1
|
Yazaki K, Dewar M, Dauz J, Akazawa Y, Hui L, Sun M, Hui W, Kabir G, Dejardin JF, Connelly KA, Heximer SP, Friedberg MK. Serial and regional assessment of the right ventricular molecular and functional response to pressure loading. Am J Physiol Heart Circ Physiol 2025; 328:H58-H74. [PMID: 39422363 DOI: 10.1152/ajpheart.00322.2024] [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: 05/21/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
Right ventricular (RV) function determines outcomes in RV pressure loading. A better understanding of the time-course and regional distribution of RV remodeling may help optimize targets and timing for therapeutic intervention. We sought to characterize RV remodeling between zero and 6 wk after the initiation of RV pressure loading. Thirty-six rats were randomized to either sham surgery or to pulmonary artery banding (PAB). After echocardiography and conductance catheter studies, groups of rats were euthanized at 1 wk, 3 wk, and 6 wk after sham surgery, or induction of RV pressure loading, for RV histological, RNA, and molecular analysis. A vigorous inflammatory response characterized by increased RV inflammatory cytokines, chemokines, and macrophage markers was observed at 1 wk following PAB. Metabolic changes, transforming growth factor-β (TGF-β)1 canonical signaling, collagenous fibrosis deposition, and apoptosis were already significantly increased by 1 wk after PAB. Genes marking fibroblast activation were upregulated at 1 wk but not at 6 wk post-PAB surgery. Mitochondrial dysfunction was evidenced by increased pyruvate dehydrogenase kinase (PDK) activity and decreased pyruvate dehydrogenase (PDH) phosphorylation significantly at 6-wk post-PAB. These processes preceded the development of overt myocardial hypertrophy and impaired echo parameters of systolic and diastolic function that occurred significantly from 3 wk after PAB. RV myocardial inflammation, metabolic shift, metabolic gene transcription, and profibrotic signaling occur early after initiation of pressure loading when RV pressures are only moderately elevated, before the development of overt myocardial hypertrophy and dysfunction, suggesting that adaptive hypertrophy and maladaptive remodeling occur simultaneously. These results suggest that therapeutic intervention to reduce adverse RV remodeling may be needed earlier and at lower thresholds than currently used.NEW & NOTEWORTHY Exploring the dynamics of right ventricular remodeling: unveiling the intricate interplay between inflammation, metabolic shifts, and fibrotic signaling in response to pressure loading. Through a comprehensive study spanning from initiation to 6 wk post-pressure loading, our research sheds light on the early onset of crucial molecular processes preceding overt hypertrophy and dysfunction. These findings challenge conventional intervention timing, advocating for early, targeted therapeutic strategies to mitigate adverse remodeling in right ventricular pressure loading.
Collapse
MESH Headings
- Animals
- Ventricular Remodeling
- Male
- Ventricular Function, Right
- Rats, Sprague-Dawley
- Fibrosis
- Rats
- Ventricular Pressure
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Disease Models, Animal
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/genetics
- Ventricular Dysfunction, Right/etiology
- Time Factors
- Apoptosis
- Pulmonary Artery/physiopathology
- Pulmonary Artery/metabolism
- Signal Transduction
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/genetics
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/pathology
- Cytokines/metabolism
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
Collapse
Affiliation(s)
- Kana Yazaki
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Michael Dewar
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - John Dauz
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Yohei Akazawa
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Lucy Hui
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Mei Sun
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Wei Hui
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Golam Kabir
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jean-Francois Dejardin
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Kim A Connelly
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Scott P Heximer
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mark K Friedberg
- Division of Cardiology, Labatt Family Heart Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Vlachakis PK, Theofilis P, Kachrimanidis I, Giannakopoulos K, Drakopoulou M, Apostolos A, Kordalis A, Leontsinis I, Tsioufis K, Tousoulis D. The Role of Inflammasomes in Heart Failure. Int J Mol Sci 2024; 25:5372. [PMID: 38791409 PMCID: PMC11121241 DOI: 10.3390/ijms25105372] [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: 04/24/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Heart failure (HF) poses a significant world health challenge due to the increase in the aging population and advancements in cardiac care. In the pathophysiology of HF, the inflammasome has been correlated with the development, progression, and complications of HF disease. Discovering biomarkers linked to inflammasomes enhances understanding of HF diagnosis and prognosis. Directing inflammasome signaling emerges as an innovative therapeutic strategy for managing HF. The present review aims to delve into this inflammatory cascade, understanding its role in the development of HF, its potential role as biomarker, as well as the prospects of modulating inflammasomes as a therapeutic approach for HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Dimitris Tousoulis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.K.V.); (P.T.); (I.K.); (K.G.); (M.D.); (A.A.); (A.K.); (I.L.); (K.T.)
| |
Collapse
|
3
|
Ramírez-Vélez R, González A, García-Hermoso A, Amézqueta IL, Izquierdo M, Díez J. Revisiting skeletal myopathy and exercise training in heart failure: Emerging role of myokines. Metabolism 2023; 138:155348. [PMID: 36410495 DOI: 10.1016/j.metabol.2022.155348] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/06/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Abstract
Exercise intolerance remains a major unmet medical need in patients with heart failure (HF). Skeletal myopathy is currently considered as the major limiting factor for exercise capacity in HF patients. On the other hand, emerging evidence suggest that physical exercise can decrease morbidity and mortality in HF patients. Therefore, mechanistic insights into skeletal myopathy may uncover critical aspects for therapeutic interventions to improve exercise performance in HF. Emerging data reviewed in this article suggest that the assessment of circulating myokines (molecules synthesized and secreted by skeletal muscle in response to contraction that display autocrine, paracrine and endocrine actions) may provide new insights into the pathophysiology, phenotyping and prognostic stratification of HF-related skeletal myopathy. Further studies are required to determine whether myokines may also serve as biomarkers to personalize the modality and dose of physical training prescribed for patients with HF and exercise intolerance. In addition, the production and secretion of myokines in patients with HF may interact with systemic alterations (e.g., inflammation and metabolic disturbances), frequently present in patients with HF. Furthermore, myokines may exert beneficial or detrimental effects on cardiac structure and function, which may influence adverse cardiac remodelling and clinical outcomes in HF patients. Collectively, these data suggest that a deeper knowledge on myokines regulation and actions may lead to the identification of novel physical exercise-based therapeutic approaches for HF patients.
