1
|
Mahmoudi L, Izadpanah P, Asadi S. The effect of calcitriol and cholecalciferol on inflammatory markers in periprocedural myocardial injury: A randomized controlled trial. Medicine (Baltimore) 2025; 104:e42103. [PMID: 40228253 PMCID: PMC11999415 DOI: 10.1097/md.0000000000042103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Inflammation is an important factor in the development of cardiac injury during and after percutaneous coronary intervention (PCI). Vitamin D receptors play a significant role in the cardiovascular system and have anti-inflammatory effects. Hence, our goal was to evaluate the impact of calcitriol and cholecalciferol as vitamin D receptors agonists on inflammatory biomarkers in patients who are undergoing elective PCI. METHODS In this controlled clinical trial, patients undergoing elective PCI were randomly assigned to receive either calcitriol and cholecalciferol or were placed in the control group from July 2021 to November 2022. Calcitriol and cholecalciferol were administered at doses of 1 mcg and 300,000 international units, respectively, before the procedure. High-sensitive C-reactive protein (hs-CRP) was evaluated as the main inflammatory biomarker and other relevant clinical and laboratory data were also included. RESULTS During the study, 180 patients were allocated into three groups, each consisting of 60 patients, with a mean age of 62.26 ± 8.73 years. The prevalence of the underlying conditions was not different among the groups. After 24 hours, hs-CRP levels were lower (P = .012), and a significantly lower increase from baseline was observed (P = .003) in the group that received calcitriol. However, no significant differences were observed in Troponin I and creatine kinase-MB levels (P > .05). CONCLUSIONS Administration of calcitriol was associated with significantly lower levels of hs-CRP, the main cardiac inflammatory marker, in patients undergoing elective PCI. Further clinical studies with a larger sample size are needed to assess the clinical impact of this anti-inflammatory effect.
Collapse
Affiliation(s)
- Laleh Mahmoudi
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Peyman Izadpanah
- Department of Cardiovascular Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Asadi
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Jiang C, Yan Y, Long T, Xu J, Chang C, Kang M, Wang X, Chen Y, Qiu J. Ferroptosis: a potential therapeutic target in cardio-cerebrovascular diseases. Mol Cell Biochem 2025:10.1007/s11010-025-05262-7. [PMID: 40148662 DOI: 10.1007/s11010-025-05262-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Cardio-cerebrovascular diseases (CCVDs) are the leading cause of global mortality, yet effective treatment options remain limited. Ferroptosis, a novel form of regulated cell death, has emerged as a critical player in various CCVDs, including atherosclerosis, myocardial infarction, ischemia-reperfusion injury, cardiomyopathy, and ischemic/hemorrhagic strokes. This review highlights the core mechanisms of ferroptosis, its pathological implications in CCVDs, and the therapeutic potential of targeting this process. Additionally, it explores the role of Chinese herbal medicines (CHMs) in mitigating ferroptosis, offering novel therapeutic strategies for CCVDs management. Ferroptosis is regulated by several key pathways. The GPX4-GSH-System Xc- axis is central to ferroptosis execution, involving GPX4 using GSH to neutralize lipid peroxides, with system Xc- being crucial for GSH synthesis. The NAD(P)H/FSP1/CoQ10 axis involves FSP1 regenerating CoQ10 via NAD(P)H, inhibiting lipid peroxidation independently of GPX4. Lipid peroxidation, driven by PUFAs and enzymes like ACSL4 and LPCAT3, and iron metabolism, regulated by proteins like TfR1 and ferritin, are also crucial for ferroptosis. Inhibiting ferroptosis shows promise in managing CCVDs. In atherosclerosis, ferroptosis inhibitors reduce iron accumulation and lipid peroxidation. In myocardial infarction, inhibitors protect cardiomyocytes by preserving GPX4 and SLC7A11 levels. In ischemia-reperfusion injury, targeting ferroptosis reduces myocardial and cerebral damage. In diabetic cardiomyopathy, Nrf2 activators alleviate oxidative stress and iron metabolism irregularities. CHMs offer natural compounds that mitigate ferroptosis. They possess antioxidant properties, chelate iron, and modulate signaling pathways like Nrf2 and AMPK. For example, Salvia miltiorrhiza and Astragalus membranaceus reduce oxidative stress, while some CHMs chelate iron, reducing its availability for ferroptosis. In conclusion, ferroptosis plays a pivotal role in CCVDs, and targeting it offers novel therapeutic avenues. CHMs show promise in reducing ferroptosis and improving patient outcomes. Future research should explore combination therapies and further elucidate the molecular interactions in ferroptosis.
Collapse
Affiliation(s)
- Chenlong Jiang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Yang Yan
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Tianlin Long
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Jiawei Xu
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Cuicui Chang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Meili Kang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Xuanqi Wang
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| | - Yuhua Chen
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China.
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China.
- School of Life and Health Science, Hainan University, No. 58 People's Avenue, Haikou, 570100, Hainan, China.
| | - Junlin Qiu
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| |
Collapse
|
3
|
Kotit S. Benefits of intravenous iron supplementation in heart failure. Glob Cardiol Sci Pract 2024; 2024:e202410. [PMID: 38746071 PMCID: PMC11090186 DOI: 10.21542/gcsp.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/14/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction: Iron deficiency (ID) is one of the most frequent comorbidities in patients with heart failure (HF) and is estimated to be present in up to 80% of acute patients regardless of their ejection fraction. Randomized controlled trials have shown that supplementary intravenous iron results in improved clinical outcomes; however, the current understanding of the effects of intravenous iron on morbidity and mortality remains limited. Study and results: The meta-analysis pooled individual participant data from three randomized placebo-controlled trials of ferric carboxymaltose (FCM) in adult patients (n = 4,501) with heart failure and iron deficiency (CONFIRM-HF, AFFIRM-AHF, and HEART-FID). FCM therapy significantly reduced the co-primary composite endpoint of total cardiovascular hospitalizations and cardiovascular death, with a rate ratio (RR 0.86; 95% CI 0.75 to 0.98; p = 0.029). FCM therapy was associated with a 17% relative rate reduction in total cardiovascular hospitalizations (RR 0.83; 95% CI 0.73 to 0.96; p = 0.009) and a 16% relative rate reduction in total heart failure hospitalizations (RR 0.84; 95% CI 0.71 to 0.98; p = 0.025). Lessons learned: The meta-analysis shows that in iron-deficient patients with heart failure and reduced or mildly reduced left ventricular ejection fraction, intravenous ferric carboxymaltose (FCM) is associated with a reduced risk of total cardiovascular hospitalization and cardiovascular mortality. These findings indicate that intravenous FCM should be considered in iron-deficient patients with heart failure and reduced or mildly reduced ejection fractions.
