after H2O2 pre-conditioning. IH-EVs from HMVEC-d and THP1 M0 elicited M2-polarity changes did not alter insulin sensitivity responses. IH induces cell-selective changes in EVs cargo, which primarily seem to target the emergence of endothelial dysfunction. Thus, changes in EVs cargo from selected cell sources in vivo may play causal roles in some of the adverse outcomes associated with OSA.
Collapse
Affiliation(s)
- David Sanz-Rubio
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
- Translational Research Unit, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón (IISAragón), 50009 Zaragoza, Spain;
| | - Abdelnaby Khalyfa
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
- Correspondence: ; Tel.: +1-573-884-7685
| | - Zhuanhong Qiao
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
| | - Jorge Ullate
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
| | - José M. Marin
- Translational Research Unit, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón (IISAragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERes), 28029 Madrid, Spain
| | - Leila Kheirandish-Gozal
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
| | - David Gozal
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
| |
Collapse
|
26
|
Elevation of CD40/CD40L Inflammatory Pathway Molecules in Carotid Plaques from Moderate-and-Severe Obstructive Sleep Apnea Patients. Diagnostics (Basel) 2021; 11:diagnostics11060935. [PMID: 34067481 PMCID: PMC8224789 DOI: 10.3390/diagnostics11060935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 11/30/2022] Open
Abstract
A chronic inflammatory process characteristic of obstructive sleep apnea promotes vascular endothelial dysfunction and atherogenesis. This process can lead to destabilization and rupture of cardiovascular plaques, which clinically manifests as an acute coronary syndrome or stroke. The aim of this study was to investigate the inflammatory pathway leading to plaque destabilization in non-to-mild and moderate-to-severe groups of OSA patients. This prospective study involved enrollment of patients scheduled for endarterectomy. A sleep study was performed prior to surgery. Immunohistochemistry was performed on atherosclerotic plaques from carotid arteries obtained during standard open endarterectomy to determine levels of CD40, CD40L receptors, MCP-1, and MMP-9. The 46 patients included 14 controls, 13 with mild, 11 with moderate, and 8 with severe OSA. Increased expression of CD40, CD40L receptors, MCP-1, and MMP-9 were found to be proportionate with OSA severity. However, significant differences among groups were observed only for MCP-1 (p = 0.014). Increased expression of inflammatory markers (CD40, CD40L, MCP-1, MMP-9) is associated with increasing OSA severity. This suggests the CD40-CD4-L inflammatory pathway may contribute to plaque instability and rupture in OSA patients.
Collapse
|
27
|
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
|
28
|
Tang J, Cheng W, Gao J, Li Y, Yao R, Rothman N, Lan Q, Campen MJ, Zheng Y, Leng S. Occupational exposure to carbon black nanoparticles increases inflammatory vascular disease risk: an implication of an ex vivo biosensor assay. Part Fibre Toxicol 2020; 17:47. [PMID: 32993720 PMCID: PMC7523398 DOI: 10.1186/s12989-020-00378-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/04/2020] [Indexed: 11/10/2022] Open
Abstract
Background Among manufactured or engineered nanoparticles, carbon black (CB) has largest production worldwide and is also an occupational respiratory hazard commonly seen in rubber industry. Few studies have assessed the risk for cardiovascular disease in carbon black exposed populations. An endothelial biosensor assay was used to quantify the capacity of sera from 82 carbon black packers (CBP) and 106 non-CBPs to induce endothelial cell activation ex vivo. The mediation effect of circulatory proinflammatory factors on the association between carbon black exposure and endothelial cell activation was assessed and further validated using in vitro intervention experiments. Results The average elemental carbon level inside carbon black bagging facilities was 657.0 μg/m3, which was 164-fold higher than that seen in reference areas (4.0 μg/m3). A global index was extracted from mRNA expression of seven candidate biosensor genes using principal component analysis and used to quantify the magnitude of endothelial cell activation. This global index was found to be significantly altered in CBPs compared to non-CBPs (P < 0.0001), however this difference did not vary by smoking status (P = 0.74). Individual gene analyses identified that de novo expression of key adhesion molecules (e.g., ICAM and VCAM) and chemotactic factors (e.g., CCL2, CCL5, and CXCL8) responsible for the recruitment of leukocytes was dramatically induced in CBPs with CXCL8 showing the highest fold of induction (relative quantification = 9.1, P < 0.0001). The combination of mediation analyses and in vitro functional validation confirmed TNF-α, IL-1β, and IL-6 as important circulatory factors mediating the effects of carbon black exposure on endothelial cell activation responses. Conclusions Inflammatory mediators in sera from CBPs may bridge carbon black exposure and endothelial cell activation response assessed ex vivo. CBPs may have elevated risk for cardiovascular diseases when comorbidity exists. Our study may serve as a benchmark for understanding health effects of engineered carbon based nanoparticles with environmental and occupational health relevance.