Collapse
Affiliation(s)
- Robinson Ramírez-Vélez
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Spain; CIBERFES, Carlos III Institute of Health, Madrid, Spain; Institute for Health Research of Navarra (IDISNA), Pamplona, Spain
| | - Arantxa González
- Institute for Health Research of Navarra (IDISNA), Pamplona, Spain; Program of Cardiovascular Diseases, Center of Applied Medical Research (CIMA), Universidad deNavarra, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Antonio García-Hermoso
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Spain; CIBERFES, Carlos III Institute of Health, Madrid, Spain; Institute for Health Research of Navarra (IDISNA), Pamplona, Spain
| | - Iñigo Latasa Amézqueta
- Program of Cardiovascular Diseases, Center of Applied Medical Research (CIMA), Universidad deNavarra, Pamplona, Spain
| | - Mikel Izquierdo
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Spain; CIBERFES, Carlos III Institute of Health, Madrid, Spain; Institute for Health Research of Navarra (IDISNA), Pamplona, Spain.
| | - Javier Díez
- Institute for Health Research of Navarra (IDISNA), Pamplona, Spain; Program of Cardiovascular Diseases, Center of Applied Medical Research (CIMA), Universidad deNavarra, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| |
Collapse
|
4
|
Gilbert-Ouimet M, Zahiriharsini A, Biron C, Langlois L, Ménard C, Lebel M, Pelletier J, Duchaine C, Beaulieu M, Truchon M. Predict, prevent and manage moral injuries in Canadian frontline healthcare workers and leaders facing the COVID-19 pandemic: Protocol of a mixed methods study. SSM - MENTAL HEALTH 2022; 2:100124. [PMID: 35669531 PMCID: PMC9158246 DOI: 10.1016/j.ssmmh.2022.100124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 05/24/2022] [Accepted: 05/28/2022] [Indexed: 11/20/2022] Open
Abstract
Moral injuries can occur when perpetrating, failing to prevent, or bearing witness to acts that transgress deeply held moral beliefs and expectations. The COVID-19 crisis highlighted the fact that psychosocial stressors at work, such as high emotional demands, are placing Canadian healthcare workers at risk of moral injuries. Evidence linking psychosocial stressors at work to moral injuries are needed to better predict, prevent and manage moral injuries, as these stressors are frequent and modifiable occupational risk factors. This protocol presents a study aiming to: 1) understand workplace events having the potential to either cause or reduce moral injuries, 2) predict the risk and severity of moral injuries using a disease prevention model, 3) identify biological signatures (biomarkers) associated with psychosocial stressors at work and moral injuries and 4) elaborate preliminary guidelines of organizational practices for frontline healthcare workers to reduce and manage moral injuries. This study is a mixed methods research with three components: qualitative, quantitative and biological. The data collection has been completed and because of the COVID-19 pandemic, it was adjusted to allow for gathering qualitative and quantitative data remotely. Frontline healthcare workers and leaders were included. Through focus groups and individual interviews, and an online questionnaire, events and psychosocial working conditions that may increase the risk of moral injuries will be documented. In addition, blood samples which were collected from a sub-sample of volunteer participants will measure an innovative set of biomarkers associated with vulnerability to stress and mental health. Data analyses are ongoing. We anticipate to identify workplace events that may trigger moral injuries. We expect that potential predictors of moral injury risk occurrence and severity will be identified from psychosocial stressors at work that can be improved by implementing organizational practices. We also expect to observe a different mental health state and biological inflammation signature across workers exposed compared to workers not exposed to psychosocial stressors at work. Based on these future findings, we intend to develop preliminary recommendations of organizational practices for managers. This research will contribute to expand our knowledge of the events in the workplace likely to generate or lessen the impact moral injuries, to build a model for predicting the risk of moral injuries at work, all in the specific context of the COVID-19 health crisis among healthcare workers.
Collapse
Affiliation(s)
- Mahée Gilbert-Ouimet
- Department of Health Sciences, Université du Québec à Rimouski, 1595 Boulevard Alphonse-Desjardins, Lévis, QC, G6V 0A6, Canada
- CHU de Québec-Laval University Research Center, 1050 Chemin Ste-Foy, Quebec City, QC, G1S 4L8, Canada
| | - Azita Zahiriharsini
- Department of Health Sciences, Université du Québec à Rimouski, 1595 Boulevard Alphonse-Desjardins, Lévis, QC, G6V 0A6, Canada
- CHU de Québec-Laval University Research Center, 1050 Chemin Ste-Foy, Quebec City, QC, G1S 4L8, Canada
| | - Caroline Biron
- Department of Management, Laval University, Quebec City, QC, G1V 0A6, Canada
| | | | - Caroline Ménard
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University and CERVO Brain Research Center, Quebec City, QC, G1V 0A6, Canada
| | - Manon Lebel
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University and CERVO Brain Research Center, Quebec City, QC, G1V 0A6, Canada
| | - Jérôme Pelletier
- Department of Health Sciences, Université du Québec à Rimouski, 1595 Boulevard Alphonse-Desjardins, Lévis, QC, G6V 0A6, Canada
| | - Caroline Duchaine
- Faculty of Medicine, Laval University, 1050 ave de la Médecine, Quebec City, QC, G1V 0A6, Canada
- CHU de Québec-Laval University Research Center, 1050 Chemin Ste-Foy, Quebec City, QC, G1S 4L8, Canada
| | | | - Manon Truchon
- School of Psychology, Laval University, Quebec City, QC, G1V 0A6, Canada
- Centre de recherche interdisciplinaire en réadaptation et intégration sociale(CIRRIS), Quebec City, QC, G1M 2S8, Canada
| |
Collapse
|
5
|
Takasu T. The Role of SGLT2 Inhibitor Ipragliflozin on Cardiac Hypertrophy and microRNA Expression Profiles in a Non-diabetic Rat Model of Cardiomyopathy. Biol Pharm Bull 2022; 45:1321-1331. [DOI: 10.1248/bpb.b22-00289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
6
|
Koleck TA, Topaz M, Tatonetti NP, George M, Miaskowski C, Smaldone A, Bakken S. Characterizing shared and distinct symptom clusters in common chronic conditions through natural language processing of nursing notes. Res Nurs Health 2021; 44:906-919. [PMID: 34637147 PMCID: PMC8641786 DOI: 10.1002/nur.22190] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 01/08/2023]
Abstract
Data-driven characterization of symptom clusters in chronic conditions is essential for shared cluster detection and physiological mechanism discovery. This study aims to computationally describe symptom documentation from electronic nursing notes and compare symptom clusters among patients diagnosed with four chronic conditions-chronic obstructive pulmonary disease (COPD), heart failure, type 2 diabetes mellitus, and cancer. Nursing notes (N = 504,395; 133,977 patients) were obtained for the 2016 calendar year from a single medical center. We used NimbleMiner, a natural language processing application, to identify the presence of 56 symptoms. We calculated symptom documentation prevalence by note and patient for the corpus. Then, we visually compared documentation for a subset of patients (N = 22,657) diagnosed with COPD (n = 3339), heart failure (n = 6587), diabetes (n = 12,139), and cancer (n = 7269) and conducted multiple correspondence analysis and hierarchical clustering to discover underlying groups of patients who have similar symptom profiles (i.e., symptom clusters) for each condition. As expected, pain was the most frequently documented symptom. All conditions had a group of patients characterized by no symptoms. Shared clusters included cardiovascular symptoms for heart failure and diabetes; pain and other symptoms for COPD, diabetes, and cancer; and a newly-identified cognitive and neurological symptom cluster for heart failure, diabetes, and cancer. Cancer (gastrointestinal symptoms and fatigue) and COPD (mental health symptoms) each contained a unique cluster. In summary, we report both shared and distinct, as well as established and novel, symptom clusters across chronic conditions. Findings support the use of electronic health record-derived notes and NLP methods to study symptoms and symptom clusters to advance symptom science.
