1
|
Pramanda AN, Farabi F, Prameswari HS, Achmad C, Tiksnadi BB. Myocardial-alternation index (MMI) is correlated with soluble suppression of tumorigenecity-2 (sST2) in patients with ischemic cardiomyopathy. Egypt Heart J 2025; 77:39. [PMID: 40261549 PMCID: PMC12014887 DOI: 10.1186/s43044-025-00634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/23/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Ischemic cardiomyopathy is a condition that represents myocardial dysfunction due to obstructive coronary artery disease. In ischemic cardiomyopathy, both structural and electrical remodeling occur. Myocardial biomarker, soluble ST2 (sST2) is able to predict patient's mortality and morbidity, and structural remodeling of the heart is responsible for its expression. ECG dispersion mapping (ECG-DM) as evaluated by myocardial micro-alternation index (MMI) may predict alteration of the myocardial electrophysiology with high sensitivity and specificity. The association between structural and electrical remodeling in ischemic cardiomyopathy is not fully understood. This study aims to evaluate the correlation between MMI and sST2 level in patients with ischemic cardiomyopathy. RESULT Total patients who met for the inclusion criteria were 30 patients. Mean age was 57.97 ± 10.04 years; most patients were male (80%). 27 (90%) patients had class II NYHA functional class. The most common risk factors were smoking (20 (66,7%)) and hypertension (17 (56,7%)). Median MMI was 34.0% (IQR: 23.0-42.3%) and median sST2 was 5.6 ng/mL (IQR: 2.0-11.5 ng/mL). This study found that MMI had a significant correlation with sST2, indicating a link between structural and electrical remodeling in ischemic cardiomyopathy (r = 0.583, p < 0,05). CONCLUSION There was a correlation between MMI and sST2 in patients with ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Andra Naufal Pramanda
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Fatih Farabi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Hawani Sasmaya Prameswari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Chaerul Achmad
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Badai Bhatara Tiksnadi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia.
| |
Collapse
|
2
|
Feng Y, He LQ. Soluble ST2: A Novel Biomarker for Diagnosis and Prognosis of Cardiovascular Disease. Curr Med Sci 2024; 44:669-679. [PMID: 39096477 DOI: 10.1007/s11596-024-2907-x] [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: 12/14/2023] [Accepted: 05/30/2024] [Indexed: 08/05/2024]
Abstract
The increasing incidence of cardiovascular disease (CVD) is a significant global health concern, affecting millions of individuals each year. Accurate diagnosis of acute CVD poses a formidable challenge, as misdiagnosis can significantly decrease patient survival rates. Traditional biomarkers have played a vital role in the diagnosis and prognosis of CVDs, but they can be influenced by various factors, such as age, sex, and renal function. Soluble ST2 (sST2) is a novel biomarker that is closely associated with different CVDs. Its low reference change value makes it suitable for continuous measurement, unaffected by age, kidney function, and other confounding factors, facilitating risk stratification of CVDs. Furthermore, the combination of sST2 with other biomarkers can enhance diagnostic accuracy and prognostic value. This review aims to provide a comprehensive overview of sST2, focusing on its diagnostic and prognostic value as a myocardial marker for different types of CVDs and discussing the current limitations of sST2.
Collapse
Affiliation(s)
- Yin Feng
- Department of Cardiology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Qun He
- Department of Cardiology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Bedo D, Beaudrey T, Florens N. Unraveling Chronic Cardiovascular and Kidney Disorder through the Butterfly Effect. Diagnostics (Basel) 2024; 14:463. [PMID: 38472936 DOI: 10.3390/diagnostics14050463] [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: 01/15/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Chronic Cardiovascular and Kidney Disorder (CCKD) represents a growing challenge in healthcare, characterized by the complex interplay between heart and kidney diseases. This manuscript delves into the "butterfly effect" in CCKD, a phenomenon in which acute injuries in one organ lead to progressive dysfunction in the other. Through extensive review, we explore the pathophysiology underlying this effect, emphasizing the roles of acute kidney injury (AKI) and heart failure (HF) in exacerbating each other. We highlight emerging therapies, such as renin-angiotensin-aldosterone system (RAAS) inhibitors, SGLT2 inhibitors, and GLP1 agonists, that show promise in mitigating the progression of CCKD. Additionally, we discuss novel therapeutic targets, including Galectin-3 inhibition and IL33/ST2 pathway modulation, and their potential in altering the course of CCKD. Our comprehensive analysis underscores the importance of recognizing and treating the intertwined nature of cardiac and renal dysfunctions, paving the way for more effective management strategies for this multifaceted syndrome.
Collapse
Affiliation(s)
- Dimitri Bedo
- Nephrology Department, Hopitaux Universitaires de Strasbourg, F-67091 Strasbourg, France
- Faculté de Médecine, Université de Strasbourg, Team 3072 "Mitochondria, Oxidative Stress and Muscle Protection", Translational Medicine Federation of Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Thomas Beaudrey
- Nephrology Department, Hopitaux Universitaires de Strasbourg, F-67091 Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, F-67000 Strasbourg, France
| | - Nans Florens
- Nephrology Department, Hopitaux Universitaires de Strasbourg, F-67091 Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, F-67000 Strasbourg, France
| |
Collapse
|
4
|
Lu P, Wang T, Wan Z, Wang M, Zhou Y, He Z, Liao S, Liu H, Shu C. Immune-Related Genes and Immune Cell Infiltration Characterize the Maturation Status of Arteriovenous Fistulas: An Integrative Bioinformatics Study and Experimental Validation Based on Transcriptome Sequencing. J Inflamm Res 2024; 17:137-152. [PMID: 38223424 PMCID: PMC10785828 DOI: 10.2147/jir.s433525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/31/2023] [Indexed: 01/16/2024] Open
Abstract
Purpose Arteriovenous fistula (AVF) is the preferred vascular access for hemodialysis, but the low maturation rate is concerning. Immune cells' impact on AVF maturation lacks bioinformatics research. The study aims to investigate the potential predictive role of immune-related genes and immune cell infiltration characteristics in AVF maturation. Patients and Methods We analyzed the high-throughput sequencing dataset to identify differentially expressed genes (DEGs). Then, we performed enrichment analyses (GO, KEGG, GSEA) on immune-related genes and pathways in mature AVF. We focused on differentially expressed immune-related genes (DEIRGs) and constructed a PPI network to identify hub genes. These hub genes were validated in other databases and experiments, including qPCR and immunohistochemistry (IHC). The immune cell infiltration characteristics in native veins, failed AVFs, and matured AVFs were analyzed by cibersortX. Partial experimental validation was conducted using clinical samples. Results Our results showed that immune-related genes and signaling pathways are significantly enriched in mature AVF. We validated this in other databases and ultimately identified three hub genes (IL1B, IL6, CXCR4) in combination with experiments. Significant differences in immune cell infiltration characteristics were observed among native veins, failed AVFs, and matured AVFs. Immune cell infiltration analysis revealed that accumulation of CD4+ T cells, dendritic cells, mast cells and M2 macrophages contribute to AVF maturation. These immune-related genes and immune cells have the potential to serve as predictive factors for AVF maturation. We partially validated this experimentally. Conclusion From a bioinformatics perspective, our results have identified, for the first time, a set of immune-related genes and immune cell infiltration features that can characterize the maturation of AVF and significantly impact AVF maturation. These features hold potential as predictive indicators for AVF maturation outcomes.