Collapse
|
4
|
Papamichail A, Kourek C, Briasoulis A, Xanthopoulos A, Tsougos E, Farmakis D, Paraskevaidis I. Targeting Key Inflammatory Mechanisms Underlying Heart Failure: A Comprehensive Review. Int J Mol Sci 2023; 25:510. [PMID: 38203681 PMCID: PMC10778956 DOI: 10.3390/ijms25010510] [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: 12/05/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Inflammation is a major component of heart failure (HF), causing peripheral vasculopathy and cardiac remodeling. High levels of circulating inflammatory cytokines in HF patients have been well recognized. The hallmark of the inflammatory imbalance is the insufficient production of anti-inflammatory mediators, a condition that leads to dysregulated cytokine activity. The condition progresses because of the pathogenic consequences of the cytokine imbalance, including the impact of endothelial dysfunction and adrenergic responsiveness deterioration, and unfavorable inotropic effects on the myocardium. Hence, to develop possible anti-inflammatory treatment options that will enhance the outcomes of HF patients, it is essential to identify the potential pathophysiological mechanisms of inflammation in HF. Inflammatory mediators, such as cytokines, adhesion molecules, and acute-phase proteins, are elevated during this process, highlighting the complex association between inflammation and HF. Therefore, these inflammatory markers can be used in predicting prognosis of the syndrome. Various immune cells impact on myocardial remodeling and recovery. They lead to stimulation, release of alarmins and risk-related molecule patterns. Targeting key inflammatory mechanisms seems a quite promising therapy strategy in HF. Cytokine modulation is only one of several possible targets in the fight against inflammation, as the potential molecular targets for therapy in HF include immune activation, inflammation, oxidative stress, alterations in mitochondrial bioenergetics, and autophagy.
Collapse
Affiliation(s)
- Adamantia Papamichail
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (A.P.); (C.K.); (A.B.)
| | - Christos Kourek
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (A.P.); (C.K.); (A.B.)
| | - Alexandros Briasoulis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (A.P.); (C.K.); (A.B.)
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece;
| | - Elias Tsougos
- Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece;
| | - Dimitrios Farmakis
- Attikon University Hospital, Medical School of Athens, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Ioannis Paraskevaidis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (A.P.); (C.K.); (A.B.)
| |
Collapse
|
5
|
Lima do Vale MR, Buckner L, Mitrofan CG, Tramontt CR, Kargbo SK, Khalid A, Ashraf S, Mouti S, Dai X, Unwin D, Bohn J, Goldberg L, Golubic R, Ray S. A synthesis of pathways linking diet, metabolic risk and cardiovascular disease: a framework to guide further research and approaches to evidence-based practice. Nutr Res Rev 2023; 36:232-258. [PMID: 34839838 DOI: 10.1017/s0954422421000378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cardiovascular disease (CVD) is the most common non-communicable disease occurring globally. Although previous literature has provided useful insights into the important role that diet plays in CVD prevention and treatment, understanding the causal role of diets is a difficult task considering inherent and introduced weaknesses of observational (e.g. not properly addressing confounders and mediators) and experimental research designs (e.g. not appropriate or well designed). In this narrative review, we organised current evidence linking diet, as well as conventional and emerging physiological risk factors, with CVD risk, incidence and mortality in a series of diagrams. The diagrams presented can aid causal inference studies as they provide a visual representation of the types of studies underlying the associations between potential risk markers/factors for CVD. This may facilitate the selection of variables to be considered and the creation of analytical models. Evidence depicted in the diagrams was systematically collected from studies included in the British Nutrition Task Force report on diet and CVD and database searches, including Medline and Embase. Although several markers and disorders linked to conventional and emerging risk factors for CVD were identified, the causal link between many remains unknown. There is a need to address the multifactorial nature of CVD and the complex interplay between conventional and emerging risk factors with natural and built environments, while bringing the life course into the spotlight.
Collapse
Affiliation(s)
| | - Luke Buckner
- NNEdPro Global Centre for Nutrition and Health, Cambridge, UK
| | | | | | | | - Ali Khalid
- NNEdPro Global Centre for Nutrition and Health, Cambridge, UK
| | - Sammyia Ashraf
- NNEdPro Global Centre for Nutrition and Health, Cambridge, UK
| | - Saad Mouti
- University of California Berkeley, Consortium for Data Analytics in Risk, Berkeley, CA, USA
| | - Xiaowu Dai
- University of California Berkeley, Consortium for Data Analytics in Risk, Berkeley, CA, USA
| | | | - Jeffrey Bohn
- University of California Berkeley, Consortium for Data Analytics in Risk, Berkeley, CA, USA
- Swiss Re Institute, Zürich, Switzerland
| | - Lisa Goldberg
- University of California Berkeley, Consortium for Data Analytics in Risk, Berkeley, CA, USA
| | - Rajna Golubic
- NNEdPro Global Centre for Nutrition and Health, Cambridge, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Diabetes Trials Unit, University of Oxford, Oxford, UK
| | - Sumantra Ray
- NNEdPro Global Centre for Nutrition and Health, Cambridge, UK
- University of Ulster, School of Biomedical Sciences, Coleraine, UK