Collapse
Affiliation(s)
- Jinglong Tang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, China
| | - Wenting Cheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, China
| | - Jinling Gao
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, China
| | - Yanting Li
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, China
| | - Ruyong Yao
- Department of Central Laboratory, Affiliated Hospital of Medical College of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, 87131, USA
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, China.
| | - Shuguang Leng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, 266021, China. .,Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA. .,Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA.
| |
Collapse
|
29
|
Kodali V, Shoeb M, Meighan TG, Eye T, Friend SA, Hubczak J, Kashon ML, Zeidler-Erdely PC, Antonini JM, Erdely A. Bioactivity of Circulatory Factors After Pulmonary Exposure to Mild or Stainless Steel Welding Fumes. Toxicol Sci 2020; 177:108-120. [PMID: 32514565 DOI: 10.1093/toxsci/kfaa084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Studies suggest that alterations in circulating factors are a driver of pulmonary-induced cardiovascular dysfunction. To evaluate, if circulating factors effect endothelial function after a pulmonary exposure to welding fumes, an exposure known to induce cardiovascular dysfunction, serum collected from Sprague Dawley rats 24 h after an intratracheal instillation exposure to 2 mg/rat of 2 compositionally distinct metal-rich welding fume particulates (manual metal arc welding using stainless steel electrodes [MMA-SS] or gas metal arc welding using mild steel electrodes [GMA-MS]) or saline was used to test molecular and functional effects of in vitro cultures of primary cardiac microvascular endothelial cells (PCMEs) or ex vivo organ cultures. The welding fumes elicited significant pulmonary injury and inflammation with only minor changes in measured serum antioxidant and cytokine levels. PCME cells were challenged for 4 h with serum collected from exposed rats, and 84 genes related to endothelial function were analyzed. Changes in relative mRNA patterns indicated that serum from rats exposed to MMA-SS, and not GMA-MS or PBS, could influence several functional aspects related to endothelial cells, including cell migration, angiogenesis, inflammation, and vascular function. The predictions were confirmed using a functional in vitro assay (scratch assay) as well as an ex vivo multicellular environment (aortic ring angiogenesis assay), validating the concept that endothelial cells can be used as an effective screening tool of exposed workers for determining bioactivity of altered circulatory factors. Overall, the results indicate that pulmonary MMA-SS fume exposure can cause altered endothelial function systemically via altered circulating factors.
Collapse
Affiliation(s)
- Vamsi Kodali
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| | - Mohammad Shoeb
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| | - Terence G Meighan
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| | - Tracy Eye
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| | - Sherri A Friend
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| | - John Hubczak
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| | - Michael L Kashon
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| | | | - James M Antonini
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| | - Aaron Erdely
- Health Effects Laboratory Division, NIOSH, Morgantown, West Virginia 26505-2888
| |
Collapse
|
30
|
Sanchez B, Zhou X, Gardiner AS, Herbert G, Lucas S, Morishita M, Wagner JG, Lewandowski R, Harkema JR, Shuey C, Campen MJ, Zychowski KE. Serum-borne factors alter cerebrovascular endothelial microRNA expression following particulate matter exposure near an abandoned uranium mine on the Navajo Nation. Part Fibre Toxicol 2020; 17:29. [PMID: 32611356 PMCID: PMC7329534 DOI: 10.1186/s12989-020-00361-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 06/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background Commercial uranium mining on the Navajo Nation has subjected communities on tribal lands in the Southwestern United States to exposures from residual environmental contamination. Vascular health effects from these ongoing exposures are an active area of study. There is an association between residential mine-site proximity and circulating biomarkers in residents, however, the contribution of mine-site derived wind-blown dusts on vascular and other health outcomes is unknown. To assess neurovascular effects of mine-site derived dusts, we exposed mice using a novel exposure paradigm, the AirCARE1 mobile inhalation laboratory, located 2 km from an abandoned uranium mine, Claim 28 in Blue Gap Tachee, AZ. Mice were exposed to filtered air (FA) (n = 6) or concentrated ambient particulate matter (CAPs) (n = 5) for 2 wks for 4 h per day. Results To assess miRNA differential expression in cultured mouse cerebrovascular cells following particulate matter (PM) exposure (average: 96.6 ± 60.4 μg/m3 for all 4 h exposures), the serum cumulative inflammatory potential (SCIP) assay was employed. MiRNA sequencing was then performed in cultured mouse cerebrovascular endothelial cells (mCECs) to evaluate transcriptional changes. Results indicated 27 highly differentially expressed (p < 0.01) murine miRNAs, as measured in the SCIP assay. Gene ontology (GO) pathway analysis revealed notable alterations in GO enrichment related to the cytoplasm, protein binding and the cytosol, while significant KEGG pathways involved pathways in cancer, axon guidance and Wnt signaling. Expression of these 27 identified, differentially expressed murine miRNAs were then evaluated in the serum. Nine of these miRNAs (~ 30%) were significantly altered in the serum and 8 of those miRNAs demonstrated the same directional change (either upregulation or downregulation) as cellular miRNAs, as measured in the SCIP assay. Significantly upregulated miRNAs in the CAPs exposure group included miRNAs in the let-7a family. Overexpression of mmu-let-7a via transfection experiments, suggested that this miRNA may mediate mCEC barrier integrity following dust exposure. Conclusions Our data suggest that mCEC miRNAs as measured in the SCIP assay show similarity to serum-borne miRNAs, as approximately 30% of highly differentially expressed cellular miRNAs in the SCIP assay were also found in the serum. While translocation of miRNAs via exosomes or an alternative mechanism is certainly possible, other yet-to-be-identified factors in the serum may be responsible for significant miRNA differential expression in endothelium following inhaled exposures. Additionally, the most highly upregulated murine miRNAs in the CAPs exposure group were in the let-7a family. These miRNAs play a prominent role in cell growth and differentiation and based on our transfection experiments, mmu-let-7a may contribute to cerebrovascular mCEC alterations following inhaled dust exposure.