Collapse
Affiliation(s)
- Peng Lu
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institute of Vascular Diseases, Central South University, Changsha, People’s Republic of China
| | - Tun Wang
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institute of Vascular Diseases, Central South University, Changsha, People’s Republic of China
| | - Zicheng Wan
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institute of Vascular Diseases, Central South University, Changsha, People’s Republic of China
| | - Mo Wang
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institute of Vascular Diseases, Central South University, Changsha, People’s Republic of China
| | - Yang Zhou
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institute of Vascular Diseases, Central South University, Changsha, People’s Republic of China
| | - Zhenyu He
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institute of Vascular Diseases, Central South University, Changsha, People’s Republic of China
| | - Sheng Liao
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institute of Vascular Diseases, Central South University, Changsha, People’s Republic of China
| | - Haiyang Liu
- Department of Geriatrics, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Chang Shu
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Institute of Vascular Diseases, Central South University, Changsha, People’s Republic of China
- Center of Vascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| |
Collapse
|
5
|
Lau C, Muthu ML, Siddiqui IF, Li L, Reinhardt DP. High-Fat Diet Has a Protective Sex-Dependent Effect on Aortic Aneurysm Severity in a Marfan Syndrome Mouse Model. Can J Cardiol 2023; 39:1553-1567. [PMID: 37482239 DOI: 10.1016/j.cjca.2023.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Marfan syndrome (MFS) is a genetic disorder caused by mutations in fibrillin-1 and is characterized by thoracic aortic aneurysms and other complications. Previous studies revealed sexual dimorphisms in formation of aortic aneurysm in patients with MFS. The current study aimed to investigate the combined role of a high-fat diet (HFD) and biological sex in aortic disease using the mgR/mgR MFS mouse model. METHODS Male and female mgR/mgR mice, as well as wild-type (WT) littermate mice, were fed a control diet (CD [10% fat]) or HFD (60% fat) from 4 to 12 weeks of age. Key aortic disease parameters analyzed included the diameter of the aortic wall; elastic fibre fragmentation; proteoglycan content; mRNA levels of Mmp12, Col1a1, Col3a1, and Fbn1; and fibrillin-1 deposition in the aortic wall. RESULTS HFD-fed female mgR/mgR mice had significantly reduced aortic diameters (35%), elastic fibre fragmentation (56%), pathologically enhanced proteoglycans (45%), and expression of Mmp12 (64%), Col1a1 (41%), and Col3a1 (43%) compared with male mgR/mgR mice on HFD. Fibrillin-1 deposition and Fbn1 mRNA levels were unaffected. The data reveal a protective effect of HFD in female mice. In contrast, CD did not exert any protective effects. CONCLUSIONS This study demonstrates a specific sexual dimorphism in MFS mice, with HFD exerting an explicit protective effect on severity of aortic disease in female mice. These preclinical data may be useful for developing nutritional recommendations for individuals with MFS in the longer term.
Collapse
Affiliation(s)
- Cori Lau
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Muthu L Muthu
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Iram Fatima Siddiqui
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Ling Li
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montréal, Québec, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
6
|
Soluble ST2 as a Potential Biomarker for Abdominal Aortic Aneurysms-A Single-Center Retrospective Cohort Study. Int J Mol Sci 2022; 23:ijms23179598. [PMID: 36076997 PMCID: PMC9455465 DOI: 10.3390/ijms23179598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
The maximal aortic diameter is the only clinically applied predictor of abdominal aortic aneurysm (AAA) progression and indicator for surgical repair. Circulating biomarkers resulting from AAA pathogenesis are attractive candidates for the diagnosis and prognosis of aneurysmal disease. Due to the reported role of interleukin 33 in AAA development, we investigated the corresponding circulating receptor molecules of soluble suppression of tumorigenesis 2 (sST2) in AAA patients regarding their marker potential in diagnosis and prognosis. We conducted a single-center retrospective cohort study in a diagnostic setting, measuring the circulating serum sST2 protein levels of 47 AAA patients under surveillance, matched with 25 peripheral artery disease (PAD) patients and 25 healthy controls. In a prognostic setting, we analyzed the longitudinal monitoring data of 50 monitored AAA patients. Slow versus fast AAA progression was defined as a <2 or ≥2 mm increase in AAA diameter over 6 months and a <4 or ≥4 mm increase over 12 months. Additionally, the association of circulating serum sST2 and AAA growth was investigated using a specifically tailored log-linear mixed model. Serum sST2 concentrations were significantly increased in AAA patients compared with healthy individuals: the median of AAA patient cohort was 112.72 ng/mL (p = 0.025) and that of AAA patient cohort 2 was 14.32 ng/mL (p = 0.039) versus healthy controls (8.82 ng/mL). Likewise, PAD patients showed significantly elevated sST2 protein levels compared with healthy controls (the median was 12.10 ng/mL; p = 0.048) but similar concentrations to AAA patients. Additionally, sST2 protein levels were found to be unsuited to identifying fast AAA progression over short-term periods of 6 or 12 months, which was confirmed by a log-linear mixed model. In conclusion, the significantly elevated protein levels of sST2 detected in patients with vascular disease may be useful in the early diagnosis of AAA but cannot distinguish between AAA and PAD or predict AAA progression.
Collapse
|
7
|
Evans BR, Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Döring Y. Inflammatory Mediators in Atherosclerotic Vascular Remodeling. Front Cardiovasc Med 2022; 9:868934. [PMID: 35600479 PMCID: PMC9114307 DOI: 10.3389/fcvm.2022.868934] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/11/2022] [Indexed: 12/23/2022] Open
Abstract
Atherosclerotic vascular disease remains the most common cause of ischemia, myocardial infarction, and stroke. Vascular function is determined by structural and functional properties of the arterial vessel wall, which consists of three layers, namely the adventitia, media, and intima. Key cells in shaping the vascular wall architecture and warranting proper vessel function are vascular smooth muscle cells in the arterial media and endothelial cells lining the intima. Pathological alterations of this vessel wall architecture called vascular remodeling can lead to insufficient vascular function and subsequent ischemia and organ damage. One major pathomechanism driving this detrimental vascular remodeling is atherosclerosis, which is initiated by endothelial dysfunction allowing the accumulation of intimal lipids and leukocytes. Inflammatory mediators such as cytokines, chemokines, and modified lipids further drive vascular remodeling ultimately leading to thrombus formation and/or vessel occlusion which can cause major cardiovascular events. Although it is clear that vascular wall remodeling is an elementary mechanism of atherosclerotic vascular disease, the diverse underlying pathomechanisms and its consequences are still insufficiently understood.
Collapse
Affiliation(s)
- Bryce R. Evans
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Anaïs Yerly
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Emiel P. C. van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- *Correspondence: Yvonne Döring
| |
Collapse
|
8
|
de Moraes Arnoso BJ, Magliaccio FM, de Araújo CA, de Andrade Soares R, Santos IB, de Bem GF, Fernandes-Santos C, Ognibene DT, de Moura RS, Resende AC, Daleprane JB, Costa CAD. Açaí seed extract (ASE) rich in proanthocyanidins improves cardiovascular remodeling by increasing antioxidant response in obese high-fat diet-fed mice. Chem Biol Interact 2022; 351:109721. [PMID: 34715092 DOI: 10.1016/j.cbi.2021.109721] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/24/2021] [Accepted: 10/23/2021] [Indexed: 02/06/2023]
Abstract
Obesity is recognized as an independent risk factor for cardiovascular diseases and is an important contributor to cardiac mortality. Açaí seed extract (ASE), rich in proanthocyanidins, has been shown to have potential anti-obesity effects. This study aimed to investigate the therapeutic effect of ASE in cardiovascular remodeling associated with obesity and compare it with that of rosuvastatin. Male C57BL/6 mice were fed a high-fat diet or a standard diet for 12 weeks. The ASE (300 mg/kg/day) and rosuvastatin (20 mg/kg/day) treatments started in the 8th week until the 12th week, totaling 4 weeks of treatment. Our data showed that treatment with ASE and rosuvastatin reduced body weight, ameliorated lipid profile, and improved cardiovascular remodeling. Treatment with ASE but not rosuvastatin reduced hyperglycemia and oxidative stress by reducing immunostaining of 8-isoprostane and increasing SOD-1 and GPx expression in HFD mice. ASE and rosuvastatin reduced NOX4 expression, increased SIRT-1 and Nrf2 expression and catalase and GPx activities, and improved vascular and cardiac remodeling in HFD mice. The therapeutic effect of ASE was similar to that of rosuvastatin in reducing dyslipidemia and cardiovascular remodeling but was superior in reducing oxidative damage and hyperglycemia, suggesting that ASE was a promising natural product for the treatment of cardiovascular alterations associated with obesity.