Collapse
Affiliation(s)
- Theresa A. Koleck
- School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Maxim Topaz
- School of Nursing, Columbia University, New York, New York
- Data Science Institute, Columbia University, New York, New York
| | - Nicholas P. Tatonetti
- Data Science Institute, Columbia University, New York, New York
- Department of Biomedical Informatics, Columbia University, New York, New York
- Department of Systems Biology, Columbia University, New York, New York
- Department of Medicine, Columbia University, New York, New York
- Institute for Genomic Medicine, Columbia University, New York, New York
| | - Maureen George
- School of Nursing, Columbia University, New York, New York
| | - Christine Miaskowski
- School of Nursing, University of California San Francisco, San Francisco, California
| | - Arlene Smaldone
- School of Nursing, Columbia University, New York, New York
- College of Dental Medicine, Columbia University, New York, New York
| | - Suzanne Bakken
- School of Nursing, Columbia University, New York, New York
- Data Science Institute, Columbia University, New York, New York
- Department of Biomedical Informatics, Columbia University, New York, New York
| |
Collapse
|
7
|
Ren Y, Wang X, Liang H, He W, Zhao X. Mechanism of miR-30b-5p-Loaded PEG-PLGA Nanoparticles for Targeted Treatment of Heart Failure. Front Pharmacol 2021; 12:745429. [PMID: 34658880 PMCID: PMC8514665 DOI: 10.3389/fphar.2021.745429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/10/2021] [Indexed: 12/02/2022] Open
Abstract
Objective: Exploring the effectiveness of miR-30b-5p-loaded PEG-PLGA nanoparticles (NPs) for the treatment of heart failure and the underlying mechanism. Methods: PEG-PLGA characteristics with different loading amounts were first examined to determine the loading, encapsulation, and release of miR-30b-5p from NPs. The effects of miR-30b-5p NPs on cardiac function and structure were assessed by immunofluorescence, echocardiography, HE/Masson staining, and TUNEL staining. The effects of NPs on the expression of factors related to cardiac hypertrophy and inflammation were examined by RT-PCR and western blotting, and the mechanism of miR-30b-5p treatment on heart failure was explored by dual luciferase reporter assay and RT-PCR. Results: The size of PEG-PLGA NPs with different loading amounts ranged from 200 to 300 nm, and the zeta potential of PEG-PLGA NPs was negative. The mean entrapment efficiency of the NPs for miR-30b-5p was high (81.8 ± 2.1%), and the release rate reached 5 days with more than 90% release. Distribution experiments showed that NPs were mainly distributed in the heart and had a protective effect on myocardial injury and cardiac function. Compared with a rat model of cardiac failure and miR-30b-5p-non-loaede NP groups, the expression of cardiac hypertrophy markers (ANP, BNPβ-MHC) and inflammatory factors (IL-1β, IL-6) were significantly decreased. Dual luciferase reporter assay assays indicated that miR-30b-5p exerted its effects mainly by targeting TGFBR2. Conclusion: PEG-PLGA NPs loaded with miR-30b-5p improved cardiac function, attenuated myocardial injury, and regulated the expression of factors associated with cardiac hypertrophy and inflammation by targeting TGFBR2.
Collapse
Affiliation(s)
- Yu Ren
- Scientific Research Department, Inner Mongolia People's Hospital, Hohhot, China
| | - Xiao Wang
- Scientific Research Department, Inner Mongolia People's Hospital, Hohhot, China
| | - Hongyu Liang
- Scientific Research Department, Inner Mongolia People's Hospital, Hohhot, China
| | - Wenshuai He
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, China
| | - Xingsheng Zhao
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, China
| |
Collapse
|
8
|
Patoulias D, Stavropoulos K, Imprialos K, Athyros V, Grassos H, Doumas M, Faselis C. Inflammatory Markers in Cardiovascular Disease; Lessons Learned and Future Perspectives. Curr Vasc Pharmacol 2021; 19:323-342. [PMID: 32188386 DOI: 10.2174/1570161118666200318104434] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiovascular disease (CVD) still remains the leading cause of morbidity and mortality worldwide. It is now established that inflammation plays a crucial role in atherosclerosis and atherothrombosis, and thus, it is closely linked to cardiovascular disease. OBJECTIVE The aim of the present review is to summarize and critically appraise the most relevant evidence regarding the potential use of inflammatory markers in the field of CVD. METHODS We conducted a comprehensive research of the relevant literature, searching MEDLINE from its inception until November 2018, primarily for meta-analyses, randomized controlled trials and observational studies. RESULTS Established markers of inflammation, mainly C-reactive protein, have yielded significant results both for primary and secondary prevention of CVD. Newer markers, such as lipoprotein-associated phospholipase A2, lectin-like oxidized low-density lipoprotein receptor-1, cytokines, myeloperoxidase, cell adhesion molecules, matrix metalloproteinases, and the CD40/CD40 ligand system, have been largely evaluated in human studies, enrolling both individuals from the general population and patients with established CVD. Some markers have yielded conflicting results; however, others are now recognized not only as promising biomarkers of CVD, but also as potential therapeutic targets, establishing the role of anti-inflammatory and pleiotropic drugs in CVD. CONCLUSION There is significant evidence regarding the role of consolidated and novel inflammatory markers in the field of diagnosis and prognosis of CVD. However, multimarker model assessment, validation of cut-off values and cost-effectiveness analyses are required in order for those markers to be integrated into daily clinical practice.
Collapse
Affiliation(s)
- Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | | | - Konstantinos Imprialos
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Vasilios Athyros
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | | | - Michael Doumas
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Charles Faselis
- VA Medical Center, and George Washington University, Washington, DC 20422, United States
| |
Collapse
|
9
|
Rasiuk AS, Walsh SR, Chan L, Viloria-Petit AM, Wootton SK, Karimi K, Bridle BW. The Role of Type I Interferon Signaling in Regulating Cytokine Production and Cell Survival in Bone Marrow-Derived Macrophages. Viral Immunol 2021; 34:470-482. [PMID: 34097550 DOI: 10.1089/vim.2020.0308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
During viral infections, cells produce type I interferons (IFNs), which are detected by the IFNα/β receptor (IFNAR). To survive in hosts, viruses have strategies to downregulate IFN-mediated signaling. We hypothesized that macrophages, which are among the first myeloid cells to respond to viral infections, would produce a different cytokine profile if responding to ligation of pattern recognition receptors (PRRs) while IFNAR-mediated signaling was compromised. Specifically, IFNAR-mediated regulation of interleukin (IL)-1α, IL-6, IL-12, and tumor necrosis factor-α was studied in cultured murine bone marrow-derived macrophages. Since viruses like vesicular stomatitis virus can ligate PRRs such as Toll-like receptor (TLR)4 and 7, macrophages were stimulated with the TLR4 and TLR7 agonists lipopolysaccharide (LPS) or imiquimod, respectively, with or without antibody-mediated IFNAR-blockade. Cytokines and viability were assessed for 3 days poststimulation. Blocking IFNARs acutely exacerbated cytokine production by macrophages and aided their survival when they were treated with LPS. In contrast, cytokine concentrations were unaffected or slightly reduced by IFNAR blockade, but macrophages died at a greater rate when imiquimod was the stimulus. This demonstrated a differential role of IFNAR signaling in regulating PRR-induced cytokines. This suggests potential mechanisms whereby macrophages responding to viruses that inhibit type I IFN responses might contribute to excessive inflammation in some cases and inappropriately low-magnitude responses in others. This also provides a well-defined cell-based model for further dissecting the role of type I IFN signaling in macrophages responding to viral and other infections.