- University of Cambridge, School of the Humanities and Social Sciences, Cambridge, UK
| |
Collapse
|
6
|
Xu Q, Wang Z, You S, Yuan L, Li H. Mechanism for effect of tanshinone IIA on alleviating cardiomyocyte injury induced by oxygen glucose deprivation. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:369-375. [PMID: 37164920 PMCID: PMC10930080 DOI: 10.11817/j.issn.1672-7347.2023.220173] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Indexed: 05/12/2023]
Abstract
OBJECTIVES Tanshinone IIA has a wide range of myocardial protective effects. AK003290 is a long noncoding RNA (lncRNA) that is highly expressed in myocardial tissue, and its expression is down-regulated when myocardial injury occurs. This study aims to explore the mechanism for tanshinone IIA in alleviating myocardial cell damage induced by oxygen glucose deprivation (OGD). METHODS OGD model was established in rat H9C2 cardiomyocytes. siRNA was transfected to reduce AK003290 expression. H9C2 cells were divided into 6 groups: A control group, a tanshinone IIA (TAN) group, an OGD group, a tanshinone IIA+OGD (TAN+OGD) group, a scrambled siRNA transfection+tanshinone IIA+OGD (scrambled siRNA+TAN+OGD) group, and a AK003290 siRNA transfection+tanshinone IIA+OGD (AK003290 siRNA+TAN+OGD) group. H9C2 cells in the TAN group were treated with 40 μmol/L tanshinone IIA for 12 h. The TAN+OGD group was treated with 40 μmol/L tanshinone IIA for 12 h, followed by OGD treatment for 12 h. The scrambled siRNA+TAN+OGD group and AK003290 siRNA+TAN+OGD group were transfected with the scrambled siRNA or AK003290 siRNA. Twenty-four hours later, the cells were treated with tanshinone IIA and OGD. Real-time RT-PCR was used to detect the expression of AK003290. Spectrophotometry was used to detect the content of lactate dehydrogenase (LDH) in cell culture medium to reflect LDH leakage rate, and enzyme-linked immunosorbent assay (ELISA) was used to detect the content of interleukin-1β (IL-1β) and interleukin-18 (IL-18). Western blotting was used to detect the protein expression of phospho-nuclear factor- κB (p-NF-κB). RESULTS Compared with the control group, the leakage rate of LDH, the content of IL-1β and IL-18 in culture medium, and the protein expression level of p-NF-κB were increased in the OGD group (P<0.01 or P<0.001). Compared with the OGD group, the leakage rate of LDH, the content of IL-1β and IL-18 in culture medium, and the protein expression level of p-NF-κB were decreased in the TAN+OGD group (P<0.05 or P<0.01). Compared with the control group, the AK003290 expression was increased in the TAN group (P<0.01) and it was decreased in the OGD group (P<0.05). Compared with the OGD group, the AK003290 expression was increased in the TAN+OGD group (P<0.05). Compared with the scrambled siRNA+TAN+OGD group, the leakage rate of LDH, the content of IL-1β and IL-18 in culture medium, and the protein expression level of p-NF-κB were increased in the AK003290 siRNA+TAN+OGD group (P<0.05 or P<0.01). CONCLUSIONS Tanshinone IIA inhibits NF-κB activity and attenuates OGD-induced inflammatory injury of cardiomyocytes through up-regulating AK003290.
Collapse
Affiliation(s)
- Qin Xu
- Department of Cardiology, Brain Hospital of Hunan Province, Changsha 410007, China.
| | - Zhaohua Wang
- Department of Cardiology, Brain Hospital of Hunan Province, Changsha 410007, China
| | - Sanli You
- Department of Cardiology, Brain Hospital of Hunan Province, Changsha 410007, China
| | - Lili Yuan
- Department of Cardiology, Brain Hospital of Hunan Province, Changsha 410007, China
| | - He Li
- Department of Cardiology, Brain Hospital of Hunan Province, Changsha 410007, China.
| |
Collapse
|
7
|
Parmana IMA, Boom CE, Rachmadi L, Hanafy DA, Widyastuti Y, Mansyur M, Siswanto BB. Correlation Between Cardiac Index, Plasma Troponin I, Myocardial Histopathology, CPB and AoX Duration in Glutamine versus No Glutamine Administered Patients with Low Ejection Fraction Undergoing Elective On-Pump CABG Surgery: Secondary Analysis of an RCT. Vasc Health Risk Manag 2023; 19:93-101. [PMID: 36880009 PMCID: PMC9985398 DOI: 10.2147/vhrm.s399925] [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: 12/05/2022] [Accepted: 02/18/2023] [Indexed: 03/04/2023] Open
Abstract
Purpose On-pump coronary artery bypass graft (CABG) causes myocardial ischemia, through the cardiopulmonary bypass (CPB) and aortic cross-clamping (AoX). Glutamine supplementation protects cardiac cells during cardiac ischemia. This study analysed the correlation between cardiac index (CI), plasma troponin I, myocardial histopathology, CPB and AoX duration in low ejection fraction patients receiving glutamine and no glutamine undergoing elective on-pump CABG. Material and Methods This was a secondary analysis of a double-blind, randomised controlled trial of 60 patients, split into control and intervention (glutamine) groups. Glutamine was administered at a dose of 0.5 g/kg/24 hours. There were 29 patients in each respective groups after a total of two patients dropped out. Results A negative correlation (p = 0.037) was observed between CPB duration and CI at 6 hours after CPB in the glutamine group. A positive correlation (p = 0.002) was also observed between AoX duration and plasma troponin I at 6 hours after CPB in the control group. However, no correlation was observed between myocardial histopathology and plasma troponin I level at 5 minutes after CPB. Conclusion Significant negative correlation between CPB duration and CI at 6 hours after CPB in the glutamine group, along with significant positive correlation between AoX duration and plasma troponin I level at 6 hours after CPB in the control group demonstrated the myocardial protection qualities of intravenous glutamine administration in patients with low ejection fraction undergoing elective on-pump CABG surgeries.