Collapse
Affiliation(s)
| | - Fabrizia Mansur Magliaccio
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Caroline Alves de Araújo
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Ricardo de Andrade Soares
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Izabelle Barcellos Santos
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Graziele Freitas de Bem
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Caroline Fernandes-Santos
- Department of Basic Sciences, Institute of Health, Fluminense Federal University, Nova Friburgo, RJ, Brazil
| | - Dayane Teixeira Ognibene
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Roberto Soares de Moura
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Angela Castro Resende
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Julio Beltrame Daleprane
- Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Cristiane Aguiar da Costa
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
9
|
Jiang W, Wang X, Gao P, Li F, Lu K, Tan X, Zheng S, Pei W, An M, Li X, Hu R, Zhong Y, Zhu J, Du J, Wang Y. Association of IL1R1 Coding Variant With Plasma-Level Soluble ST2 and Risk of Aortic Dissection. Front Cardiovasc Med 2021; 8:710425. [PMID: 34409081 PMCID: PMC8365023 DOI: 10.3389/fcvm.2021.710425] [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: 05/16/2021] [Accepted: 06/28/2021] [Indexed: 01/16/2023] Open
Abstract
Objective: Aortic dissection (AD) is characterized by an acute onset, rapid progress, and high mortality. Levels of soluble ST2 (sST2) on presentation are elevated in patients with acute AD, which can be used to discriminate AD patients from patients with chest pain. sST2 concentrations were found to be highly heritable in the general population. The aim of this study was to investigate the associations of variations in ST2-related gene expression with sST2 concentrations and AD risk. Methods: This case-control study involving a total of 2,277 participants were conducted, including 435 AD patients and age- and sex-matched 435 controls in the discovery stage, and 464 patients and 943 controls in the validation stage. Eight ST2-related genes were selected by systematic review. Tag single-nucleotide polymorphisms (SNPs) were screened out from the Chinese population of the 1,000 Genomes Database. Twenty-one ST2-related SNPs were genotyped, and plasma sST2 concentrations were measured. Results: In the discovery stage, rs13019803 located in IL1R1 was significantly associated with AD after Bonferroni correction (p = 0.0009) and was correlated with circulating sST2 levels in patients with type A AD(AAD) [log-sST2 per C allele increased by 0.180 (95%) CI: 0.002 - 0.357] but not in type B. Combining the two stages together, rs13019803C was associated with plasma sST2 level in AAD patients [log-sST2 increased by 0.141 (95% CI: 0.055-0.227) for per C allele]. Odds ratio of rs13019803 on the risk of AAD is 1.67 (95% CI: 1.33-2.09). Conclusions: The IL1R1 SNP rs13019803C is associated with higher sST2 levels and increased risk of AAD.
Collapse
Affiliation(s)
- Wenxi Jiang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Xue Wang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Pei Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Fengjuan Li
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Ke Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Xin Tan
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Shuai Zheng
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Wang Pei
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Meiyu An
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Xi Li
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Rong Hu
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yongliang Zhong
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junming Zhu
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jie Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Yuan Wang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, The Collaborative Innovation Center for Cardiovascular Disorders, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Department of Vascular Biology, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| |
Collapse
|
10
|
Interference of LPS H. pylori with IL-33-Driven Regeneration of Caviae porcellus Primary Gastric Epithelial Cells and Fibroblasts. Cells 2021; 10:cells10061385. [PMID: 34199843 PMCID: PMC8227243 DOI: 10.3390/cells10061385] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Lipopolysaccharide (LPS) of Helicobacter pylori (Hp) bacteria causes disintegration of gastric tissue cells in vitro. It has been suggested that interleukin (IL)-33 is involved in healing gastric injury. Aim: To elucidate whether Hp LPS affects regeneration of gastric barrier initiated by IL-33. Methods: Primary gastric epithelial cells or fibroblasts from Caviae porcellus were transfected with siRNA IL-33. Such cells, not exposed or treated with LPS Hp, were sub-cultured in the medium with or without exogenous IL-33. Then cell migration was assessed in conjunction with oxidative stress and apoptosis, activation of extracellular signal-regulated kinase (Erk), production of collagen I and soluble ST2 (IL-33 decoy). Results: Control cells not treated with LPS Hp migrated in the presence of IL-33. The pro-regenerative activity of IL-33 was related to stimulation of cells to collagen I production. Wound healing by cells exposed to LPS Hp was inhibited even in the presence of IL-33. This could be due to increased oxidative stress and apoptosis in conjunction with Erk activation, sST2 elevation and modulation of collagen I production. Conclusions: The recovery of gastric barrier cells during Hp infection potentially can be affected due to downregulation of pro-regenerative activity of IL-33 by LPS Hp.
Collapse
|
11
|
Oxidative Stress and Vascular Damage in the Context of Obesity: The Hidden Guest. Antioxidants (Basel) 2021; 10:antiox10030406. [PMID: 33800427 PMCID: PMC7999611 DOI: 10.3390/antiox10030406] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
The vascular system plays a central role in the transport of cells, oxygen and nutrients between different regions of the body, depending on the needs, as well as of metabolic waste products for their elimination. While the structure of different components of the vascular system varies, these structures, especially those of main arteries and arterioles, can be affected by the presence of different cardiovascular risk factors, including obesity. This vascular remodeling is mainly characterized by a thickening of the media layer as a consequence of changes in smooth muscle cells or excessive fibrosis accumulation. These vascular changes associated with obesity can trigger functional alterations, with endothelial dysfunction and vascular stiffness being especially common features of obese vessels. These changes can also lead to impaired tissue perfusion that may affect multiple tissues and organs. In this review, we focus on the role played by perivascular adipose tissue, the activation of the renin-angiotensin-aldosterone system and endoplasmic reticulum stress in the vascular dysfunction associated with obesity. In addition, the participation of oxidative stress in this vascular damage, which can be produced in the perivascular adipose tissue as well as in other components of the vascular wall, is updated.
Collapse
|
12
|
Matilla L, Arrieta V, Jover E, Garcia-Peña A, Martinez-Martinez E, Sadaba R, Alvarez V, Navarro A, Fernandez-Celis A, Gainza A, Santamaria E, Fernandez-Irigoyen J, Rossignol P, Zannad F, Lopez-Andres N. Soluble St2 Induces Cardiac Fibroblast Activation and Collagen Synthesis via Neuropilin-1. Cells 2020; 9:cells9071667. [PMID: 32664340 PMCID: PMC7408622 DOI: 10.3390/cells9071667] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/21/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
Circulating levels of soluble interleukin 1 receptor-like 1 (sST2) are increased in heart failure and associated with poor outcome, likely because of the activation of inflammation and fibrosis. We investigated the pathogenic role of sST2 as an inductor of cardiac fibroblasts activation and collagen synthesis. The effects of sST2 on human cardiac fibroblasts was assessed using proteomics and immunodetection approaches to evidence the upregulation of neuropilin-1 (NRP-1), a regulator of the profibrotic transforming growth factor (TGF)-β1. In parallel, sST2 increased fibroblast activation, collagen and fibrosis mediators. Pharmacological inhibition of nuclear factor-kappa B (NF-κB) restored NRP-1 levels and blocked profibrotic effects induced by sST2. In NRP-1 knockdown cells, sST2 failed to induce fibroblast activation and collagen synthesis. Exogenous NRP-1 enhanced cardiac fibroblast activation and collagen synthesis via NF-κB. In a pressure overload rat model, sST2 was elevated in association with cardiac fibrosis and was positively correlated with NRP-1 expression. Our study shows that sST2 induces human cardiac fibroblasts activation, as well as the synthesis of collagen and profibrotic molecules. These effects are mediated by NRP-1. The blockade of NF-κB restored NRP-1 expression, improving the profibrotic status induced by sST2. These results show a new pathogenic role for sST2 and its mediator, NRP-1, as cardiac fibroblast activators contributing to cardiac fibrosis.