Collapse
Affiliation(s)
| | - Scott R Walsh
- Department of Pathobiology and University of Guelph, Guelph, Canada
| | - Lily Chan
- Department of Pathobiology and University of Guelph, Guelph, Canada
| | | | - Sarah K Wootton
- Department of Pathobiology and University of Guelph, Guelph, Canada
| | - Khalil Karimi
- Department of Pathobiology and University of Guelph, Guelph, Canada
| | - Byram W Bridle
- Department of Pathobiology and University of Guelph, Guelph, Canada
| |
Collapse
|
10
|
Nemec Svete A, Verk B, Čebulj-Kadunc N, Salobir J, Rezar V, Domanjko Petrič A. Inflammation and its association with oxidative stress in dogs with heart failure. BMC Vet Res 2021; 17:176. [PMID: 33902566 PMCID: PMC8077822 DOI: 10.1186/s12917-021-02878-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 04/13/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Inflammation and oxidative stress can contribute to the development and progression of heart failure. This study aimed to investigate the association between inflammatory and oxidative stress markers in dogs with congestive heart failure (CHF). Associations between the disease severity marker N-terminal pro-B-type natriuretic peptide (NT-proBNP) and markers of inflammation and oxidative stress were also determined. RESULTS Thirty-seven dogs with cardiovascular diseases (dilated cardiomyopathy, DCM (16 dogs), myxomatous mitral valve disease, MMVD (21 dogs)) and ten healthy dogs were included in this prospective study. The patients were further divided into groups with (26) and without CHF (11). We found a significantly higher serum concentration of C-reactive protein (P = 0.012), white blood cell (P = 0.001), neutrophil (P = 0.001) and monocyte counts (P = 0.001) in patients with CHF compared to control dogs. The concentration of tumor necrosis factor-alpha (TNF-α) was significantly higher in patients with CHF compared to patients without CHF (P = 0.030). No significant difference was found in most of the measured parameters between MMVD and DCM patients, except for glutathione peroxidase (GPX) and NT-proBNP. In patients with CHF, TNF-α correlated positively with malondialdehyde (P = 0.014, r = 0.474) and negatively with GPX (P = 0.026, r = - 0.453), and interleukin-6 correlated negatively with GPX (P = 0.046, r = - 0.412). NT-proBNP correlated positively with malondialdehyde (P = 0.011, r = 0.493). In patients without CHF none of the inflammatory and oxidative stress markers correlated significantly. Furthermore, in the group of all cardiac patients, GPX activity significantly negatively correlated with NT-proBNP (P = 0.050, r = - 0.339) and several markers of inflammation, including TNF-α (P = 0.010, r = - 0.436), interleukin-6 (P = 0.026, r = - 0.382), white blood cell (P = 0.032, r = - 0.369), neutrophil (P = 0.027, r = - 0.379) and monocyte counts (P = 0.024, r = - 0.386). CONCLUSION Inflammatory and oxidative stress markers are linked in canine CHF patients, but not in patients without CHF. These results suggest complex cross communication between the two biological pathways in advanced stages of CHF.
Collapse
Affiliation(s)
- Alenka Nemec Svete
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Barbara Verk
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Nina Čebulj-Kadunc
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Janez Salobir
- Institute of Nutrition, Biotechnical Faculty, University of Ljubljana, Groblje 3, 1230, Domžale, Slovenia
| | - Vida Rezar
- Institute of Nutrition, Biotechnical Faculty, University of Ljubljana, Groblje 3, 1230, Domžale, Slovenia
| | - Aleksandra Domanjko Petrič
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia.
| |
Collapse
|
11
|
Menzel A, Samouda H, Dohet F, Loap S, Ellulu MS, Bohn T. Common and Novel Markers for Measuring Inflammation and Oxidative Stress Ex Vivo in Research and Clinical Practice-Which to Use Regarding Disease Outcomes? Antioxidants (Basel) 2021; 10:antiox10030414. [PMID: 33803155 PMCID: PMC8001241 DOI: 10.3390/antiox10030414] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Many chronic conditions such as cancer, chronic obstructive pulmonary disease, type-2 diabetes, obesity, peripheral/coronary artery disease and auto-immune diseases are associated with low-grade inflammation. Closely related to inflammation is oxidative stress (OS), which can be either causal or secondary to inflammation. While a low level of OS is physiological, chronically increased OS is deleterious. Therefore, valid biomarkers of these signalling pathways may enable detection and following progression of OS/inflammation as well as to evaluate treatment efficacy. Such biomarkers should be stable and obtainable through non-invasive methods and their determination should be affordable and easy. The most frequently used inflammatory markers include acute-phase proteins, essentially CRP, serum amyloid A, fibrinogen and procalcitonin, and cytokines, predominantly TNFα, interleukins 1β, 6, 8, 10 and 12 and their receptors and IFNγ. Some cytokines appear to be disease-specific. Conversely, OS-being ubiquitous-and its biomarkers appear less disease or tissue-specific. These include lipid peroxidation products, e.g., F2-isoprostanes and malondialdehyde, DNA breakdown products (e.g., 8-OH-dG), protein adducts (e.g., carbonylated proteins), or antioxidant status. More novel markers include also -omics related ones, as well as non-invasive, questionnaire-based measures, such as the dietary inflammatory-index (DII), but their link to biological responses may be variable. Nevertheless, many of these markers have been clearly related to a number of diseases. However, their use in clinical practice is often limited, due to lacking analytical or clinical validation, or technical challenges. In this review, we strive to highlight frequently employed and useful markers of inflammation-related OS, including novel promising markers.
Collapse
Affiliation(s)
- Alain Menzel
- Laboratoires Réunis, 38, Rue Hiehl, L-6131 Junglinster, Luxembourg; (A.M.); (F.D.)
| | - Hanen Samouda
- Nutrition and Health Research Group, Department of Population Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, L-1445 Strassen, Luxembourg;
| | - Francois Dohet
- Laboratoires Réunis, 38, Rue Hiehl, L-6131 Junglinster, Luxembourg; (A.M.); (F.D.)
| | - Suva Loap
- Clinic Cryo Esthetic, 11 Rue Éblé, 75007 Paris, France;
| | - Mohammed S. Ellulu
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Al-Azhar University of Gaza (AUG), Gaza City 00970, Palestine;
| | - Torsten Bohn
- Nutrition and Health Research Group, Department of Population Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, L-1445 Strassen, Luxembourg;
- Correspondence:
| |
Collapse
|
12
|
Perera GS, Ahmed T, Heiss L, Walia S, Bhaskaran M, Sriram S. Rapid and Selective Biomarker Detection with Conductometric Sensors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005582. [PMID: 33502115 DOI: 10.1002/smll.202005582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/18/2020] [Indexed: 06/12/2023]
Abstract
The biomarker detection in human body fluids is crucial as biomarkers are important in diagnosing diseases. Conventional invasive techniques for biomarker detection are associated with infection, tissue damage, and discomfort. Non-invasive devices are an attractive alternative. Here, metal oxide (oxygen-deficient zinc oxide, ZnO) based conductometric sensors with two-terminal electrodes for rapid detection of biomarkers in real-time, are presented. This platform can be engineered for non-invasive, sensitive, and on-demand selective detection of biomarkers based on surface functionalization. The three novelties in this biosensing technique include an on-demand target selection device platform, short (<10 min) incubation times, and real-time monitoring of the biomarker of interest by electrical (resistance change) measurements. Cardiac inflammatory biomarkers interleukin 6 (IL-6) and C-reactive protein (CRP) are used as the model antigens. The devices can detect 100× lower concentration of IL-6 than healthy levels in human saliva and sweat and 1000× and ≈50× lower CRP concentrations than healthy levels in human saliva and sweat, respectively. The devices show high selectivity for IL-6 and CRP antigens when tested with a mixture of biomarkers. This sensor platform can be extended to selective measurements for viruses or DNA screening, which enables a new category of compact and rapid point-of-care medical devices.