Collapse
Affiliation(s)
- I Made Adi Parmana
- Department of Anesthesiology and Intensive Care, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Cindy Elfira Boom
- Department of Anesthesiology and Intensive Care, National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Lisnawati Rachmadi
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia/Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Dudy Arman Hanafy
- Department of Cardiothoracic and Vascular Surgery, Faculty of Medicine, Universitas Indonesia/National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| | - Yunita Widyastuti
- Department of Anesthesiology and Intensive Care, Universitas Gadjah Mada/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Muchtaruddin Mansyur
- Department of Community Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Bambang Budi Siswanto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia/National Cardiovascular Center Harapan Kita, Jakarta, Indonesia
| |
Collapse
|
8
|
Kuna J, Żuber Z, Chmielewski G, Gromadziński L, Krajewska-Włodarczyk M. Role of Distinct Macrophage Populations in the Development of Heart Failure in Macrophage Activation Syndrome. Int J Mol Sci 2022; 23:2433. [PMID: 35269577 PMCID: PMC8910409 DOI: 10.3390/ijms23052433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage activation syndrome (MAS) is one of the few entities in rheumatology with the potential to quickly cause multiple organ failure and loss of life, and as such, requires urgent clinical intervention. It has a broad symptomatology, depending on the organs it affects. One especially dangerous aspect of MAS's course of illness is myocarditis leading to acute heart failure and possibly death. Research in recent years has proved that macrophages settled in different organs are not a homogenous group, with particular populations differing in both structure and function. Within the heart, we can determine two major groups, based on the presence of the C-C 2 chemokine receptor (CCR2): CCR2+ and CCR2-. There are a number of studies describing their function and the changes in the population makeup between normal conditions and different illnesses; however, to our knowledge, there has not been one touching on the matter of changes occurring in the populations of heart macrophages during MAS and their possible consequences. This review summarizes the most recent knowledge on heart macrophages, the influence of select cytokines (those particularly significant in the development of MAS) on their activity, and both the immediate and long-term consequences of changes in the makeup of specific macrophage populations-especially the loss of CCR2- cells that are responsible for regenerative processes, as well as the substitution of tissue macrophages by the highly proinflammatory CCR2+ macrophages originating from circulating monocytes. Understanding the significance of these processes may lead to new discoveries that could improve the therapeutic methods in the treatment of MAS.
Collapse
Affiliation(s)
- Jakub Kuna
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Zbigniew Żuber
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Kraków University, 30-705 Kraków, Poland;
| | - Grzegorz Chmielewski
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Leszek Gromadziński
- Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| |
Collapse
|
9
|
Mykula YI, Kupnovytska IH, Danulyk OI. Indicators of endothelial function and systemic immune inflammatory response in patients with chronic heart failure and coexisting primary hypothyroidism. PHARMACIA 2022. [DOI: 10.3897/pharmacia.69.e77225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Chronic heart failure is one of the leading causes of death globally, affecting 1.5 to 2% of the total world population and 2.9 to 3.9% of the total Western European population. Chronic heart failure often progresses rapidly in coexistence with endocrine pathology, namely hypothyroidism, that results in a more rapid development and further progression of endothelial dysfunction and the development of a systemic inflammatory response. The aim of our research was to study the levels of endothelin-1, C-reactive protein, tumor necrosis factor α and their correlation with the levels of thyroid-stimulating hormone, thyroxine in patients with chronic heart failure and coexisting hypothyroidism. There were examined 38 patients with chronic heart failure and coexisting hypothyroidism and 42 patients with chronic heart failure without hypothyroidism. The serum levels of endothelin-1, C-reactive protein, tumor necrosis factor α were determined by the enzyme-linked immunosorbent assay, while the levels of thyroid-stimulating hormone and thyroxine were determined by the electrochemiluminescence immunoassay. In patients with chronic heart failure and coexisting hypothyroidism, the levels of endothelin-1, C-reactive protein, and tumor necrosis factor α were 2.9, 1.5 and 2.27 times higher than those in patients without hypothyroidism. In Group I, there was a moderate positive correlation between the serum levels of endothelin-1 and thyroid-stimulating hormone and a weak negative correlation between the levels of thyroxine and endothelin-1. In Group II, there was a weak correlation between the levels of endothelin-1 and thyroid-stimulating hormone and no correlation between the levels of thyroxine and endothelin-1. In Group I, there was a strong positive correlation between C-reactive protein and thyroid-stimulating hormone levels as well; in Group II, no similar correlation was found. In Group I, there was found a moderate negative correlation between tumor necrosis factor α and thyroxine levels. According to our results, there was a close correlation between the markers of endothelial dysfunction, immune inflammatory response, and single markers of hypothyroidism.
Collapse
|
10
|
Fang Y, Wang S, Lv J, Zhao Z, Guo N, Wu G, Tong J, Wang Z. Slc39a2-Mediated Zinc Homeostasis Modulates Innate Immune Signaling in Phenylephrine-Induced Cardiomyocyte Hypertrophy. Front Cardiovasc Med 2021; 8:736911. [PMID: 34790705 PMCID: PMC8592093 DOI: 10.3389/fcvm.2021.736911] [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: 07/06/2021] [Accepted: 10/12/2021] [Indexed: 01/05/2023] Open
Abstract
Zinc dyshomeostasis has been involved in the pathogenesis of cardiac hypertrophy; however, the dynamic regulation of intracellular zinc and its downstream signaling in cardiac hypertrophy remain largely unknown. Using Zincpyr1 staining, we found a significant decrease of intracellular Zinc concentration in phenylephrine (PE)-induced hypertrophy of neonatal rat ventricular myocytes (NRVMs). We then screened SLC39 family members responsible for zinc uptake and identified Slc39a2 as the only one altered by PE treatment. Slc39a2 knockdown in NRVMs reduced the intracellular Zinc level, and exacerbated the hypertrophic responses to PE treatment. In contrast, adenovirus-mediated Slc39a2 overexpression enhanced zinc uptake and suppressed PE-induced Nppb expression. RNA sequencing analysis showed a pro-hypertrophic transcriptome reprogramming after Slc39a2 knockdown. Interestingly, the innate immune signaling pathways, including NOD signaling, TOLL-like receptor, NFκB, and IRFs, were remarkably enriched in the Slc39a2-regulated genes. Slc39a2 deficiency enhanced the phosphorylation of P65 NFκB and STAT3, and reduced the expression of IκBα. Finally, the expression of IRF7 was significantly increased by Slc39a2 knockdown, which was in turn suppressed by IRF7 knockdown. Our data demonstrate that zinc homeostasis mediated by a Slc39a2/IRF7 regulatory circuit contributes to the alteration of innate immune signaling in cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Yu Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Lv
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenyi Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,School of Pharmacy, Health Science Center, Shenzhen University, Shenzhen, China
| | - Ningning Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Tong
- School of Life Sciences, Central China Normal University, Wuhan, China
| | - Zhihua Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.,State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Mirna M, Holnthoner M, Topf A, Jirak P, Fejzic D, Paar V, Kellermair J, Blessberger H, Reiter C, Kammler J, Motloch LJ, Jung C, Kretzschmar D, Franz M, Alushi B, Lauten A, Hoppe UC, Steinwender C, Lichtenauer M. Tumor necrosis factor alpha-an underestimated risk predictor in patients undergoing transcatheter aortic valve replacement (TAVR)? J Clin Lab Anal 2021; 35:e23977. [PMID: 34562276 PMCID: PMC8605157 DOI: 10.1002/jcla.23977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/06/2021] [Accepted: 08/14/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Systemic inflammation has been identified as a major cardiovascular risk factor in patients undergoing transcatheter aortic valve replacement (TAVR), yet currently, it is not adequately portrayed in scores for pre-interventional risk assessment. The aim of this study was to investigate the predictive ability of TNF-α in TAVR. METHODS A total of 431 patients undergoing transfemoral TAVR were enrolled in this study. Blood samples were drawn prior to intervention, 24 h post-intervention, 4, 5, and 7 days post-intervention, and 1, 3, and 6 months post-TAVR. RESULTS In a univariate Cox proportional hazard analysis, plasma concentrations of TNF-α after 24 h and after 5 days were associated with mortality after 12 months (after 24 h: HR 1.002 (1.000-1.004), p = 0.028; after 5d: HR 1.003 (1.001-1.005), p = 0.013). This association remained significant even after correction for confounders in a multivariate Cox regression analysis. Additionally, cut-offs were calculated. Patients above the cut-off for TNF-α after 5d had a significantly worse 12-month mortality than patients below the cut-off (18.8% vs. 2.8%, p = 0.046). CONCLUSION Plasma levels of TNF-α after 24 h and 5 days were independently associated with 12-month mortality in patients undergoing TAVR. Thus, TNF-α could represent a novel biomarker for enhanced risk stratification in these patients.