Collapse
Affiliation(s)
- Bethany Sanchez
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Xixi Zhou
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Amy S Gardiner
- Department of Cell Biology and Physiology, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Selita Lucas
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Masako Morishita
- Department of Family Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - James G Wagner
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Ryan Lewandowski
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Jack R Harkema
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Chris Shuey
- Southwest Research and Information Center, Albuquerque, NM, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Katherine E Zychowski
- College of Nursing, MSC09 53601 University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA.
| |
Collapse
|
31
|
Recent advances in the management of secondary hypertension—obstructive sleep apnea. Hypertens Res 2020; 43:1338-1343. [DOI: 10.1038/s41440-020-0494-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 12/22/2022]
|
32
|
The effect of in-lab polysomnography and home sleep polygraphy on sleep position. Sleep Breath 2020; 25:251-255. [PMID: 32418018 PMCID: PMC7987683 DOI: 10.1007/s11325-020-02099-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/26/2020] [Accepted: 05/01/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE Little is known regarding the influence of in-laboratory polysomnography (PSG) equipment on sleep position, especially on the prevalence of supine positioning, which in many cases may lead to a more severe sleep apnea diagnosis. The aim of this study was to assess the percentage of supine sleep during an in-laboratory PSG compared to that seen during a home sleep apnea test (HSAT). METHODS This was a retrospective cohort study comparing in-laboratory PSG and HSAT using a peripheral arterial tone (PAT) technology device. RESULTS Of 445 PSG and 416 HSAT studies analyzed, there was no significant difference in the proportion of supine sleep time between PSG (44%) and HSAT (45%, p = 0.53). Analysis of the differences in sleep position (supine versus non-supine), analyzed by sex, BMI (≥ 30 kg/m2 versus < 30 kg/m2), and age (≥ 60 years versus < 60 years), was significant only for women, who had more supine sleep during HSAT at 61 ± 24% than during PSG at 45 ± 26% (p < 0.001). CONCLUSION Overall there was no difference in the percentage of supine sleep when comparing in-laboratory PSG to HSAT. However, women had more supine sleep with HSAT than with PSG.
Collapse
|
33
|
Zychowski KE, Wheeler A, Sanchez B, Harmon M, Steadman Tyler CR, Herbert G, Lucas SN, Ali AM, Avasarala S, Kunda N, Robinson P, Muttil P, Cerrato JM, Bleske B, Smirnova O, Campen MJ. Toxic Effects of Particulate Matter Derived from Dust Samples Near the Dzhidinski Ore Processing Mill, Eastern Siberia, Russia. Cardiovasc Toxicol 2020; 19:401-411. [PMID: 30963444 DOI: 10.1007/s12012-019-09507-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ambient particulate matter (PM) is associated with increased mortality and morbidity, an effect influenced by the metal components of the PM. We characterized five sediment samples obtained near a tungsten-molybdenum ore-processing complex in Zakamensk, Russia for elemental composition and PM toxicity with regard to pulmonary, vascular, and neurological outcomes. Elemental and trace metals analysis of complete sediment and PM10 (the respirable fraction, < 10 µm mass mean aerodynamic diameter) were performed using inductively coupled plasma optical emission spectrometry (ICP-OES) and mass spectrometry (ICP-MS). Sediment samples and PM10 consisted largely of silicon and iron and silicon and sodium, respectively. Trace metals including manganese and uranium in the complete sediment, as well as copper and lead in the PM10 were observed. Notably, metal concentrations were approximately 10 × higher in the PM10 than in the sediment. Exposure to 100 µg of PM10 via oropharyngeal aspiration in C56BL/6 mice resulted in pulmonary inflammation across all groups. In addition, mice exposed to three of the five PM10 samples exhibited impaired endothelial-dependent relaxation, and correlative analysis revealed associations between pulmonary inflammation and levels of lead and cadmium. A tendency for elevated cortical ccl2 and Tnf-α mRNA expression was induced by all samples and significant upregulation was noted following exposure to PM10 samples Z3 and Z4, respectively. Cortical Nqo1 mRNA levels were significantly upregulated in mice exposed to PM10 Z2. In conclusion, pulmonary exposure to PM samples from the Zakamensk region sediments induced varied pulmonary and systemic effects that may be influenced by elemental PM composition. Further investigation is needed to pinpoint putative drivers of neurological outcomes.
Collapse
Affiliation(s)
- Katherine E Zychowski
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA
| | - Abigail Wheeler
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA
| | - Bethany Sanchez
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA
| | - Molly Harmon
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA
| | | | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA
| | - Selita N Lucas
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA
| | - Abdul-Mehdi Ali
- Department of Civil Engineering, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Sumant Avasarala
- Department of Civil Engineering, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Nitesh Kunda
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA
| | - Paul Robinson
- Southwest Research and Information Center, Albuquerque, NM, 87196, USA
| | - Pavan Muttil
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA
| | - Jose M Cerrato
- Department of Civil Engineering, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Barry Bleske
- Pharmacy Practice and Administrative Sciences, University of New Mexico-Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Olga Smirnova
- Geological Institute, Siberian Branch, Russian Academy of Sciences, Moscow, Russia
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico-Health Sciences Center, MSC09 5360, Albuquerque, NM, 87131, USA.