Collapse
Affiliation(s)
- Ganganath S Perera
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Taimur Ahmed
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Leah Heiss
- School of Design, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Sumeet Walia
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, Victoria, 3001, Australia
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Madhu Bhaskaran
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Sharath Sriram
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, Victoria, 3001, Australia
| |
Collapse
|
13
|
Che Man R, Sulaiman N, Ishak MF, Bt Hj Idrus R, Abdul Rahman MR, Yazid MD. The Effects of Pro-Inflammatory and Anti-Inflammatory Agents for the Suppression of Intimal Hyperplasia: An Evidence-Based Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17217825. [PMID: 33114632 PMCID: PMC7672569 DOI: 10.3390/ijerph17217825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Anti-atherogenic therapy is crucial in halting the progression of inflammation-induced intimal hyperplasia. The aim of this concise review was to methodically assess the recent findings of the different approaches, mainly on the recruitment of chemokines and/or cytokine and its effects in combating the intimal hyperplasia caused by various risk factors. Pubmed and Scopus databases were searched, followed by article selection based on pre-set inclusion and exclusion criteria. The combination of keywords used were monocyte chemoattractant protein-1 OR MCP-1 OR TNF-alpha OR TNF-α AND hyperplasia OR intimal hyperplasia OR neointimal hyperplasia AND in vitro. These keywords combination was incorporated in the study and had successfully identified 77 articles, with 22 articles were acquired from Pubmed, whereas 55 articles were obtained from Scopus. However, after title screening, only twelve articles meet the requirements of defined inclusion criteria. We classified the data into 4 different approaches, i.e., utilisation of natural product, genetic manipulation and protein inhibition, targeted drugs in clinical setting, and chemokine and cytokines induction. Most of the articles are working on genetic manipulation targeted on specific pathway to inhibit the pro-inflammatory factors expression. We also found that the utilisation of chemokine- and cytokine-related treatments are emerging throughout the years. However, there is no study utilising the combination of approaches that might give a better outcome in combating intimal hyperplasia. Hopefully, this concise review will provide an insight regarding the usage of different novel approaches in halting the progression of intimal hyperplasia, which serves as a key factor for the development of atherosclerosis in cardiovascular disease.
Collapse
Affiliation(s)
- Rohaina Che Man
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Mohamad Fikeri Ishak
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Ruszymah Bt Hj Idrus
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Correspondence: ; Tel.: +603-9145-6995
| |
Collapse
|
14
|
Bouwens E, Schuurman AS, Akkerhuis KM, Baart SJ, Caliskan K, Brugts JJ, van Ramshorst J, Germans T, Umans VAWM, Boersma E, Kardys I. Serially Measured Cytokines and Cytokine Receptors in Relation to Clinical Outcome in Patients With Stable Heart Failure. Can J Cardiol 2020; 36:1587-1591. [PMID: 32827637 DOI: 10.1016/j.cjca.2020.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/04/2020] [Accepted: 08/10/2020] [Indexed: 11/19/2022] Open
Abstract
In this prospective cohort study of 250 stable heart failure patients with trimonthly blood sampling, we investigated associations of 17 repeatedly measured cytokines and cytokine receptors with clinical outcome during a median follow-up of 2.2 (25th-75th percentile, 1.4-2.5) years. Sixty-six patients reached the primary end point (composite of cardiovascular mortality, heart failure hospitalization, heart transplantation, left ventricular assist device implantation). Repeatedly measured levels of 8 biomarkers correlated with clinical outcomes independent of clinical characteristics. Rates of change over time (slopes of biomarker evolutions) remained independently associated with outcome for 15 biomarkers. Thus, temporal patterns of cytokines and cytokine receptors, in particular tumour necrosis factor ligand superfamily member 13B and interleukin-1 receptor type 1, might contribute to personalized risk assessment.
Collapse
Affiliation(s)
- Elke Bouwens
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Sara J Baart
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Kadir Caliskan
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Jasper J Brugts
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Jan van Ramshorst
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | | | - Eric Boersma
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
15
|
Yan Y, Song D, Zhang X, Hui G, Wang J. GEO Data Sets Analysis Identifies COX-2 and Its Related Micro RNAs as Biomarkers for Non-Ischemic Heart Failure. Front Pharmacol 2020; 11:1155. [PMID: 32848764 PMCID: PMC7419645 DOI: 10.3389/fphar.2020.01155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 07/15/2020] [Indexed: 01/06/2023] Open
Abstract
Heart failure (HF) is a heterogeneous clinical syndrome with a variety of causes, risk factors, and pathology. Clinically, only brain natriuretic peptide (BNP) or its precursor N-terminus proBNP (NTproBNP) has been validated for HF diagnosis, but they are also affected by other conditions, such as female gender, renal disease, and acute coronary syndromes, and false low levels in the setting of obesity or flash pulmonary edema. In addition, there is no one biomarker which could encompass all heart failure phenotypes. Advances in bioinformatics have provided us with large databases that characterize the complex genetic and epigenetic changes associated with human diseases. The use of data mining strategies on public access databases to identify previously unknown disease markers is an innovative approach to identify potential biomarkers or even new therapeutic targets in complex diseases such as heart failure (HF). In this study, we analyzed the genomic and transcription data of HF peripheral blood mononuclear cell (PBMC) samples obtained from the Gene Expression Omnibus data sets using Omicsbean online database (http://www.omicsbean.cn/) and found that the prostaglandin-endoperoxide synthase 2 (PTGS2), also named as cyclooxygenase-2 (COX-2), as well as its related micro RNAs including miR-1297 and miR-4649-3p might be used as potential biomarkers for non-ischemic heart failure. Our result showed that plasma COX-2 and miR-4649-3p were significantly up-regulated, whereas the plasma miR-1297 was significantly decreased, and miR-4649-3p displayed high predictive power for non-ischemic heart failure.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
| | - Gang Hui