Collapse
Affiliation(s)
- Lara Matilla
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Vanessa Arrieta
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Eva Jover
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Amaia Garcia-Peña
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Ernesto Martinez-Martinez
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
- Departamento de Fisiología, Facultad Medicina, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, 28040 Madrid, Spain
| | - Rafael Sadaba
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Virginia Alvarez
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Adela Navarro
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Amaya Fernandez-Celis
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Alicia Gainza
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
| | - Enrique Santamaria
- Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Institute for Health Research, Universidad Pública de Navarra, IdiSNA, 31008 Pamplona, Spain; (E.S.); (J.F.-I.)
| | - Joaquín Fernandez-Irigoyen
- Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Institute for Health Research, Universidad Pública de Navarra, IdiSNA, 31008 Pamplona, Spain; (E.S.); (J.F.-I.)
| | - Patrick Rossignol
- INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, 54035 Nancy, France; (P.R.); (F.Z.)
| | - Faiez Zannad
- INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, 54035 Nancy, France; (P.R.); (F.Z.)
| | - Natalia Lopez-Andres
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain; (L.M.); (V.A.); (E.J.); (A.G.-P.); (E.M.-M.); (R.S.); (V.A.); (A.N.); (A.F.-C.); (A.G.)
- INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT (Cardiovascular and Renal Clinical Trialists), Université de Lorraine, 54035 Nancy, France; (P.R.); (F.Z.)
- Correspondence: ; Tel.: +34-848428539; Fax: +34-848422300
| |
Collapse
|
13
|
Song B, Yao B, Dang H, Dong R. Soluble ST2, Galectin-3 and clinical prognosis of patients with hypertrophic cardiomyopathy undergoing ventricular septal myectomy: a correlation analysis. Cardiovasc Diagn Ther 2020; 10:145-152. [PMID: 32420094 DOI: 10.21037/cdt.2020.01.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Hypertrophic cardiomyopathy (HCM) is the most common chromosomal abnormal heart disease. The pathophysiological mechanism of HCM is complex. Several studies have suggested that the level of Soluble ST2 (sST2) may be a biomarker of chronic systolic heart failure, however, the role of sST2 in HCM remains unclear. So we performed this study to analyze the role of Soluble ST2 (sST2), Galectin-3 (Gal-3) and its correlations with clinical prognosis of patients with hypertrophic cardiomyopathy (HCM) undergoing ventricular septal myectomy. Methods HCM patients who underwent modified Morrow surgery in our hospital during June 2016-June 2018 were included. We divided the patients into different groups stratified by sST2 and Gal-3 level. Besides, we included volunteers without heart disease for medical examination as normal controls. Biochemical analyses were conducted to identify the biomarkers difference. The predictive value of sST2 and Gal-3 on all-cause mortality was evaluated with Cox regression analysis. Results A total of 125 HCM patients were included in this present study. The sST2 and Gal-3 levels in HCM patients were significantly higher than that in control group (all P<0.001); there were significant differences in the incidence of all-cause mortality for HCM patients stratified by the sST2 and Gal-3 level; Cox univariate regression survival analysis showed that the hypertension (HR =1.19, 95% CI: 1.01-1.38), maximum wall thickness (HR =1.48, 95% CI: 1.04-1.98), Log sST2 (HR =1.02, 95% CI: 1.01-1.05), Log Gal-3 (HR =1.17, 95% CI: 1.09-1.32) were the predictors for all-cause mortality in patients with HCM, and Cox multivariate risk regression showed that maximum wall thickness was the independent predictors of all-cause mortality in patients with HCM (HR =1.63, 95% CI: 1.35-1.97). Conclusions Even through sST2 and Gal-3 were not associated with clinical prognosis of patients with HCM undergoing ventricular septal myectomy, it may be involved in the progress of HCM, more studies are warranted to identify the potential mechanism and reverence value.
Collapse
Affiliation(s)
- Bangrong Song
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Bo Yao
- Cardiology Section 2, Zibo City Linzi District People's Hospital, Zibo 255400, China
| | - Haiming Dang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ran Dong
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
14
|
An HS, Lee JY, Choi EB, Jeong EA, Shin HJ, Kim KE, Park KA, Jin Z, Lee JE, Koh JS, Kwak W, Kim WH, Roh GS. Caloric restriction reverses left ventricular hypertrophy through the regulation of cardiac iron homeostasis in impaired leptin signaling mice. Sci Rep 2020; 10:7176. [PMID: 32346034 PMCID: PMC7188880 DOI: 10.1038/s41598-020-64201-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/13/2020] [Indexed: 12/18/2022] Open
Abstract
Leptin-deficient and leptin-resistant mice manifest obesity, insulin resistance, and left ventricular hypertrophy (LVH); however, LVH’s mechanisms are not fully understood. Cardiac iron dysregulation has been recently implicated in cardiomyopathy. Here we investigated the protective effects of caloric restriction on cardiac remodeling in impaired leptin signaling obese mice. RNA-seq analysis was performed to assess the differential gene expressions in the heart of wild-type and ob/ob mice. In particular, to investigate the roles of caloric restriction on iron homeostasis-related gene expressions, 10-week-old ob/ob and db/db mice were assigned to ad libitum or calorie-restricted diets for 12 weeks. Male ob/ob mice exhibited LVH, cardiac inflammation, and oxidative stress. Using RNA-seq analysis, we identified that an iron uptake-associated gene, transferrin receptor, was upregulated in obese ob/ob mice with LVH. Caloric restriction attenuated myocyte hypertrophy, cardiac inflammation, fibrosis, and oxidative stress in ob/ob and db/db mice. Furthermore, we found that caloric restriction reversed iron homeostasis-related lipocalin 2, divalent metal transporter 1, transferrin receptor, ferritin, ferroportin, and hepcidin expressions in the heart of ob/ob and db/db mice. These findings demonstrate that the cardioprotective effects of caloric restriction result from the cellular regulation of iron homeostasis, thereby decreasing oxidative stress, inflammation, and cardiac remodeling. We suggest that decreasing iron-mediated oxidative stress and inflammation offers new therapeutic approaches for obesity-induced cardiomyopathy.
Collapse
Affiliation(s)
- Hyeong Seok An
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Jong Youl Lee
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Eun Bee Choi
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Eun Ae Jeong
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Hyun Joo Shin
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Kyung Eun Kim
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Kyung-Ah Park
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Zhen Jin
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Jung Eun Lee
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Jin Sin Koh
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea
| | - Woori Kwak
- C&K genomics, Songpa-gu, Seoul, Republic of Korea
| | - Won-Ho Kim
- Division of Cardiovascular Diseases, Center for Biomedical Sciences, National Institute of Health, Cheongju, Chungbuk, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea. .,Bio Anti-aging Medical Research Center, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Gyeongnam, Republic of Korea.
| |
Collapse
|
15
|
da Silva-Bertani DCT, Vileigas DF, Mota GAF, de Souza SLB, Sant'Ana PG, Freire PP, de Tomasi LC, Corrêa CR, Padovani CR, Fernandes T, de Oliveira EM, Cicogna AC. Increased angiotensin II from adipose tissue modulates myocardial collagen I and III in obese rats. Life Sci 2020; 252:117650. [PMID: 32294475 DOI: 10.1016/j.lfs.2020.117650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023]
Abstract
It has been described that the cardiac dysfunction in the obesity model is because of collagen imbalance and that angiotensin II (Ang II) contributes to myocardial fibrosis. However, it remains undefined if changes in collagen I and III metabolism in obesity is due to the renin-angiotensin system (RAS) dysregulation from myocardium or excessive adipose tissue. AIM This study aimed to verify whether the changes in myocardial collagen metabolism result from RAS deregulation of cardiac or adipose tissue in an obesity model. MAIN METHODS Wistar rats were fed with control (CD) and high-fat (HFD) diets for 30 weeks. After the dietary intervention, animals were assigned to be treated with losartan at the 30 mg/kg/day dosage or kept untreated for an additional five weeks. KEY FINDINGS HFD induced obesity, comorbidities, and cardiac collagen overexpression. The HFD group presented an increase in Ang II levels in both adipose tissue and plasma, as well as AT1 receptor expression in cardiac tissue. Of note, the myocardial Ang II was not changed in the HFD group. Losartan administration reduced some obesity-induced comorbidities regardless of weight loss. The AT1 receptor blockade also decreased the release of Ang II from adipose tissue and myocardial AT1 receptor and collagen. SIGNIFICANCE It was seen that excessive adipose tissue is responsible for the exacerbated circulating Ang II, which induced cardiac fibrosis development.