Collapse
Affiliation(s)
- Moritz Mirna
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Mario Holnthoner
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Albert Topf
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Peter Jirak
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Dzeneta Fejzic
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Vera Paar
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Jörg Kellermair
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Hermann Blessberger
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Christian Reiter
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Jürgen Kammler
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Paracelsus Medical University of Salzburg, Linz, Austria
| | - Lukas J Motloch
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Dusseldorf, Germany
| | - Daniel Kretzschmar
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
| | - Marcus Franz
- Department of Cardiology, Clinic of Internal Medicine I, Universitätsherzzentrum Thüringen, Friedrich Schiller University Jena, Jena, Germany
| | - Brunilda Alushi
- Department of General and Interventional Cardiology and Rhythmology, Helios Clinic, Erfurt, Germany
| | - Alexander Lauten
- Department of General and Interventional Cardiology and Rhythmology, Helios Clinic, Erfurt, Germany
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Clemens Steinwender
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Paracelsus Medical University of Salzburg, Linz, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| |
Collapse
|
12
|
Haley KE, Almas T, Shoar S, Shaikh S, Azhar M, Cheema FH, Hameed A. The role of anti-inflammatory drugs and nanoparticle-based drug delivery models in the management of ischemia-induced heart failure. Biomed Pharmacother 2021; 142:112014. [PMID: 34391184 DOI: 10.1016/j.biopha.2021.112014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 12/20/2022] Open
Abstract
Ongoing advancements in the treatment of acute myocardial infarction (MI) have significantly decreased MI related mortality. Consequently, the number of patients experiencing post-MI heart failure (HF) has continued to rise. Infarction size and the extent of left ventricular (LV) remodeling are largely determined by the extent of ischemia at the time of myocardial injury. In the setting of MI or acute phase of post-MI LV remodeling, anti-inflammatory drugs including intravenous immunoglobulin (IVIG) and Pentoxifylline have shown potential efficacy in preventing post-MI remodeling in-vitro and in some clinical trials. However, systemic administration of anti-inflammatory drugs are not without their off-target side effects. Herein, we explore the clinical feasibility of targeted myocardial delivery of anti-inflammatory drugs via biodegradable polymers, liposomes, hydrogels, and nano-particle based drug delivery models (NDDM) based on existing pre-clinical and clinical models. We summarize the barriers to clinical application of targeted anti-inflammatory delivery post-MI, including challenges in achieving sufficient retention and distribution, as well as the potential need for multiple dosing. Collectively, we suggest that localized delivery of anti-inflammatory agents to the myocardium using NDDM is a promising approach for successful treatment of ischemic HF. Future studies will be instrumental in determining the most effective target and delivery modalities for orchestrating NDDM-mediated treatment of HF.
Collapse
Affiliation(s)
- Kathryn E Haley
- Graduate Entry Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland
| | - Talal Almas
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; School of Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland
| | - Saeed Shoar
- HCA Healthcare Gulf Coast Division, Houston, TX, USA
| | - Shan Shaikh
- HCA Healthcare Gulf Coast Division, Houston, TX, USA
| | - Maimoona Azhar
- Graduate Entry Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Department of Surgery, St. Vincent's University Hospital, Dublin 4 Dublin, Ireland
| | - Faisal Habib Cheema
- HCA Healthcare Gulf Coast Division, Houston, TX, USA; University of Houston, College of Medicine, Houston, TX, USA
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 Dublin, Ireland; Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.