| |
Collapse
|
34
|
Lian N, Zhang S, Huang J, Lin T, Lin Q. Resveratrol Attenuates Intermittent Hypoxia-Induced Lung Injury by Activating the Nrf2/ARE Pathway. Lung 2020; 198:323-331. [DOI: 10.1007/s00408-020-00321-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/02/2020] [Indexed: 12/22/2022]
|
35
|
Friedlander AH, Boström KI, Tran HA, Chang TI, Polanco JC, Lee UK. Severe Sleep Apnea Associated With Increased Systemic Inflammation and Decreased Serum Bilirubin. J Oral Maxillofac Surg 2019; 77:2318-2323. [DOI: 10.1016/j.joms.2019.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 11/16/2022]
|
36
|
Mostovenko E, Young T, Muldoon PP, Bishop L, Canal CG, Vucetic A, Zeidler-Erdely PC, Erdely A, Campen MJ, Ottens AK. Nanoparticle exposure driven circulating bioactive peptidome causes systemic inflammation and vascular dysfunction. Part Fibre Toxicol 2019; 16:20. [PMID: 31142334 PMCID: PMC6542040 DOI: 10.1186/s12989-019-0304-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background The mechanisms driving systemic effects consequent pulmonary nanoparticle exposure remain unclear. Recent work has established the existence of an indirect process by which factors released from the lung into the circulation promote systemic inflammation and cellular dysfunction, particularly on the vasculature. However, the composition of circulating contributing factors and how they are produced remains unknown. Evidence suggests matrix protease involvement; thus, here we used a well-characterized multi-walled carbon nanotube (MWCNT) oropharyngeal aspiration model with known vascular effects to assess the distinct contribution of nanoparticle-induced peptide fragments in driving systemic pathobiology. Results Data-independent mass spectrometry enabled the unbiased quantitative characterization of 841 significant MWCNT-responses within an enriched peptide fraction, with 567 of these factors demonstrating significant correlation across animal-paired bronchoalveolar lavage and serum biofluids. A database search curated for known matrix protease substrates and predicted signaling motifs enabled identification of 73 MWCNT-responsive peptides, which were significantly associated with an abnormal cardiovascular phenotype, extracellular matrix organization, immune-inflammatory processes, cell receptor signaling, and a MWCNT-altered serum exosome population. Production of a diverse peptidomic response was supported by a wide number of upregulated matrix and lysosomal proteases in the lung after MWCNT exposure. The peptide fraction was then found bioactive, producing endothelial cell inflammation and vascular dysfunction ex vivo akin to that induced with whole serum. Results implicate receptor ligand functionality in driving systemic effects, exemplified by an identified 59-mer thrombospondin fragment, replete with CD36 modulatory motifs, that when synthesized produced an anti-angiogenic response in vitro matching that of the peptide fraction. Other identified peptides point to integrin ligand functionality and more broadly to a diversity of receptor-mediated bioactivity induced by the peptidomic response to nanoparticle exposure. Conclusion The present study demonstrates that pulmonary-sequestered nanoparticles, such as multi-walled carbon nanotubes, acutely upregulate a diverse profile of matrix proteases, and induce a complex peptidomic response across lung and blood compartments. The serum peptide fraction, having cell-surface receptor ligand properties, conveys peripheral bioactivity in promoting endothelial cell inflammation, vasodilatory dysfunction and inhibiting angiogenesis. Results here establish peptide fragments as indirect, non-cytokine mediators and putative biomarkers of systemic health outcomes from nanoparticle exposure.
Collapse
Affiliation(s)
- Ekaterina Mostovenko
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Tamara Young
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Pretal P Muldoon
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Lindsey Bishop
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Christopher G Canal
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Aleksandar Vucetic
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Patti C Zeidler-Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA.
| |
Collapse
|
37
|
Chatterjee S, Tao JQ, Johncola A, Guo W, Caporale A, Langham MC, Wehrli FW. Acute exposure to e-cigarettes causes inflammation and pulmonary endothelial oxidative stress in nonsmoking, healthy young subjects. Am J Physiol Lung Cell Mol Physiol 2019; 317:L155-L166. [PMID: 31042077 DOI: 10.1152/ajplung.00110.2019] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The effects of e-cigarette (e-cig) aerosol inhalation by nonsmokers have not been examined to date. The present study was designed to evaluate the acute response to aerosol inhalation of non-nicotinized e-cigarettes in terms of oxidative stress and indices of endothelial activation in human pulmonary microvascular endothelial cells (HPMVEC). Ten smoking-naïve healthy subjects (mean age ± SD = 28.7 ± 5.5 yr) were subjected to an e-cig challenge, following which their serum was monitored for markers of inflammation [C-reactive protein (CRP) and soluble intercellular adhesion molecule (sICAM)] and nitric oxide metabolites (NOx). The oxidative stress and inflammation burden of the circulating serum on the vascular network was also assessed by measuring reactive oxygen species (ROS) production and induction of ICAM-1 expression on HPMVEC. Our results show that serum indices of oxidative stress and inflammation increased significantly (P < 0.05 as compared with baseline), reaching a peak at approximately 1-2 h post-e-cig aerosol inhalation and returning to baseline levels at 6 h. The circulatory burden of the serum (ICAM-1 and ROS) increased significantly at 2 h and returned to baseline values 6 h post-e-cig challenge. ROS production by HPMVEC was found to occur via activation of the NADPH oxidase 2 (NOX2) pathways. These findings suggest that even in the absence of nicotine, acute e-cig aerosol inhalation leads to a transient increase in oxidative stress and inflammation. This can adversely affect the vascular endothelial network by promoting oxidative stress and immune cell adhesion. Thus e-cig inhalation has the potential to drive the onset of vascular pathologies.