Collapse
Affiliation(s)
| | - Danielle Fernandes Vileigas
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo 18618687, Brazil
| | - Gustavo Augusto Ferreira Mota
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo 18618687, Brazil
| | - Sérgio Luiz Borges de Souza
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo 18618687, Brazil
| | - Paula Grippa Sant'Ana
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo 18618687, Brazil
| | - Paula Paccielli Freire
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618970, Brazil
| | - Loreta Casquel de Tomasi
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo 18618687, Brazil
| | - Camila Renata Corrêa
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo 18618687, Brazil
| | - Carlos Roberto Padovani
- Department of Biostatistics, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618970, Brazil
| | - Tiago Fernandes
- Laboratory of Biochemistry and Molecular Biology of the Exercise, School of Physical Education and Sport, University of São Paulo (USP), 05508-900 São Paulo, Brazil
| | - Edilamar Menezes de Oliveira
- Laboratory of Biochemistry and Molecular Biology of the Exercise, School of Physical Education and Sport, University of São Paulo (USP), 05508-900 São Paulo, Brazil
| | - Antonio Carlos Cicogna
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo 18618687, Brazil.
| |
Collapse
|
16
|
Morello F, Bartalucci A, Bironzo M, Santoro M, Pivetta E, Ianniello A, Rumbolo F, Mengozzi G, Lupia E. Prospective diagnostic accuracy study of plasma soluble ST2 for diagnosis of acute aortic syndromes. Sci Rep 2020; 10:3103. [PMID: 32080259 PMCID: PMC7033105 DOI: 10.1038/s41598-020-59884-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 01/28/2020] [Indexed: 01/16/2023] Open
Abstract
Acute aortic syndromes (AASs) are difficult to diagnose emergencies. Plasma soluble ST2 (sST2), a prognostic biomarker for heart failure, has been proposed as a diagnostic biomarker of AASs outperforming D-dimer, the current diagnostic standard. We performed a prospective diagnostic accuracy study of sST2 for AASs in the Emergency Department (ED). In 2017–2018, patients were enrolled if they had ≥1 red-flag symptoms (chest/abdominal/back pain, syncope, perfusion deficit) and a clinical suspicion of AAS. sST2 was detected with the Presage® assay. Adjudication was based on computed tomography angiography (CTA) or on diagnostic outcome inclusive of 30-day follow-up. 297 patients were enrolled, including 88 with AASs. The median age was 67 years. In 162 patients with CTA, the median sST2 level was 41.7 ng/mL (IQR 29.4–103.2) in AASs and 34.6 ng/mL (IQR 21.4–51.5) in alternative diagnoses (P = 0.005). In ROC analysis, the AUC of sST2 was 0.63, as compared to 0.82 of D-dimer (P < 0.001). Sensitivity and specificity values of sST2 associated with different cutoffs were: 95.5% and 10.8% (≥12 ng/mL), 84.1% and 29.7% (≥23.7 ng/mL), 35.2% and 85.1% (≥66.5 ng/mL). Results were similar in the full cohort. In conclusion, in patients from a European ED, plasma sST2 provided modest accuracy for diagnosis of AASs.
Collapse
Affiliation(s)
- Fulvio Morello
- S.C.U. Medicina d'Urgenza, Molinette Hospital, A.O.U. Città della Salute e della Scienza, Torino, Italy. .,Dipartimento di Scienze Mediche, Università degli Studi di Torino, Torino, Italy.
| | - Alice Bartalucci
- S.C.U. Medicina d'Urgenza, Molinette Hospital, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Marco Bironzo
- S.C.U. Medicina d'Urgenza, Molinette Hospital, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Marco Santoro
- S.C.U. Medicina d'Urgenza, Molinette Hospital, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Emanuele Pivetta
- S.C.U. Medicina d'Urgenza, Molinette Hospital, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Alice Ianniello
- S.C. Biochimica Clinica, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Francesca Rumbolo
- S.C. Biochimica Clinica, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Giulio Mengozzi
- S.C. Biochimica Clinica, A.O.U. Città della Salute e della Scienza, Torino, Italy
| | - Enrico Lupia
- S.C.U. Medicina d'Urgenza, Molinette Hospital, A.O.U. Città della Salute e della Scienza, Torino, Italy.,Dipartimento di Scienze Mediche, Università degli Studi di Torino, Torino, Italy
| |
Collapse
|
17
|
Vianello E, Dozio E, Bandera F, Froldi M, Micaglio E, Lamont J, Tacchini L, Schmitz G, Corsi Romanelli MM. Correlative Study on Impaired Prostaglandin E2 Regulation in Epicardial Adipose Tissue and its Role in Maladaptive Cardiac Remodeling via EPAC2 and ST2 Signaling in Overweight Cardiovascular Disease Subjects. Int J Mol Sci 2020; 21:ijms21020520. [PMID: 31947646 PMCID: PMC7014202 DOI: 10.3390/ijms21020520] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/10/2020] [Accepted: 01/12/2020] [Indexed: 12/12/2022] Open
Abstract
There is recent evidence that the dysfunctional responses of a peculiar visceral fat deposit known as epicardial adipose tissue (EAT) can directly promote cardiac enlargement in the case of obesity. Here, we observed a newer molecular pattern associated with LV dysfunction mediated by prostaglandin E2 (PGE2) deregulation in EAT in a cardiovascular disease (CVD) population. A series of 33 overweight CVD males were enrolled and their EAT thickness, LV mass, and volumes were measured by echocardiography. Blood, plasma, EAT, and SAT biopsies were collected for molecular and proteomic assays. Our data show that PGE2 biosynthetic enzyme (PTGES-2) correlates with echocardiographic parameters of LV enlargement: LV diameters, LV end diastolic volume, and LV masses. Moreover, PTGES-2 is directly associated with EPAC2 gene (r = 0.70, p < 0.0001), known as a molecular inducer of ST2/IL-33 mediators involved in maladaptive heart remodelling. Furthermore, PGE2 receptor 3 (PTEGER3) results are downregulated and its expression is inversely associated with ST2/IL-33 expression. Contrarily, PGE2 receptor 4 (PTGER4) is upregulated in EAT and directly correlates with ST2 molecular expression. Our data suggest that excessive body fatness can shift the EAT transcriptome to a pro-tissue remodelling profile, may be driven by PGE2 deregulation, with consequent promotion of EPAC2 and ST2 signalling.
Collapse
Affiliation(s)
- Elena Vianello
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (E.D.); (F.B.); (L.T.); (M.M.C.R.)
- Correspondence: ; Tel.: +39-02-50315342
| | - Elena Dozio
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (E.D.); (F.B.); (L.T.); (M.M.C.R.)
| | - Francesco Bandera
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (E.D.); (F.B.); (L.T.); (M.M.C.R.)