- The Department of 31656 Troops of Chinese People's Liberation Army, Leshan, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Sattayaprasert P, Vasireddi SK, Bektik E, Jeon O, Hajjiri M, Mackall JA, Moravec CS, Alsberg E, Fu J, Laurita KR. Human Cardiac Mesenchymal Stem Cells Remodel in Disease and Can Regulate Arrhythmia Substrates. Circ Arrhythm Electrophysiol 2020; 13:e008740. [PMID: 32755466 PMCID: PMC7578059 DOI: 10.1161/circep.120.008740] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The mesenchymal stem cell (MSC), known to remodel in disease and have an extensive secretome, has recently been isolated from the human heart. However, the effects of normal and diseased cardiac MSCs on myocyte electrophysiology remain unclear. We hypothesize that in disease the inflammatory secretome of cardiac human MSCs (hMSCs) remodels and can regulate arrhythmia substrates. METHODS hMSCs were isolated from patients with or without heart failure from tissue attached to extracted device leads and from samples taken from explanted/donor hearts. Failing hMSCs or nonfailing hMSCs were cocultured with normal human cardiac myocytes derived from induced pluripotent stem cells. Using fluorescent indicators, action potential duration, Ca2+ alternans, and spontaneous calcium release (SCR) incidence were determined. RESULTS Failing and nonfailing hMSCs from both sources exhibited similar trilineage differentiation potential and cell surface marker expression as bone marrow hMSCs. Compared with nonfailing hMSCs, failing hMSCs prolonged action potential duration by 24% (P<0.001, n=15), increased Ca2+ alternans by 300% (P<0.001, n=18), and promoted spontaneous calcium release activity (n=14, P<0.013) in human cardiac myocytes derived from induced pluripotent stem cells. Failing hMSCs exhibited increased secretion of inflammatory cytokines IL (interleukin)-1β (98%, P<0.0001) and IL-6 (460%, P<0.02) compared with nonfailing hMSCs. IL-1β or IL-6 in the absence of hMSCs prolonged action potential duration but only IL-6 increased Ca2+ alternans and promoted spontaneous calcium release activity in human cardiac myocytes derived from induced pluripotent stem cells, replicating the effects of failing hMSCs. In contrast, nonfailing hMSCs prevented Ca2+ alternans in human cardiac myocytes derived from induced pluripotent stem cells during oxidative stress. Finally, nonfailing hMSCs exhibited >25× higher secretion of IGF (insulin-like growth factor)-1 compared with failing hMSCs. Importantly, IGF-1 supplementation or anti-IL-6 treatment rescued the arrhythmia substrates induced by failing hMSCs. CONCLUSIONS We identified device leads as a novel source of cardiac hMSCs. Our findings show that cardiac hMSCs can regulate arrhythmia substrates by remodeling their secretome in disease. Importantly, therapy inhibiting (anti-IL-6) or mimicking (IGF-1) the cardiac hMSC secretome can rescue arrhythmia substrates.
Collapse
Affiliation(s)
- Prasongchai Sattayaprasert
- Heart and Vascular Research Center, MetroHealth Campus, Case Western Reserve University, Cleveland, OH (P.S., S.K.V., M.H., K.R.L.)
| | - Sunil K Vasireddi
- Heart and Vascular Research Center, MetroHealth Campus, Case Western Reserve University, Cleveland, OH (P.S., S.K.V., M.H., K.R.L.)
| | - Emre Bektik
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA (E.B.)
| | - Oju Jeon
- Departments of Biomedical Engineering (O.J., E.A.), University of Illinois at Chicago
| | - Mohammad Hajjiri
- Heart and Vascular Research Center, MetroHealth Campus, Case Western Reserve University, Cleveland, OH (P.S., S.K.V., M.H., K.R.L.)
| | - Judith A Mackall
- Harrington Heart & Vascular Institute, University Hospitals Cleveland Medical Center (J.A.M.)
| | - Christine S Moravec
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland (C.S.M.)
| | - Eben Alsberg
- Departments of Biomedical Engineering (O.J., E.A.), University of Illinois at Chicago.,Orthopaedics (E.A.), University of Illinois at Chicago.,Pharmacology (E.A.), University of Illinois at Chicago.,Mechanical & Industrial Engineering (E.A.), University of Illinois at Chicago
| | - Jidong Fu
- Department of Physiology & Cell Biology, The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus (J.F.)
| | - Kenneth R Laurita
- Heart and Vascular Research Center, MetroHealth Campus, Case Western Reserve University, Cleveland, OH (P.S., S.K.V., M.H., K.R.L.)
| |
Collapse
|
17
|
Borchert T, Hess A, Lukačević M, Ross TL, Bengel FM, Thackeray JT. Angiotensin-converting enzyme inhibitor treatment early after myocardial infarction attenuates acute cardiac and neuroinflammation without effect on chronic neuroinflammation. Eur J Nucl Med Mol Imaging 2020; 47:1757-1768. [PMID: 32125488 PMCID: PMC7248052 DOI: 10.1007/s00259-020-04736-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Purpose Myocardial infarction (MI) triggers a local inflammatory response which orchestrates cardiac repair and contributes to concurrent neuroinflammation. Angiotensin-converting enzyme (ACE) inhibitor therapy not only attenuates cardiac remodeling by interfering with the neurohumoral system, but also influences acute leukocyte mobilization from hematopoietic reservoirs. Here, we seek to dissect the anti-inflammatory and anti-remodeling contributions of ACE inhibitors to the benefit of heart and brain outcomes after MI. Methods C57BL/6 mice underwent permanent coronary artery ligation (n = 41) or sham surgery (n = 9). Subgroups received ACE inhibitor enalapril (20 mg/kg, oral) either early (anti-inflammatory strategy; 10 days treatment beginning 3 days prior to surgery; n = 9) or delayed (anti-remodeling; continuous from 7 days post-MI; n = 16), or no therapy (n = 16). Cardiac and neuroinflammation were serially investigated using whole-body macrophage- and microglia-targeted translocator protein (TSPO) PET at 3 days, 7 days, and 8 weeks. In vivo PET signal was validated by autoradiography and histopathology. Results Myocardial infarction evoked higher TSPO signal in the infarct region at 3 days and 7 days compared with sham (p < 0.001), with concurrent elevation in brain TSPO signal (+ 18%, p = 0.005). At 8 weeks after MI, remote myocardium TSPO signal was increased, consistent with mitochondrial stress, and corresponding to recurrent neuroinflammation. Early enalapril treatment lowered the acute TSPO signal in the heart and brain by 55% (p < 0.001) and 14% (p = 0.045), respectively. The acute infarct signal predicted late functional outcome (r = 0.418, p = 0.038). Delayed enalapril treatment reduced chronic myocardial TSPO signal, consistent with alleviated mitochondrial stress. Early enalapril therapy tended to lower TSPO signal in the failing myocardium at 8 weeks after MI (p = 0.090) without an effect on chronic neuroinflammation. Conclusions Whole-body TSPO PET identifies myocardial macrophage infiltration and neuroinflammation after MI, and altered cardiomyocyte mitochondrial density in chronic heart failure. Improved chronic cardiac outcome by enalapril treatment derives partially from acute anti-inflammatory activity with complementary benefits in later stages. Whereas early ACE inhibitor therapy lowers acute neuroinflammation, chronic alleviation is not achieved by early or delayed ACE inhibitor therapy, suggesting a more complex mechanism underlying recurrent neuroinflammation in ischemic heart failure. Electronic supplementary material The online version of this article (10.1007/s00259-020-04736-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tobias Borchert
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Annika Hess
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Mario Lukačević
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg-Str. 1, D-30625, Hannover, Germany.