Collapse
Affiliation(s)
- Shampa Chatterjee
- Institute for Environmental Medicine and Department of Physiology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Jian-Qin Tao
- Institute for Environmental Medicine and Department of Physiology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Alyssa Johncola
- Laboratory for Structural, Physiologic and Functional Imaging, Department of Radiology, University of Pennsylvania Health System , Philadelphia, Pennsylvania
| | - Wensheng Guo
- Department of Biostatistics and Epidemiology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Alessandra Caporale
- Laboratory for Structural, Physiologic and Functional Imaging, Department of Radiology, University of Pennsylvania Health System , Philadelphia, Pennsylvania
| | - Michael C Langham
- Laboratory for Structural, Physiologic and Functional Imaging, Department of Radiology, University of Pennsylvania Health System , Philadelphia, Pennsylvania
| | - Felix W Wehrli
- Laboratory for Structural, Physiologic and Functional Imaging, Department of Radiology, University of Pennsylvania Health System , Philadelphia, Pennsylvania
| |
Collapse
|
38
|
Zhao D, Yin CY, Ye XW, Wan ZF, Zhao DG, Zhang XY. Mitochondrial separation protein inhibitor inhibits cell apoptosis in rat lungs during intermittent hypoxia. Exp Ther Med 2019; 17:2349-2358. [PMID: 30867720 DOI: 10.3892/etm.2019.7201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/22/2018] [Indexed: 12/27/2022] Open
Abstract
Obstructive sleep apnoea (OSA) is a very common sleep and breathing disorder that occurs in worldwide. It is important to develop a more effective treatment for OSA to overcome lung cell apoptosis during intermittent hypoxia (IH). A mitochondrial separation protein inhibitor (Mdivi-1) has been demonstrated to be a powerful tool for inhibiting apoptosis. In the present study, the protective effect and possible mechanism of apoptosis in lung cells during IH was investigated using in vivo and in vitro experiments. Following IH exposure for 4 weeks, the lung tissues of Sprague Dawley rats exhibited interstitial lesions, while Mdivi-1 reduced these pulmonary interstitial lesions. B-cell lymphoma (Bcl)-2 mRNA and protein expression levels were decreased however caspase-3, caspase-9 and dynamin-related protein 1 (Drp-1) mRNA and protein expression levels were increased. Following Mdivi-1 intervention, Bcl-2 mRNA and protein expression levels were increased while caspase-3, caspase-9 and Drp-1 mRNA and protein expression levels were decreased (P<0.05). After exposure to IH for 12 h, the apoptosis rate of WTRL1 cells in rats increased gradually with the IH time (P<0.05). Bcl-2 mRNA and protein expression levels were decreased, whereas caspase-3, caspase-9, cytochrome C (Cyt-C) and Drp-1 mRNA levels were increased, and caspase-3, caspase-9 and Drp-1 protein expression levels were increased. After Mdivi-1 intervention, Bcl-2 mRNA and protein expression levels were increased but caspase-3, caspase-9, Cyt-C and Drp-1 mRNA levels were decreased along with caspase-9, Cyt-C and Drp-1 protein expression levels which were decreased (P<0.05). The results of the present study suggest that Mdivi-1 may be a potential agent for treating OSA because it inhibits the mitochondrial pathway and reduces apoptosis.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China.,Life Sciences College of Guizhou University, Guiyang, Guizhou 550025, P.R. China
| | - Chen-Yi Yin
- Department of Graduate School, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xian-Wei Ye
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Zi-Fen Wan
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - De-Gang Zhao
- Life Sciences College of Guizhou University, Guiyang, Guizhou 550025, P.R. China.,The Key Laboratory of Plant Resources Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), Guizhou University, Guiyang, Guizhou 550025, P.R. China
| | - Xiang-Yan Zhang
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
39
|
Obstructive Sleep Apnea and Inflammation: Proof of Concept Based on Two Illustrative Cytokines. Int J Mol Sci 2019; 20:ijms20030459. [PMID: 30678164 PMCID: PMC6387387 DOI: 10.3390/ijms20030459] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/12/2019] [Accepted: 01/15/2019] [Indexed: 12/19/2022] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a markedly prevalent condition across the lifespan, particularly in overweight and obese individuals, which has been associated with an independent risk for neurocognitive, behavioral, and mood problems as well as cardiovascular and metabolic morbidities, ultimately fostering increases in overall mortality rates. In adult patients, excessive daytime sleepiness (EDS) is the most frequent symptom leading to clinical referral for evaluation and treatment, but classic EDS features are less likely to be reported in children, particularly among those with normal body-mass index. The cumulative evidence collected over the last two decades supports a conceptual framework, whereby sleep-disordered breathing in general and more particularly OSAS should be viewed as low-grade chronic inflammatory diseases. Accordingly, it is assumed that a proportion of the morbid phenotypic signature in OSAS is causally explained by underlying inflammatory processes inducing end-organ dysfunction. Here, the published links between OSAS and systemic inflammation will be critically reviewed, with special focus on the pro-inflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6), since these constitute classical prototypes of the large spectrum of inflammatory molecules that have been explored in OSAS patients.