| |
Collapse
|
13
|
Opincariu D, Rodean I, Rat N, Hodas R, Benedek I, Benedek T. Systemic Vulnerability, as Expressed by I-CAM and MMP-9 at Presentation, Predicts One Year Outcomes in Patients with Acute Myocardial Infarction-Insights from the VIP Clinical Study. J Clin Med 2021; 10:jcm10153435. [PMID: 34362217 PMCID: PMC8347806 DOI: 10.3390/jcm10153435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/18/2022] Open
Abstract
(1) Background: The prediction of recurrent events after acute myocardial infarction (AMI) does not sufficiently integrate systemic inflammation, coronary morphology or ventricular function in prediction algorithms. We aimed to evaluate the accuracy of inflammatory biomarkers, in association with angiographical and echocardiographic parameters, in predicting 1-year MACE after revascularized AMI. (2) Methods: This is an extension of a biomarker sub-study of the VIP trial (NCT03606330), in which 225 AMI patients underwent analysis of systemic vulnerability and were followed for 1 year. Hs-CRP, MMP-9, IL-6, I-CAM, V-CAM and E-selectin were determined at 1 h after revascularization. The primary end-point was the 1-year MACE rate. (3) Results: The MACE rate was 24.8% (n = 56). There were no significant differences between groups in regard to IL-6, V-CAM and E-selectin. The following inflammatory markers were significantly higher in MACE patients: hs-CRP (11.1 ± 13.8 vs. 5.1 ± 4.4 mg/L, p = 0.03), I-CAM (452 ± 283 vs. 220.5 ± 104.6, p = 0.0003) and MMP-9 (2255 ± 1226 vs. 1099 ± 706.1 ng/mL p = 0.0001). The most powerful predictor for MACE was MMP-9 of >1155 ng/mL (AUC-0.786, p < 0.001) even after adjustments for diabetes, LVEF, acute phase complications and other inflammatory biomarkers. For STEMI, the most powerful predictors for MACE included I-CAM > 239.7 ng/mL, V-CAM > 877.9 ng/mL and MMP-9 > 1393 ng/mL. (4) Conclusions: High levels of I-CAM and MMP-9 were the most powerful predictors for recurrent events after AMI for the overall study population. For STEMI subjects, the most important predictors included increased levels of I-CAM, V-CAM and MMP-9, while none of the analyzed parameters had proven to be predictive. Inflammatory biomarkers assayed during the acute phase of AMI presented a more powerful predictive capacity for MACE than the LVEF.
Collapse
Affiliation(s)
- Diana Opincariu
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
- Correspondence: or (D.O.); or (R.H.); Tel.: +40-756-787-587 (D.O.); +40-742-385-600 (R.H.)
| | - Ioana Rodean
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
| | - Nora Rat
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| | - Roxana Hodas
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Correspondence: or (D.O.); or (R.H.); Tel.: +40-756-787-587 (D.O.); +40-742-385-600 (R.H.)
| | - Imre Benedek
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| | - Theodora Benedek
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| |
Collapse
|
14
|
Sun P, Wang Y, Ding Y, Luo J, Zhong J, Xu N, Zhang Y, Xie W. Canagliflozin attenuates lipotoxicity in cardiomyocytes and protects diabetic mouse hearts by inhibiting the mTOR/HIF-1α pathway. iScience 2021; 24:102521. [PMID: 34142035 PMCID: PMC8188479 DOI: 10.1016/j.isci.2021.102521] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/12/2021] [Accepted: 05/05/2021] [Indexed: 11/19/2022] Open
Abstract
Lipotoxicity plays an important role in the development of diabetic heart failure (HF). Canagliflozin (CAN), a marketed sodium-glucose co-transporter 2 inhibitor, has significantly beneficial effects on HF. In this study, we evaluated the protective effects and mechanism of CAN in the hearts of C57BL/6J mice induced by high-fat diet/streptozotocin (HFD/STZ) for 12 weeks in vivo and in HL-1 cells (a type of mouse cardiomyocyte line) induced by palmitic acid (PA) in vitro. The results showed that CAN significantly ameliorated heart functions and inflammatory responses in the hearts of the HFD/STZ-induced diabetic mice. CAN significantly attenuated the inflammatory injury induced by PA in the HL-1 cells. Furthermore, CAN seemed to bind to the mammalian target of rapamycin (mTOR) and then inhibit mTOR phosphorylation and hypoxia-inducible factor-1α (HIF-1α) expression. These results indicated that CAN might attenuate lipotoxicity in cardiomyocytes by inhibiting the mTOR/HIF-1α pathway and then show protective effects on diabetic hearts.
Collapse
Affiliation(s)
- Pengbo Sun
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- State Key Laboratory of Chemical Oncogenomic, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Key Lab in Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Yangyang Wang
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- State Key Laboratory of Chemical Oncogenomic, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Key Lab in Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Yipei Ding
- State Key Laboratory of Chemical Oncogenomic, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Key Lab in Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Jingyi Luo
- State Key Laboratory of Chemical Oncogenomic, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Key Lab in Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Jin Zhong
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- State Key Laboratory of Chemical Oncogenomic, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Key Lab in Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Naihan Xu
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- State Key Laboratory of Chemical Oncogenomic, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Key Lab in Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Yaou Zhang
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- State Key Laboratory of Chemical Oncogenomic, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Key Lab in Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Weidong Xie
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- State Key Laboratory of Chemical Oncogenomic, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Key Lab in Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Corresponding author
| |
Collapse
|
15
|
Dutka M, Bobiński R, Ulman-Włodarz I, Hajduga M, Bujok J, Pająk C, Ćwiertnia M. Various aspects of inflammation in heart failure. Heart Fail Rev 2021; 25:537-548. [PMID: 31705352 PMCID: PMC7181445 DOI: 10.1007/s10741-019-09875-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Despite significant advances in the prevention and treatment of heart failure (HF), the prognosis in patients who have been hospitalised on at least one occasion due to exacerbation of HF is still poor. Therefore, a better understanding of the underlying pathophysiological mechanisms of HF is crucial in order to achieve better results in the treatment of this clinical syndrome. One of the areas that, for years, has aroused the interest of researchers is the activation of the immune system and the elevated levels of biomarkers of inflammation in patients with both ischaemic and non-ischaemic HF. Additionally, it is intriguing that the level of circulating pro-inflammatory biomarkers correlates with the severity of the disease and prognosis in this group of patients. Unfortunately, clinical trials aimed at assessing interventions to modulate the inflammatory response in HF have been disappointing, and the modulation of the inflammatory response has had either no effect or even a negative effect on the HF prognosis. The article presents a summary of current knowledge on the role of immune system activation and inflammation in the pathogenesis of HF. Understanding the immunological mechanisms pathogenetically associated with left ventricular remodelling and progression of HF may open up new therapeutic possibilities for HF.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland.
| | - Rafał Bobiński
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Izabela Ulman-Włodarz
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Maciej Hajduga
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Jan Bujok
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Celina Pająk
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| | - Michał Ćwiertnia
- Faculty of Health Sciences, Department of Emergency Medicine, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biala, Poland
| |
Collapse
|
16
|
Gottschlich A, Endres S, Kobold S. Therapeutic Strategies for Targeting IL-1 in Cancer. Cancers (Basel) 2021; 13:477. [PMID: 33530653 PMCID: PMC7865618 DOI: 10.3390/cancers13030477] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/19/2022] Open
Abstract
Since its discovery, interleukin-1 has been extensively studied in a wide range of medical fields. Besides carrying out vital physiological functions, it has been implicated with a pivotal role in the progression and spreading of different cancer entities. During the last years, several clinical trials have been conducted, shedding light on the role of IL-1 blocking agents for the treatment of cancer. Additionally, recent developments in the field of immuno-oncology have implicated IL-1-induced signaling cascades as a major driver of severe chimeric antigen receptor T cell-associated toxicities such as cytokine release syndrome and immune effector cell-associated neurotoxicity. In this review, we summarize current clinical trials investigating the role of IL-1 blockade in cancer treatment and elaborate the proposed mechanism of these innovative treatment approaches. Additionally, we highlight cutting-edge developments utilizing IL-1 blocking agents to enhance the safety and efficacy of adoptive T cell therapy.