- Cardiology University Department, Heart Failure Unit, IRCCS Policlinico San Donato, 20097 Milan, Italy
| | - Marco Froldi
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- Internal Medicine Unit IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Emanuele Micaglio
- U.O.C. SMEL-1 of Clinical Pathology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy;
| | - John Lamont
- Randox Laboratories LTD, R&D, Crumlin-Antrim, Belfast, BT29, Northen Ireland, UK
| | - Lorenza Tacchini
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (E.D.); (F.B.); (L.T.); (M.M.C.R.)
| | - Gerd Schmitz
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Massimiliano Marco Corsi Romanelli
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (E.D.); (F.B.); (L.T.); (M.M.C.R.)
- U.O.C. SMEL-1 of Clinical Pathology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy;
| |
Collapse
|
18
|
Lind L, Figarska S, Sundström J, Fall T, Ärnlöv J, Ingelsson E. Changes in Proteomic Profiles are Related to Changes in BMI and Fat Distribution During 10 Years of Aging. Obesity (Silver Spring) 2020; 28:178-186. [PMID: 31804015 PMCID: PMC6986305 DOI: 10.1002/oby.22660] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study investigated how changes in 84 proteins over a 10-year period of aging were related to changes in measures of body fat and distribution over the same period. METHODS Cardiovascular candidate proteins were measured using the proximal extension assay technique, along with BMI and waist-hip ratio (WHR), at ages 70, 75, and 80 in 1,016 participants of the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) cohort. Associations of changes in plasma protein levels, BMI, and WHR over time were analyzed using linear mixed models. RESULTS Changes in 19 and 16 proteins were significantly associated with changes in BMI and WHR, respectively (P < 0.00059), over the investigated 10-year period. Leptin and fatty acid-binding protein 4 were among the proteins most strongly associated with changes in both BMI and WHR. Four of the proteins significantly tracked with change in BMI (P < 0.00059) but not WHR (P > 0.05): endothelial cell-specific molecule 1, pentraxin-related protein PTX3, ST2 protein (also known as interleukin-1 receptor-like 1), and spondin-1. Five proteins tracked with change in WHR (P < 0.00059) but not BMI (P > 0.05): caspase-8, cathepsin L1, oxidized low-density lipoprotein receptor 1, interleukin-6 receptor subunit alpha, and C-C motif chemokine 20. CONCLUSIONS This is the first large longitudinal study of how changes in plasma protein signatures are associated with changes in measures of body fat and distribution over 10 years of aging.
Collapse
Affiliation(s)
- Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Sylwia Figarska
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA
- Stanford Diabetes Research Center, Stanford, CA 94305, USA
| | - Johan Sundström
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Ärnlöv
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- School of Health and Social Sciences, Dalarna University, Falun, Sweden
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA
- Stanford Diabetes Research Center, Stanford, CA 94305, USA
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
19
|
|
20
|
Soluble ST2 promotes oxidative stress and inflammation in cardiac fibroblasts: an in vitro and in vivo study in aortic stenosis. Clin Sci (Lond) 2019; 133:1537-1548. [DOI: 10.1042/cs20190475] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/08/2019] [Indexed: 11/17/2022]
Abstract
Abstract
Background: Soluble ST2 (interleukin 1 receptor-like 1) (sST2) is involved in inflammatory diseases and increased in heart failure (HF). We herein investigated sST2 effects on oxidative stress and inflammation in human cardiac fibroblasts and its pathological role in human aortic stenosis (AS).
Methods and results: Using proteomics and immunodetection approaches, we have identified that sST2 down-regulated mitofusin-1 (MFN-1), a protein involved in mitochondrial fusion, in human cardiac fibroblasts. In parallel, sST2 increased nitrotyrosine, protein oxidation and peroxide production. Moreover, sST2 enhanced the secretion of pro-inflammatory cytokines interleukin (IL)-6, IL-1β and monocyte chemoattractant protein-1 (CCL-2). Pharmacological inhibition of transcriptional factor nuclear factor κB (NFκB) restored MFN-1 levels and improved oxidative status and inflammation in cardiac fibroblasts. Mito-Tempo, a mitochondria-specific superoxide scavenger, as well as Resveratrol, a general antioxidant, attenuated oxidative stress and inflammation induced by sST2. In myocardial biopsies from 26 AS patients, sST2 up-regulation paralleled a decrease in MFN-1. Cardiac sST2 inversely correlated with MFN-1 levels and positively associated with IL-6 and CCL-2 in myocardial biopsies from AS patients.
Conclusions: sST2 affected mitochondrial fusion in human cardiac fibroblasts, increasing oxidative stress production and inflammatory markers secretion. The blockade of NFκB or mitochondrial reactive oxygen species restored MFN-1 expression, improving oxidative stress status and reducing inflammatory markers secretion. In human AS, cardiac sST2 levels associated with oxidative stress and inflammation. The present study reveals a new pathogenic pathway by which sST2 promotes oxidative stress and inflammation contributing to cardiac damage.
Collapse
|
21
|
Pusceddu I, Dieplinger B, Mueller T. ST2 and the ST2/IL-33 signalling pathway-biochemistry and pathophysiology in animal models and humans. Clin Chim Acta 2019; 495:493-500. [PMID: 31136737 DOI: 10.1016/j.cca.2019.05.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/26/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022]
Abstract
ST2 is an interleukin (IL)-1 receptor family member with transmembrane (ST2L) and soluble (sST2) isoforms. Structurally, the ST2 gene products are very similar in mice and humans. In humans and in mice, alternative promoter activation and splicing produce ST2L and sST2. ST2L represents the longest transcript, whereas sST2 is the truncated, soluble isoform. ST2L is the biological receptor for IL-33, a member of the IL-1 family. IL-33 is the functional ligand of ST2L and signals the presence of tissue damage to local immune cells. IL-33/ST2L signalling leads to the production of inflammatory cytokines/chemokines and to the induction of the immune response. Conversely, sST2 functions as a decoy receptor for IL-33, inhibiting the effects of IL-33/ST2L signalling. Animal studies have allowed the investigation of ST2 and the IL-33/ST2L signalling pathway at multiple levels. However, clinical studies have mainly focused on the determination of sST2 in the circulation. In humans, plasma concentrations of sST2 increase in several diseases, such as heart disease, pulmonary disease, burn injury and graft-versus-host disease. Consequently, increased plasma concentrations of sST2 are not specific for a single disorder in humans and are thus of limited value for diagnostic purposes. However, increased plasma concentrations of sST2 have been linked to a worse prognosis in numerous diseases. Nevertheless, the major source of circulating sST2 in healthy and diseased humans is currently not fully established. In addition, whether the downregulation of sST2 can improve the outcome of patients in the clinical setting has not been elucidated. The aim of the present review was to provide an update on the findings regarding the biochemistry and pathophysiology of ST2 and the sST2 signalling pathway in humans and experimental models.
Collapse
Affiliation(s)
- Irene Pusceddu
- Department of Clinical Pathology, Hospital of Bolzano, Bolzano, Italy
| | - Benjamin Dieplinger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz, Linz, Austria
| | - Thomas Mueller
- Department of Clinical Pathology, Hospital of Bolzano, Bolzano, Italy.
| |
Collapse
|
22
|
Kim HL, Lee JP, Lim WH, Seo JB, Zo JH, Kim MA, Kim SH. Association between the level of serum soluble ST2 and invasively measured aortic pulse pressure in patients undergoing coronary angiography. Medicine (Baltimore) 2019; 98:e14215. [PMID: 30813128 PMCID: PMC6407930 DOI: 10.1097/md.0000000000014215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Despite the well-documented value of ST2 in heart failure and myocardial infarction, the role of ST2 in vascular biology has not yet been well defined. This study was performed to investigate the association between serum soluble ST2 (sST2) and invasively measured aortic pulse pressure (APP). A total of 167 consecutive patients with suspected coronary artery disease (CAD) (65.1 ± 9.8 years; men, 65.9%) referred for invasive coronary angiography was prospectively enrolled. APP was measured at the ascending aorta with a pig-tail catheter, and arterial blood samples for the measurement of sST2 were collected before coronary angiography. Serum sST2 levels were quantified by radioimmunoassay. Most of the patients (73.9%) had significant CAD (stenosis ≥ 50%) on coronary angiography. Patients with higher APP (≥76 mmHg) showed a significantly higher sST2 level compared to those with lower APP (<76 mmHg) (31.7 ± 13.9 ng/mL vs 26.2 ± 10.2 ng/mL, P < .001). In simple correlation analysis, there was a significant positive correlation between sST2 levels and APP (r = 0.413, P < .001). In multiple linear regression analysis, sST2 had an independent association with APP even after controlling for potential confounders (β = 0.331, P < .001). The serum sST2 level may be independently associated with invasively measured APP in patients undergoing coronary angiography. The result of this study gives insight into the role of sST2 in aortic stiffening, and suggests that the sST2 level may be a useful marker of aortic stiffness.