Collapse
|
40
|
Abstract
Obstructive sleep apnea (OSA) is common among patients with cardiac rhythm disorders. OSA may contribute to arrhythmias due to acute mechanisms, such as generation of negative intrathoracic pressure during futile efforts to breath, intermittent hypoxia, and surges in sympathetic activity. In addition, OSA may lead to heart remodeling and increases arrhythmia susceptibility. Atrial distension and remodeling, that has been shown to be associated with OSA, is a well-known anatomical substrate for atrial fibrillation (AF). AF is the arrhythmia most commonly described in patients with OSA. Several observational studies have shown that the treatment of OSA with continuous positive airway pressure (CPAP) reduces recurrence of AF after electrical cardioversion and catheter ablation. There is also evidence that nocturnal hypoxemia, a hallmark of OSA, predicts sudden cardiac death (SCD) independently of well-established cardiovascular risk factors. Among patients with an implantable cardiac defibrillator, those with OSA have a higher risk of receiving treatment for life-threatening arrhythmias. Nocturnal hypoxemia may also increase vagal tone, which increases susceptibility to bradycardic and conduction rhythm disorders that have also been described in patients with OSA. In conclusion, there are several biological pathways linking OSA and increased cardiac arrhythmogenesis propensity. However, the independent association is derived from observational studies and the direction of the association still needs clarification due to the lack of large clinical trials. This review focuses on the current scientific evidence linking OSA to cardiac rhythm disorders and point out future directions.
Collapse
Affiliation(s)
- Glaucylara Reis Geovanini
- Sleep Laboratory, Pulmonary Division, Heart Institute (InCor), Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Genetics and Molecular Cardiology Laboratory, Heart Institute (InCor), Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Geraldo Lorenzi-Filho
- Sleep Laboratory, Pulmonary Division, Heart Institute (InCor), Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Arslanian S, Bacha F, Grey M, Marcus MD, White NH, Zeitler P. Evaluation and Management of Youth-Onset Type 2 Diabetes: A Position Statement by the American Diabetes Association. Diabetes Care 2018; 41:2648-2668. [PMID: 30425094 PMCID: PMC7732108 DOI: 10.2337/dci18-0052] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Silva Arslanian
- Division of Pediatric Endocrinology, Metabolism, and Diabetes Mellitus, University of Pittsburgh, Pittsburgh, PA
- Center for Pediatric Research in Obesity and Metabolism, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Fida Bacha
- Children's Nutrition Research Center, Texas Children's Hospital and Baylor College of Medicine, Houston, TX
| | - Margaret Grey
- Yale School of Nursing, New Haven, CT
- Yale School of Medicine, New Haven, CT
| | | | - Neil H White
- Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Philip Zeitler
- Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
42
|
Pan H, Zhang M. Serum of coronary atherosclerotic heart disease patients induces oxidative stress injury on endothelial cells. Pteridines 2018. [DOI: 10.1515/pteridines-2018-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Endothelial cell (EC) dysfunction has a fundamental role in the development of atherosclerosis, which leads to myocardial infarction and stroke. The aim of this study is to investigate the effect of serum from patients with coronary atherosclerotic heart disease (CAD) on endothelial cells and investigate the possible mechanism underlying these effects. Serum from 35 patients with CAD and 35 healthy volunteers was collected. Human umbilical vein endothelial cell (HUVEC) proliferation and apoptosis were assessed by a CCK‑8 assay and a flow cytometry assay, respectively. The synthesis of nitric oxide (NO) and reactive oxygen species (ROS) was measured using the nitrate reduction method and DCFH2-DA staining, respectively. The proliferation of HUVECs was inhibited by treatment with serum from CAD patients (P<0.05). Suppression of HUVEC proliferation by CAD serum occurred in a concentration-dependent manner. The synthesis of NO was also reduced in the CAD serum-treated group. Furthermore, the serum from CAD patients increased both apoptosis and intracellular ROS production in HUVECs. Moreover, treatment with tempol antagonized CAD serum-meditated HUVEC injuries. Taken together, these results suggest that HUVEC injury via CAD serum treatment is mediated by ROS production. Tempol may partly reverse this effect by abolishing HUVEC apoptosis.