Collapse
Affiliation(s)
- Adrian Gottschlich
- Center for Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (A.G.); (S.E.)
| | - Stefan Endres
- Center for Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (A.G.); (S.E.)
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, 80337 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
| | - Sebastian Kobold
- Center for Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (A.G.); (S.E.)
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, 80337 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
| |
Collapse
|
17
|
Therapeutic Application of Exosomes in Inflammatory Diseases. Int J Mol Sci 2021; 22:ijms22031144. [PMID: 33498928 PMCID: PMC7865921 DOI: 10.3390/ijms22031144] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Immunomodulation is on the cusp of being an important therapy for treating many diseases, due to the significant role of the immune system in defending the human body. Although the immune system is an essential defense system, overactivity can result in diverse sicknesses such as inflammation and autoimmune disease. Exosomes are emerging as a state-of-the-art therapeutic strategy for treating an overactive immune system. Thus, in this review, we will thoroughly review therapeutic applications of exosomes in various inflammatory and autoimmune diseases. Finally, issues for an outlook to the future of exosomal therapy will be introduced.
Collapse
|
18
|
Daci A, Da Dalt L, Alaj R, Shurdhiqi S, Neziri B, Ferizi R, Danilo Norata G, Krasniqi S. Rivaroxaban improves vascular response in LPS-induced acute inflammation in experimental models. PLoS One 2020; 15:e0240669. [PMID: 33301454 PMCID: PMC7728205 DOI: 10.1371/journal.pone.0240669] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/30/2020] [Indexed: 12/18/2022] Open
Abstract
Rivaroxaban (RVX) was suggested to possess anti-inflammatory and vascular tone modulatory effects. The goal of this study was to investigate whether RVX impacts lipopolysaccharide (LPS)-induced acute vascular inflammatory response. Male rats were treated with 5 mg/kg RVX (oral gavage) followed by 10 mg/kg LPS i.p injection. Circulating levels of IL-6, MCP-1, VCAM-1, and ICAM-1 were measured in plasma 6 and 24 hours after LPS injection, while isolated aorta was used for gene expression analysis, immunohistochemistry, and vascular tone evaluation. RVX pre-treatment significantly reduced LPS mediated increase after 6h and 24h for IL-6 (4.4±2.2 and 2.8±1.7 fold), MCP-1 (1.4±1.5 and 1.3±1.4 fold) VCAM-1 (1.8±2.0 and 1.7±2.1 fold). A similar trend was observed in the aorta for iNOS (5.5±3.3 and 3.3±1.9 folds reduction, P<0.01 and P<0.001, respectively), VCAM-1 (1.3±1.2 and 1.4±1.3 fold reduction, P<0.05), and MCP-1 (3.9±2.2 and 1.9±1.6 fold reduction, P<0.01). Moreover, RVX pre-treatment, improved LPS-induced PE contractile dysfunction in aortic rings (Control vs LPS, Emax reduction = 35.4 and 31.19%, P<0.001; Control vs LPS+RVX, Emax reduction = 10.83 and 11.48%, P>0.05, respectively), resulting in 24.5% and 19.7% change in maximal constriction in LPS and LPS+RVX respectively. These data indicate that RVX pre-treatment attenuates LPS-induced acute vascular inflammation and contractile dysfunction.
Collapse
Affiliation(s)
- Armond Daci
- Department of Pharmacy, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Lorenzo Da Dalt
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Rame Alaj
- Cardiovascular Surgery Clinic, University Clinical Center of Kosovo, Prishtina, Kosovo
| | - Shpejtim Shurdhiqi
- Cardiovascular Surgery Clinic, University Clinical Center of Kosovo, Prishtina, Kosovo
| | - Burim Neziri
- Institute of Pathophysiology, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Rrahman Ferizi
- Department of Premedical Courses-Biology, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | - Giuseppe Danilo Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
- Centro SISA per lo Studio dell’Aterosclerosi, Ospedale Bassini, Cinisello Balsamo, Italy
| | - Shaip Krasniqi
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
- * E-mail:
| |
Collapse
|
19
|
Dutka M, Bobiński R, Ulman-Włodarz I, Hajduga M, Bujok J, Pająk C, Ćwiertnia M. Sodium glucose cotransporter 2 inhibitors: mechanisms of action in heart failure. Heart Fail Rev 2020; 26:603-622. [PMID: 33150520 PMCID: PMC8024236 DOI: 10.1007/s10741-020-10041-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
Diabetes is a key independent risk factor in the development of heart failure (HF) and a strong, adverse prognostic factor in HF patients. HF remains the primary cause of hospitalisation for diabetics and, as previous studies have shown, when HF occurs in these patients, intensive glycaemic control does not directly improve the prognosis. Recent clinical studies assessing a new class of antidiabetic drugs, sodium-glucose cotransporter 2 inhibitors (SGLT2is) showed some unexpected beneficial results. Patients treated with SGLT2is had a significant decrease in both cardiovascular (CV) and all-cause mortality and less hospitalisations due to HF compared to those given a placebo. These significant clinical benefits occurred quickly after the drugs were administered and were not solely due to improved glycaemic control. These groundbreaking clinical trials’ results have already changed clinical practice in the management of patients with diabetes at high CV risk. These trials have triggered numerous experimental studies aimed at explaining the mechanisms of action of this unique group of drugs. This article presents the current state of knowledge about the mechanisms of action of SGLT2is developed for the treatment of diabetes and which, thanks to their cardioprotective effects, may, in the future, become a treatment for patients with HF.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Rafał Bobiński
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Izabela Ulman-Włodarz
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Maciej Hajduga
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Jan Bujok
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Celina Pająk
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Michał Ćwiertnia
- Faculty of Health Sciences, Department of Emergency Medicine, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
20
|
Wunderer F, Traeger L, Sigurslid HH, Meybohm P, Bloch DB, Malhotra R. The role of hepcidin and iron homeostasis in atherosclerosis. Pharmacol Res 2020; 153:104664. [PMID: 31991168 PMCID: PMC7066581 DOI: 10.1016/j.phrs.2020.104664] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/20/2019] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