Collapse
Affiliation(s)
| | - Jung Pyo Lee
- Division of Nephrology, Department of Internal Medicine, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Woo-Hyun Lim
- Division of Cardiology, Department of Internal Medicine
| | - Jae-Bin Seo
- Division of Cardiology, Department of Internal Medicine
| | - Joo-Hee Zo
- Division of Cardiology, Department of Internal Medicine
| | - Myung-A Kim
- Division of Cardiology, Department of Internal Medicine
| | - Sang-Hyun Kim
- Division of Cardiology, Department of Internal Medicine
| |
Collapse
|
23
|
Wang X, Shao X, Liu X, Qin Q, Xu J, Zhang JA. Dysregulated Interleukin -33/ST2 Pathway Perpetuates Chronic Inflammation in Hashimoto's Thyroiditis. Endocr Metab Immune Disord Drug Targets 2019; 19:1012-1021. [PMID: 30819087 DOI: 10.2174/1871530319666190226164309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Hashimoto's Thyroiditis (HT) is an autoimmune disease, characterized by chronic inflammation of the thyroid gland with unknown etiologies. Recently, interleukin-33/ST2 (IL- 33/ST2) pathway reveals its participation in the process of several autoimmune diseases. In this study, the role of IL-33/ST2 pathway in the development of HT is investigated. METHODS The levels of plasma IL-33, sST2 and the frequency of circulating CD4+ST2L+T cells in 30 HT patients and 20 healthy controls were determined by enzyme-linked immunosorbent assay (ELISA) and flow cytometry respectively. The mRNA expressions of related molecules in IL-33/ST2 pathway in thyroid tissues (12 HT patients and 10 controls) were detected by real-time quantitative PCR (RTqPCR). The protein expressions of IL-33 and ST2 were determined by Western blot and immunohistochemistry staining. RESULTS The mRNA expressions of plasma IL-33 and sST2 were elevated in HT patients, with an increased ratio of IL-33/sST2. The number of CD4+ST2L+ T cells in PBMCs of HT group was significantly increased when compared to the control group (CON) by Flow cytometry assay. MRNA Expression of IL-33 and ST2 in thyroid tissue and the level of IL-1β and IL-18 were significantly upregulated in HT patients, while IL-5 was down-regulated in HT patients, compared to CON. The expression of IL-1β and IL-18 were positively correlated with the expression of IL-33. Results of western blot and immunohistochemical staining were consistent with qPCR. CONCLUSION IL-33/ST2 pathway participates in HT via affecting the production of inflammatory cytokines.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Xiaoqing Shao
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xinhao Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiu Qin
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Jian Xu
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Jin A Zhang
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
24
|
Italiani P, Puxeddu I, Napoletano S, Scala E, Melillo D, Manocchio S, Angiolillo A, Migliorini P, Boraschi D, Vitale E, Di Costanzo A. Circulating levels of IL-1 family cytokines and receptors in Alzheimer's disease: new markers of disease progression? J Neuroinflammation 2018; 15:342. [PMID: 30541566 PMCID: PMC6292179 DOI: 10.1186/s12974-018-1376-1] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/22/2018] [Indexed: 01/20/2023] Open
Abstract
Background Although the mechanisms underlying AD neurodegeneration are not fully understood, it is now recognised that inflammation could play a crucial role in the initiation and progression of AD neurodegeneration. A neuro-inflammatory network, based on the anomalous activation of microglial cells, includes the production of a number of inflammatory cytokines both locally and systemically. These may serve as diagnostic markers or therapeutic targets for AD neurodegeneration. Methods We have measured the levels of the inflammation-related cytokines and receptors of the IL-1 family in serum of subjects with AD, compared to mild cognitive impairment (MCI), subjective memory complaints (SMC), and normal healthy subjects (NHS). Using a custom-made multiplex ELISA array, we examined ten factors of the IL-1 family, the inflammation-related cytokines IL-1α, IL-1β, IL-18, and IL-33, the natural inhibitors IL-1Ra and IL-18BP, and the soluble receptors sIL-1R1, sIL-1R2, sIL-1R3, and sIL-1R4. Results The inflammatory cytokines IL-1α and IL-1β, their antagonist IL-1Ra, and their soluble receptor sIL-1R1 were increased in AD. The decoy IL-1 receptor sIL-1R2 was only increased in MCI. IL-33 and its soluble receptor sIL-1R4 were also significantly higher in AD. The soluble form of the accessory receptor for both IL-1 and IL-33 receptor complexes, sIL-1R3, was increased in SMC and even more in AD. Total IL-18 levels were unchanged, whereas the inhibitor IL-18BP was significantly reduced in MCI and SMC, and highly increased in AD. The levels of free IL-18 were significantly higher in MCI. Conclusions AD is characterised by a significant alteration in the circulating levels of the cytokines and receptors of the IL-1 family. The elevation of sIL-1R4 in AD is in agreement with findings in other diseases and can be considered a marker of ongoing inflammation. Increased levels of IL-1Ra, sIL-1R1, sIL-1R4, and IL-18BP distinguished AD from MCI and SMC, and from other inflammatory diseases. Importantly, sIL-1R1, sIL-1R3, sIL-1R4, and IL-18BP negatively correlated with cognitive impairment. A significant elevation of circulating sIL-1R2 and free IL-18, not present in SMC, is characteristic of MCI and disappears in AD, making them additional interesting markers for evaluating progression from MCI to AD. Electronic supplementary material The online version of this article (10.1186/s12974-018-1376-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paola Italiani
- Laboratory of Innate Immunity and Inflammation, Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Ilaria Puxeddu
- Clinical Immunology Unit, Department Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Sabrina Napoletano
- NeurOmics Laboratory, Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Emanuele Scala
- Laboratory of Innate Immunity and Inflammation, Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Daniela Melillo
- Laboratory of Innate Immunity and Inflammation, Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Simone Manocchio
- Centre for Research and Training in Medicine for Aging, Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Località Tappino, 86100, Campobasso, Italy
| | - Antonella Angiolillo
- Centre for Research and Training in Medicine for Aging, Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Località Tappino, 86100, Campobasso, Italy
| | - Paola Migliorini
- Clinical Immunology Unit, Department Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Diana Boraschi
- Laboratory of Innate Immunity and Inflammation, Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy.
| | - Emilia Vitale
- NeurOmics Laboratory, Institute of Protein Biochemistry, National Research Council, Via Pietro Castellino 111, 80131, Naples, Italy.