Collapse
Affiliation(s)
- Huichao Pan
- Division Of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School Of Medicine, Shanghai , China
| | - Min Zhang
- Division of Cardiology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
43
|
Gabryelska A, Łukasik ZM, Makowska JS, Białasiewicz P. Obstructive Sleep Apnea: From Intermittent Hypoxia to Cardiovascular Complications via Blood Platelets. Front Neurol 2018; 9:635. [PMID: 30123179 PMCID: PMC6085466 DOI: 10.3389/fneur.2018.00635] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/16/2018] [Indexed: 12/31/2022] Open
Abstract
Obstructive sleep apnea is a chronic condition characterized by recurrent episodes of apneas or hypopneas during sleep leading to intermittent hypoxemia and arousals. The prevalence of the sleep disordered breathing is estimated that almost 50% of men and 24% of women suffer from moderate to severe form of the disorder. Snoring, collapse of upper airways and intermittent hypoxia are main causes of smoldering systemic inflammation in patients suffering from obstructive sleep apnea. The systematic inflammation is considered one of the key mechanisms leading to significant cardiovascular complications. Blood platelets, formerly not even recognized as cells, are currently gaining attention as crucial players in the immune continuum. Platelet surface is endowed with receptors characteristic for cells classically belonging to the immune system, which enables them to recognize pathogens, immune complexes, and interact in a homo- and heterotypic aggregates. Platelets participate in the process of transcellular production of bioactive lipids by delivering both specific enzymes and substrate molecules. Despite their lack of nucleus, platelets synthetize proteins in a stimuli-dependent manner. Atherosclerosis and consequent cardiovascular complications result from disruption in homeostasis of both of the platelet roles: blood coagulation and inflammatory processes modulation. Platelet parameters, routinely evaluated as a part of complete blood count test, were proposed as markers of cardiovascular comorbidity in patients with obstructive sleep apnea. Platelets were found to be excessively activated in this group of patients, especially in obese subjects. Persistent activation results in enhanced spontaneous aggregability and change in cytokine production. Platelet-lymphocyte ratio was suggested as an independent marker for cardiovascular disease in obstructive sleep apnea syndrome and continuous positive air pressure therapy was found to have an impact on platelet parameters and phenotype. In this literature review we summarize the current knowledge on the subject of platelets involvement in obstructive sleep apnea syndrome and consider the possible pathways in which they contribute to cardiovascular comorbidity.
Collapse
Affiliation(s)
- Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| | - Zuzanna M Łukasik
- Department of Rheumatology, Medical University of Lodz, Lodz, Poland
| | - Joanna S Makowska
- Department of Rheumatology, Medical University of Lodz, Lodz, Poland
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
44
|
Castaneda A, Jauregui-Maldonado E, Ratnani I, Varon J, Surani S. Correlation between metabolic syndrome and sleep apnea. World J Diabetes 2018; 9:66-71. [PMID: 29765510 PMCID: PMC5951892 DOI: 10.4239/wjd.v9.i4.66] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/06/2018] [Accepted: 05/10/2018] [Indexed: 02/05/2023] Open
Abstract
The so-called “metabolic syndrome” (MS), constitutes a cluster of metabolic and cardiovascular abnormalities, including fasting glucose, blood pressure, triglycerides, high density lipoprotein cholesterol (HDL-C), and waist circumference that arise from insulin resistance. Obstructive sleep apnea (OSA) syndrome is characterized by recurrent episodes of partial or complete obstruction of the upper airway, involving cessation or significant decreased airflow, with intermittent hypoxemia, frequent arousals from sleep and recurrent oxyhemoglobin desaturations that interfere with normal sleep patterns generating difficulty falling asleep, unrefreshing sleep and loud snoring. The relation between these two entities is known as “Syndrome Z”, and there is no question about the impact of these risk factors on health and disease. This clinical condition presents a growing epidemic Worldwide, affecting approximately 60% of the general population with both MS and OSA due to the constant increase of body mass index in humans. This article presents evidence-based data that focuses on the direct relationship between MS and OSA.
Collapse
Affiliation(s)
| | | | - Iqbal Ratnani
- DeBakey Heart and Vascular Center, Houston Methodist, Houston, TX 77030, United States
| | - Joseph Varon
- Department of Acute and Continuing Care. The University of Texas Health Science Center Houston, TX 77030, United States
| | - Salim Surani
- Division of Pulmonary, Critical Care and Sleep Medicine, Texas A and M University, Health Science Center, Corpus Christi, TX 78404, United States
| |
Collapse
|
45
|
Kim HJ, Noh JS, Song YO. Beneficial Effects of Kimchi, a Korean Fermented Vegetable Food, on Pathophysiological Factors Related to Atherosclerosis. J Med Food 2017; 21:127-135. [PMID: 29271694 DOI: 10.1089/jmf.2017.3946] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Atherosclerosis is a progressive disease that is characterized by accumulation of lipids and fibrous elements in large arteries. Its etiology is involved with pathophysiological factors such as lipoprotein oxidation, inflammation, and dyslipidemia. Kimchi is a Korean fermented vegetable side dish made with vegetables and kimchi condiments. To date, numerous in vitro, in vivo, and human studies have cited the health benefits of kimchi. 3-(4'-Hydroxyl-3',5'-dimethoxyphenyl)propionic acid is one of the active compounds of kimchi, and its antioxidant and anti-atherosclerosclerotic effects have been reported. This review presents the laboratory and clinical evidence of the anti-atherosclerotic effects of kimchi based on its lipid-lowering, antioxidant, and anti-inflammatory activities.