Atherosclerotic cardiovascular disease is a major burden on global health and a leading cause of death worldwide. The pathophysiology of this chronic disease is complex, involving inflammation, lipoprotein oxidation and accumulation, plaque formation, and calcification. In 1981, Dr. Jerome Sullivan formulated the 'Iron Hypothesis', suggesting that higher levels of stored iron promote cardiovascular diseases, whereas iron deficiency may have an atheroprotective effect. This hypothesis has stimulated research focused on clarifying the role of iron in the development of atherosclerosis. However, preclinical and clinical studies have produced contradictory results and the observation that patients with hemochromatosis do not appear to have an increased risk of atherosclerosis seemed incongruous with Sullivan's initial hypothesis. The 'paradox' of systemic iron overload not being accompanied by an increased risk for atherosclerosis led to a refinement of the iron hypothesis focusing on intracellular macrophage iron. More recent in vitro and animal studies have elucidated the complex signaling pathways regulating iron, with a particular focus on hepcidin, the master regulator of body iron homeostasis. Bone morphogenetic protein (BMP) signaling is the major pathway that is required for induction of hepcidin expression in response to increasing levels of iron. Strong links between iron homeostasis, BMP signaling, inflammation and atherosclerosis have been established in both mechanistic and human studies. This review summarizes the current understanding of the role of iron homeostasis and hepcidin in the development of atherosclerosis and discusses the BMP-hepcidin-ferroportin axis as a novel therapeutic target for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Florian Wunderer
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Lisa Traeger
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Haakon H. Sigurslid
- Cardiovascular Research Center and the Cardiology Division of the Department of medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Patrick Meybohm
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
- Department of Anaesthesiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Donald B. Bloch
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Rheumatology, Allergy and Immunology of the Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Rajeev Malhotra
- Cardiovascular Research Center and the Cardiology Division of the Department of medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
21
|
Zouein FA, Booz GW, Altara R. STAT3 and Endothelial Cell-Cardiomyocyte Dialog in Cardiac Remodeling. Front Cardiovasc Med 2019; 6:50. [PMID: 31069236 PMCID: PMC6491672 DOI: 10.3389/fcvm.2019.00050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/08/2019] [Indexed: 12/18/2022] Open
Abstract
This article presents an overview of the central role of STAT3 in the crosstalk between endothelial cells and cardiac myocytes in the heart. Endothelial cell STAT3 has a key role in inflammation that underlies cardiovascular disease and impacts on cardiac structure and function. STAT3 in endothelial cells contributes to adverse cardiomyocyte genetic reprograming, for instance, during peripartum cardiomyopathy. Conversely, cardiomyocyte STAT3 is important for maintaining endothelial cell function and capillary integrity with aging and hypertension. In addition, STAT3 serves as a sentinel for stress in the heart. Recent evidence has revealed that the redox nature of STAT3 is regulated, and STAT3 is responsive to oxidative stress (ischemia-reperfusion) so as to induce protective genes. At the level of the mitochondrion, STAT3 is important in regulating reactive oxygen species (ROS) formation, metabolism, and mitochondrial integrity. STAT3 may also control calcium release from the ER so as to limit its subsequent uptake by mitochondria and the induction of cell death. Under normal conditions, some STAT3 localizes to intercalated discs of cardiomyocytes and serves to transmit pro-fibrotic gene induction signals in the nucleus with increased blood pressure. Further research is needed to understand how the sentinel role of STAT3 in both endothelial cells and cardiomyocytes is integrated in order to coordinate the response of the heart to both physiological and pathological demands.
Collapse
Affiliation(s)
- Fouad A Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Raffaele Altara
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States.,Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research, Oslo, Norway
| |
Collapse
|
22
|
Mattina GF, Van Lieshout RJ, Steiner M. Inflammation, depression and cardiovascular disease in women: the role of the immune system across critical reproductive events. Ther Adv Cardiovasc Dis 2019; 13:1753944719851950. [PMID: 31144599 PMCID: PMC6545651 DOI: 10.1177/1753944719851950] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 04/29/2019] [Indexed: 12/18/2022] Open
Abstract
Women are at increased risk for developing depression and cardiovascular disease (CVD) across the lifespan and their comorbidity is associated with adverse outcomes that contribute significantly to rates of morbidity and mortality in women worldwide. Immune-system activity has been implicated in the etiology of both depression and CVD, but it is unclear how inflammation contributes to sex differences in this comorbidity. This narrative review provides an updated synthesis of research examining the association of inflammation with depression and CVD, and their comorbidity in women. Recent research provides evidence of pro-inflammatory states and sex differences associated with alterations in the hypothalamic-pituitary-adrenal axis, the renin-angiotensin-aldosterone system and the serotonin/kynurenine pathway, that likely contribute to the development of depression and CVD. Changes to inflammatory cytokines in relation to reproductive periods of hormonal fluctuation (i.e. the menstrual cycle, perinatal period and menopause) are highlighted and provide a greater understanding of the unique vulnerability women experience in developing both depressed mood and adverse cardiovascular events. Inflammatory biomarkers hold substantial promise when combined with a patient's reproductive and mental health history to aid in the prediction, identification and treatment of the women most at risk for CVD and depression. However, more research is needed to improve our understanding of the mechanisms underlying inflammation in relation to their comorbidity, and how these findings can be translated to improve women's health.
Collapse
Affiliation(s)
- Gabriella F. Mattina
- Neuroscience Graduate Program, McMaster University, 1280 Main Street West, ON L8S 4L8, Canada
| | - Ryan J. Van Lieshout
- Neuroscience Graduate Program, McMaster University, ON, Canada
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Meir Steiner
- Women’s Health Concerns Clinic, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|