| |
Collapse
|
18
|
Nejm MB, Guimarães-Marques MJ, Oliveira LF, Damasceno L, Andersen ML, Tufik S, Fonseca F, Olszewer E, Leça R, de Almeida ACG, Scorza FA, Scorza CA. Assessment of vitamin D and inflammatory markers profile in cardiac tissue on Parkinson disease animal model. Pharmacol Rep 2020; 72:296-304. [PMID: 32124387 DOI: 10.1007/s43440-020-00074-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/13/2019] [Accepted: 12/30/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cardiovascular dysfunctions are common non-motor symptoms in patients with Parkinson's disease (PD) that can result in reduced quality of life and even death. Research in animal models designed to characterize the pathological association between PD and cardiovascular abnormalities is still in its infancy. This study assessed the early impact of the nigrostriatal dopaminergic damage on cardiological features in the unilateral 6-OHDA rat model of PD. METHODS Male Wistar rats received unilateral intrastriatal injections of 6-OHDA and sham rats were injected with saline. Animals were studied 15 days later. Immunohistochemistry was used for visualization of tyrosine hydroxylase (TH)-positive neurons in the nigrostriatal system. Electrocardiogram recordings of heart rate were performed in conscious rats. Heart levels of vitamin D, inflammatory cytokines and C-reactive protein were assessed through electrochemiluminescence immunoassay, quantitative reverse transcription PCR and turbidimetric method, respectively. RESULTS We found a post-injury reduction of TH-immunoreactivity of approximately 45% in the substantia nigra pars compacta and 20% in the striatum. Heart rate reduction was found in 6-OHDA-lesioned rats as compared with sham counterparts. Reduced levels of vitamin D and increased levels of inflammatory factors (C-reactive protein, IL-6, TNF-α and TGF-β) were detected in the heart tissue of PD rats in comparison with sham. CONCLUSION Our findings suggest a link between cardiac tissue changes and cardiac functional changes early after the central dopaminergic damage induced by 6-OHDA. Knowledge of the cardiac abnormalities in the 6-OHDA model is critical in identifying future therapeutic targets and disease-modifying approaches for PD non-motor features.
Collapse
Affiliation(s)
- Mariana Bocca Nejm
- Departamento de Neurologia/Neurociência, Universidade Federal de São Paulo-Escola Paulista de Medicina (UNIFESP/EPM), Av. Pedro de Toledo, 699, 1º andar, São Paulo, SP, Brazil
| | - Marcia Jonathas Guimarães-Marques
- Departamento de Neurologia/Neurociência, Universidade Federal de São Paulo-Escola Paulista de Medicina (UNIFESP/EPM), Av. Pedro de Toledo, 699, 1º andar, São Paulo, SP, Brazil
| | - Leandro Freitas Oliveira
- Departamento de Neurologia/Neurociência, Universidade Federal de São Paulo-Escola Paulista de Medicina (UNIFESP/EPM), Av. Pedro de Toledo, 699, 1º andar, São Paulo, SP, Brazil
| | - Laís Damasceno
- Departamento de Neurologia/Neurociência, Universidade Federal de São Paulo-Escola Paulista de Medicina (UNIFESP/EPM), Av. Pedro de Toledo, 699, 1º andar, São Paulo, SP, Brazil
| | - Monica L Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo-Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo-Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Fernando Fonseca
- Laboratório de Análises Clínicas da Faculdade de Medicina do ABC, Santo André, SP, Brazil
- Departamento de Ciências Farmacêuticas da, Universidade Federal de São Paulo (UNIFESP), Diadema, SP, Brazil
| | - Efrain Olszewer
- Fundação de Apoio à Pesquisa e Estudo na Área de Saúde (FAPES), São Paulo, SP, Brazil
| | - Renato Leça
- Departamento de Cirurgia II, Faculdade de Medicina do ABC, Santo André, SP, Brazil
| | - Antonio Carlo G de Almeida
- Departamento de Engenharia de Biossistemas, Universidade Federal de São João Del Rei (UFSJ), São João Del Rei, MG, Brazil
| | - Fulvio Alexandre Scorza
- Departamento de Neurologia/Neurociência, Universidade Federal de São Paulo-Escola Paulista de Medicina (UNIFESP/EPM), Av. Pedro de Toledo, 699, 1º andar, São Paulo, SP, Brazil
| | - Carla Alessandra Scorza
- Departamento de Neurologia/Neurociência, Universidade Federal de São Paulo-Escola Paulista de Medicina (UNIFESP/EPM), Av. Pedro de Toledo, 699, 1º andar, São Paulo, SP, Brazil.
| |
Collapse
|
19
|
Yuan Y, Su M, Liu J, Lou Y, Xia Y, Zhou B. Four-week intravenous repeated dose toxicity study of vitacamphorae injection in rats. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:2001-2007. [PMID: 31960155 DOI: 10.1007/s00210-020-01820-6] [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: 09/23/2019] [Accepted: 01/10/2020] [Indexed: 01/15/2023]
Abstract
The study was undertaken to evaluate the safety of vitacamphorae (VCP) injection in Sprague-Dawley (SD) rats. Rats were intravenously administered with VCP at the doses of 0, 5, 15, and 50 mg/kg/day (equivalent to 0, 5, 15, and 50 times the clinical equivalent dose) for 4 weeks, respectively. In addition, we also tested oxidative stress-related parameters and cytokine levels in rat serum. In the current study, intravenous administration of VCP at a dose of 50 mg/kg/day caused significant pathophysiological responses in rats. Compared with the control group, different doses of VCP exposure had no significant effect on body weight, food consumption, and clinic pathology of rats after 4 weeks of VCP administration. Rats in high-dose group (50 mg/kg/day) showed general symptoms of convulsions after VCP administration. The toxicological significance of VCP exposure in the spleen of high-dose female rats was observed, which showed a significant increase in the relative spleen weights (P < 0.01) and mild lymphocyte proliferation in splenic pathology. Furthermore, the results of oxidative stress and cytokine detection showed that the levels of antioxidant enzymes SOD increased in each administration group, but the levels of a series of pro-inflammatory cytokines IL-1β, IL-6, IL-8, IL-12, and IFN-γ also increased in these groups. Above changes caused by VCP exposure can be reversed after 4 weeks of recovery. Overall, the results showed that the no-observed-adverse-effect-level (NOAEL) of VCP injection for 4-week toxicity was 15 mg/kg/day.