Collapse
Affiliation(s)
- Hyun Ju Kim
- 1 Industrial Technology Research Group, Research and Development Division, World Institute of Kimchi , Gwangju, Korea
| | - Jeong Sook Noh
- 2 Department of Food Science and Nutrition, Tongmyong University , Busan, Korea
| | - Yeong Ok Song
- 3 Department of Food Science and Nutrition, Kimchi Research Institute, Pusan National University , Busan, Korea
| |
Collapse
|
46
|
Riad E, Nasser M, Hussein MH, Toraih EA, Fawzy MS. Serum S100B: A possible biomarker for severity of obstructive sleep apnea. EGYPTIAN JOURNAL OF CHEST DISEASES AND TUBERCULOSIS 2017. [DOI: 10.1016/j.ejcdt.2017.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
47
|
Koehler U, Hildebrandt O, Krönig J, Grimm W, Otto J, Hildebrandt W, Kinscherf R. [Chronic hypoxia and cardiovascular risk : Clinical significance of different forms of hypoxia]. Herz 2017; 43:291-297. [PMID: 28474128 DOI: 10.1007/s00059-017-4570-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/13/2017] [Accepted: 04/04/2017] [Indexed: 12/19/2022]
Abstract
It is of fundamental importance to differentiate whether chronic hypoxia occurs intermittently or persistently. While chronic intermittent hypoxia (CIH) is found typically in patients with obstructive sleep apnea (OAS), chronic persistent hypoxia (CPH) is typically diagnosed in patients with chronic lung disease. Cardiovascular risk is markedly increased in patients with CIH compared to patients with CPH. The frequent change between oxygen desaturation and reoxygenation in patients with CIH is associated with increased hypoxic stress, increased systemic inflammation, and enhanced adrenergic activation followed by endothelial dysfunction and increased arteriosclerosis. The pathophysiologic consequences of CPH are less well understood. The relationship between CPH and the development of pulmonary hypertension, pulmonary heart disease as well as polycythemia has been established.
Collapse
Affiliation(s)
- U Koehler
- Klinik für Innere Medizin, SP Pneumologie, Intensiv- und Schlafmedizin, Philipps-Universität, Baldingerstraße 1, 35043, Marburg, Deutschland.
| | - O Hildebrandt
- Klinik für Innere Medizin, SP Pneumologie, Intensiv- und Schlafmedizin, Philipps-Universität, Baldingerstraße 1, 35043, Marburg, Deutschland
| | - J Krönig
- Klinik für Innere Medizin, SP Pneumologie, Intensiv- und Schlafmedizin, Philipps-Universität, Baldingerstraße 1, 35043, Marburg, Deutschland
| | - W Grimm
- Klinik für Innere Medizin, SP Kardiologie, Intensivmedizin und Angiologie, Philipps-Universität, Marburg, Deutschland
| | - J Otto
- Medizinische Zellbiologie, Institut für Anatomie und Zellbiologie, Philipps-Universität, Marburg, Deutschland
| | - W Hildebrandt
- Medizinische Zellbiologie, Institut für Anatomie und Zellbiologie, Philipps-Universität, Marburg, Deutschland
| | - R Kinscherf
- Medizinische Zellbiologie, Institut für Anatomie und Zellbiologie, Philipps-Universität, Marburg, Deutschland
| |
Collapse
|
48
|
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a common disease, distinguished by recurrent episodes of upper airway obstruction during sleep, with an inflammatory component. C-reactive protein (CRP) and high-sensitivity C-reactive protein (hs-CRP) are markers of systemic inflammation and may serve as biomarkers of OSA. METHODS Scientific studies published from January 1, 2006, to January 1, 2016 were obtained via searches of PubMed, Embase, SCI, and China National Knowledge Internet (CNKI) using relevant terms. Studies concerning serum CRP level/ hs-CRP in OSA patients were reviewed by 2 independent reviewers. Studies were included if they conform with our specific criteria of inclusion. Eligible studies were subjected to quality review, data extraction, and meta-analysis by using RevMan (version 5.2) and STATA (version 12.0). RESULTS There were 15 studies that met inclusion criteria that included a total of 1297 subjects. Meta-analysis revealed that serum CRP levels in the OSA group were 1.98 mmol/L higher than those in control group (95% confidence interval: 1.39-2.58, P < .01). Similarly, serum hs-CRP levels in the OSA group were 1.57 mmol/L higher than that in the control group (95% confidence interval: 0.96-2.18, P < .01). Subgroup analysis showed greater differences between OSA patients and controls in the setting of obesity (body mass index)> = 30. The total weighted mean difference (WMD) between OSA and controls within the subgroup of subjects who had a CRP was 2.10; for hs-CRP, the WMD was 2.49. Comparing OSA patients of mean apnea hypopnea index> = 15 and controls, the total WMD for the CRP subgroup was 2.19; for the hs-CRP subgroup, the WMD was 1.70. CONCLUSION In our meta-analysis, serum CRP/hs-CRP levels were discovered to be higher in OSA patients compared with control subjects. Those with higher body mass index and apnea hyponea index demonstrated larger differences in CRP/hs-CRP levels. These data are consistent with an inflammatory component of OSA pathophysiology and support the role of CRP/hs-CRP as a biomarker in this disease.
Collapse
|
49
|
Hoyos CM, Drager LF, Patel SR. OSA and cardiometabolic risk: What's the bottom line? Respirology 2017; 22:420-429. [DOI: 10.1111/resp.12984] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Camilla M. Hoyos
- Centre for Sleep and Chronobiology, Woolcock Institute of Medical Research; University of Sydney; Sydney New South Wales Australia
- Healthy Brain Ageing Program, School of Psychology, Faculty of Science; University of Sydney; Sydney New South Wales Australia
| | - Luciano F. Drager
- Hypertension Unit - Heart Institute (InCor); University of Sao Paulo Medical School; Sao Paulo Brazil
| | - Sanjay R. Patel
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine; University of Pittsburgh; Pittsburgh Pennsylvania USA
| |
Collapse
|