Collapse
Affiliation(s)
- Yazhi Yuan
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Mei Su
- Jiangsu Carephar Pharmaceutical Co., Ltd, Nanjing, 210009, China
| | - Jing Liu
- Jiangsu Center for Safety Evaluation of Drugs, School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, China
| | - Yajing Lou
- Jiangsu Carephar Pharmaceutical Co., Ltd, Nanjing, 210009, China
| | - Yufeng Xia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Bo Zhou
- Jiangsu Center for Safety Evaluation of Drugs, School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, China.
| |
Collapse
|
20
|
Majeed MH, Ali AA, Khalil HA, Bacon D, Imran HM. A Review of the Pharmacological Management of Chronic Pain in Patients with Heart Failure. INNOVATIONS IN CLINICAL NEUROSCIENCE 2019; 16:25-27. [PMID: 32082939 PMCID: PMC7006868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background: The incidence rates of heart failure (HF) and chronic pain increase with age. In the geriatric population, both disorders often coexist and pose a challenge to clinicians in treating them simultaneously. Methods: We conducted an online literature search for reports of the heart failure effects of pharmacological treatments for chronic pain. Results: Topical pain medications are favored agents because of their efficacy, tolerability, and favorable side-effect profile. Acetaminophen is a preferred oral medication for the treatment of pain in patients with HF. Due to deleterious effects including HF, the long-term use of oral nonsteroidal anti-inflammatory drugs and gabapentinoids are discouraged. Conclusion: Prescribers should thoroughly consider the risk-benefit ratio and individual patient-risk profile before instituting pharmacological treatment for chronic pain in patients with HF.
Collapse
Affiliation(s)
- Muhammad Hassan Majeed
- Dr. Majeed is with the Department of Anesthesiology and Critical Care Management, Johns Hopkins Hospital in Baltimore, Maryland
- Dr. Ali is with the Department of Psychiatry, Icahn School of Medicine at Mount Sinai (Elmhurst) in Queens, New York
- Dr. Khalil is with Good Samariton Hospital in Lebanon, Pennsylvania
- Dr. Bacon is with the Department of Anesthesiology, The University of Mississippi Medical Center School of Medicine in Jackson, Mississippi
- Dr. Imran is with Providence VA Medical Center/Rhode Island Hospital in Providence, Rhode Island
| | - Ali Ahsan Ali
- Dr. Majeed is with the Department of Anesthesiology and Critical Care Management, Johns Hopkins Hospital in Baltimore, Maryland
- Dr. Ali is with the Department of Psychiatry, Icahn School of Medicine at Mount Sinai (Elmhurst) in Queens, New York
- Dr. Khalil is with Good Samariton Hospital in Lebanon, Pennsylvania
- Dr. Bacon is with the Department of Anesthesiology, The University of Mississippi Medical Center School of Medicine in Jackson, Mississippi
- Dr. Imran is with Providence VA Medical Center/Rhode Island Hospital in Providence, Rhode Island
| | - Hafiza Ayesha Khalil
- Dr. Majeed is with the Department of Anesthesiology and Critical Care Management, Johns Hopkins Hospital in Baltimore, Maryland
- Dr. Ali is with the Department of Psychiatry, Icahn School of Medicine at Mount Sinai (Elmhurst) in Queens, New York
- Dr. Khalil is with Good Samariton Hospital in Lebanon, Pennsylvania
- Dr. Bacon is with the Department of Anesthesiology, The University of Mississippi Medical Center School of Medicine in Jackson, Mississippi
- Dr. Imran is with Providence VA Medical Center/Rhode Island Hospital in Providence, Rhode Island
| | - Douglas Bacon
- Dr. Majeed is with the Department of Anesthesiology and Critical Care Management, Johns Hopkins Hospital in Baltimore, Maryland
- Dr. Ali is with the Department of Psychiatry, Icahn School of Medicine at Mount Sinai (Elmhurst) in Queens, New York
- Dr. Khalil is with Good Samariton Hospital in Lebanon, Pennsylvania
- Dr. Bacon is with the Department of Anesthesiology, The University of Mississippi Medical Center School of Medicine in Jackson, Mississippi
- Dr. Imran is with Providence VA Medical Center/Rhode Island Hospital in Providence, Rhode Island
| | - Hafiz Muhammad Imran
- Dr. Majeed is with the Department of Anesthesiology and Critical Care Management, Johns Hopkins Hospital in Baltimore, Maryland
- Dr. Ali is with the Department of Psychiatry, Icahn School of Medicine at Mount Sinai (Elmhurst) in Queens, New York
- Dr. Khalil is with Good Samariton Hospital in Lebanon, Pennsylvania
- Dr. Bacon is with the Department of Anesthesiology, The University of Mississippi Medical Center School of Medicine in Jackson, Mississippi
- Dr. Imran is with Providence VA Medical Center/Rhode Island Hospital in Providence, Rhode Island
| |
Collapse
|
21
|
Kosmas CE, Silverio D, Sourlas A, Montan PD, Guzman E, Garcia MJ. Anti-inflammatory therapy for cardiovascular disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:147. [PMID: 31157268 DOI: 10.21037/atm.2019.02.34] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic subclinical inflammation is a central process in the pathogenesis of cardiovascular disease (CVD) and it has been linked with both the initiation and progression of atherosclerosis. Several pro-inflammatory cytokines, such as the C-reactive protein (CRP), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) have been described as independent risk factors for coronary heart disease and promoters of atherogenesis. Thus, extensive research is being conducted to assess the role of anti-inflammatory therapy in the primary and secondary prevention of CVD. Our review aims to provide the clinical and scientific data pertaining to the effects of different anti-inflammatory agents administered in patients with CVD.
Collapse
Affiliation(s)
| | - Delia Silverio
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | | | - Peter D Montan
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | - Eliscer Guzman
- Department of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Mario J Garcia
- Department of Medicine, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
22
|
Pascual-Figal DA, Bayes-Genis A, Asensio-Lopez MC, Hernández-Vicente A, Garrido-Bravo I, Pastor-Perez F, Díez J, Ibáñez B, Lax A. The Interleukin-1 Axis and Risk of Death in Patients With Acutely Decompensated Heart Failure. J Am Coll Cardiol 2019; 73:1016-1025. [DOI: 10.1016/j.jacc.2018.11.054] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
|
23
|
Sydykov A, Mamazhakypov A, Petrovic A, Kosanovic D, Sarybaev AS, Weissmann N, Ghofrani HA, Schermuly RT. Inflammatory Mediators Drive Adverse Right Ventricular Remodeling and Dysfunction and Serve as Potential Biomarkers. Front Physiol 2018; 9:609. [PMID: 29875701 PMCID: PMC5974151 DOI: 10.3389/fphys.2018.00609] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 05/04/2018] [Indexed: 01/07/2023] Open
Abstract
Adverse right ventricular (RV) remodeling leads to ventricular dysfunction and failure that represents an important determinant of outcome in patients with pulmonary hypertension (PH). Recent evidence indicates that inflammatory activation contributes to the pathogenesis of adverse RV remodeling and dysfunction. It has been shown that accumulation of inflammatory cells such as macrophages and mast cells in the right ventricle is associated with maladaptive RV remodeling. In addition, inhibition of inflammation in animal models of RV failure ameliorated RV structural and functional impairment. Furthermore, a number of circulating inflammatory mediators have been demonstrated to be associated with RV performance. This work reviews the role of inflammation in RV remodeling and dysfunction and discusses anti-inflammatory strategies that may attenuate adverse structural alterations while promoting improvement of RV function.
Collapse
Affiliation(s)
- Akylbek Sydykov
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany.,Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek, Kyrgyzstan
| | - Argen Mamazhakypov
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Aleksandar Petrovic
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Djuro Kosanovic
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Akpay S Sarybaev
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek, Kyrgyzstan
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary System, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Justus Liebig University of Giessen, Giessen, Germany
| |
Collapse
|