| | - Alfonso Di Costanzo
- Centre for Research and Training in Medicine for Aging, Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Località Tappino, 86100, Campobasso, Italy
| |
Collapse
|
25
|
Altara R, Ghali R, Mallat Z, Cataliotti A, Booz GW, Zouein FA. Conflicting vascular and metabolic impact of the IL-33/sST2 axis. Cardiovasc Res 2018; 114:1578-1594. [PMID: 29982301 DOI: 10.1093/cvr/cvy166] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/28/2018] [Indexed: 02/15/2024] Open
Abstract
Interleukin 33 (IL-33), which is expressed by several immune cell types, endothelial and epithelial cells, and fibroblasts, is a cytokine of the IL-1 family that acts both intra- and extracellularly to either enhance or resolve the inflammatory response. Intracellular IL-33 acts in the nucleus as a regulator of transcription. Once released from cells by mechanical stress, inflammatory cytokines, or necrosis, extracellular IL-33 is proteolytically processed to act in an autocrine/paracrine manner as an 'alarmin' on neighbouring or various immune cells expressing the ST2 receptor. Thus, IL-33 may serve an important role in tissue preservation and repair in response to injury; however, the actions of IL-33 are dampened by a soluble form of ST2 (sST2) that acts as a decoy receptor and is produced by endothelial and certain immune cells. Accumulating evidence supports the conclusion that sST2 is a biomarker of vascular health with diagnostic and/or prognostic value in various cardiovascular diseases, including coronary artery disease, myocardial infarction, atherosclerosis, giant-cell arteritis, acute aortic dissection, and ischaemic stroke, as well as obesity and diabetes. Although sST2 levels are positively associated with cardiovascular disease severity, the assumption that IL-33 is always beneficial is naïve. It is increasingly appreciated that the pathophysiological importance of IL-33 is highly dependent on cellular and temporal expression. Although IL-33 is atheroprotective and may prevent obesity and type 2 diabetes by regulating lipid metabolism, IL-33 appears to drive endothelial inflammation. Here, we review the current knowledge of the IL-33/ST2/sST2 signalling network and discuss its pathophysiological and translational implications in cardiovascular diseases.
Collapse
Affiliation(s)
- Raffaele Altara
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Building 7, 4th floor, Kirkeveien 166, Oslo, Norway
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | - Rana Ghali
- Department of Pharmacology and Toxicology, American University of Beirut & Medical Center, Faculty of Medicine, Riad El-Solh, Beirut-Lebanon
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
- Institut National de la Sante et de la Recherche Medicale (Inserm), Unit 970, Paris Cardiovascular Research Center, Paris, France
| | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Building 7, 4th floor, Kirkeveien 166, Oslo, Norway
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, 2500 North State St., Jackson, MS, USA
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, American University of Beirut & Medical Center, Faculty of Medicine, Riad El-Solh, Beirut-Lebanon
| |
Collapse
|
26
|
Offor O, Utay N, Reynoso D, Somasunderam A, Currier J, Lake J. Adiponectin and the steatosis marker Chi3L1 decrease following switch to raltegravir compared to continued PI/NNRTI-based antiretroviral therapy. PLoS One 2018; 13:e0196395. [PMID: 29746485 PMCID: PMC5944924 DOI: 10.1371/journal.pone.0196395] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/12/2018] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND People with HIV are at for metabolic syndrome (MetS) and fatty liver disease, but the role of Antiretroviral therapy (ART) is poorly understood. MetS and fatty liver disease been associated with changes in adiponectin, soluble ST2 (sST2), chitinase 3-like 1 (Chi3L1), hyaluronic acid (HA), tissue inhibitor of metalloproteinase-1 (TIMP-1), lysyl oxidase-like-2 (LOXL2) and transforming growth factor β (TGF-β) concentrations in HIV-uninfected populations. Protease (PI) and non-nucleoside reverse transcriptase inhibitors (NNRTI) may contribute to these comorbidities, but the effects of switching from PI- or NNRTI to raltegravir (RAL) on these biomarkers is unknown. METHODS Cryopreserved plasma was obtained from a completed, prospective trial of HIV-infected women with central adiposity on NNRTI- or PI-based ART during which they were randomized to remain on their current ART or switch to a RAL based regimen. Biomarker concentrations were quantified using ELISA and Multiplex assays at baseline and 24 weeks after randomization. Wilcoxon-signed rank test evaluated within-group changes, Spearman and linear regression models evaluated correlations between biomarkers and clinical covariates. RESULTS Participants had a median age of 43 years, CD4+ T lymphocyte count 558 cells/mm3 and BMI 32 kg/m2; 35% met criteria for MetS. At baseline, higher adiponectin levels correlated with higher Chi3L1 levels (r = 0.42, p = 0.02), as did declines after 24 weeks (r = 0.40, p = 0.03). Changes in sST2 correlated with changes in Chi3L1 (r = 0.43, p = 0.02) and adiponectin (r = 0.40, p = 0.03). Adiponectin and Chi3L1 levels decreased significantly in women switched to RAL vs continue PI/NNRTI. CONCLUSION In women with HIV and central obesity, the hepatic steatosis/fibrosis marker Chi3L1 and adiponectin decrease in conjunction with sST2 decreases following switch to RAL. Whether switching from NNRTI/PI-based regimens to RAL can improve hepatic steatosis and dysmetabolism requires further study. TRIAL REGISTRATION Clinicaltrials.gov NCT00656175.
Collapse
Affiliation(s)
- Obiageli Offor
- Department of Epidemiology, University of Texas Health Science Center Houston, Texas, United States of America
| | - Netanya Utay
- Department of Internal Medicine, University of Texas Health Science Center Houston, Texas, United States of America
| | - David Reynoso
- Department of Infectious Disease, University of Texas Medical Branch Galveston, Texas, United States of America
| | - Anoma Somasunderam
- Department of Internal Medicine, University of Texas Health Science Center Houston, Texas, United States of America
| | - Judith Currier
- Department of Infectious Disease, University of California Los Angeles, California, United States of America
| | - Jordan Lake
- Department of Infectious Disease, University of Texas Health Science Center Houston, Texas, United States of America
| |
Collapse
|
27
|
Capecchi R, Italiani P, Puxeddu I, Pratesi F, Tavoni A, Boraschi D, Migliorini P. IL-1 family cytokines and receptors in IgG4-related disease. Cytokine 2018; 102:145-148. [DOI: 10.1016/j.cyto.2017.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/28/2017] [Accepted: 08/02/2017] [Indexed: 12/24/2022]
|
28
|
Dichotomous function of IL-33 in health and disease: From biology to clinical implications. Biochem Pharmacol 2018; 148:238-252. [PMID: 29309756 DOI: 10.1016/j.bcp.2018.01.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022]
Abstract
Interleukin (IL)-33 is a cytokine that is released from epithelial and endothelial cells at barrier surfaces upon tissue stress or damage to operate as an alarmin. IL-33 has been primarily implicated in the induction of T helper (Th) 2 type immune responses. Therefore, IL-33 has attracted a lot of interest as a potential therapeutic target in asthma and other allergic diseases. Over the years, it has become clear that IL-33 has a much broader activity and also contributes to Th1 immunity, expanding the possibilities for therapeutic modulation of IL-33 activity to multiple inflammatory diseases. However, more recently IL-33 has also been shown to mediate immunosuppression and tissue repair by activating regulatory T cells (Treg) and promoting M2 macrophage polarization. These pleiotropic activities of IL-33 illustrate the need for a tight molecular regulation of IL-33 activity, and have to be taken into account when IL-33 or its receptor is targeted for therapeutic modulation. Here we review the multiple molecular mechanisms that regulate IL-33 activity and describe how IL-33 can shape innate and adaptive immune responses by promoting Th1, Th2 and Treg function. Finally, we will discuss the possibilities for therapeutic modulation of IL-33 signaling as well as possible safety issues.
Collapse
|
29
|
Chen J, Wang S, Luo M, Zhang Z, Dai X, Kong M, Cai L, Wang Y, Shi B, Tan Y. From the Cover: Zinc Deficiency Worsens and Supplementation Prevents High-Fat Diet Induced Vascular Inflammation, Oxidative Stress, and Pathological Remodeling. Toxicol Sci 2016; 153:124-136. [PMID: 27370414 DOI: 10.1093/toxsci/kfw110] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2024] Open
Abstract
Obesity has become a common public health problem in the world and raises the risk of various cardiovascular diseases. Zinc is essential for multiple organs in terms of normal structure and function. The present study investigated the effects of high fat diet (HFD) induced obesity on the aorta in mice, and evaluated whether it can be affected by zinc deficiency or supplementation. Four-week-old male C57BL/6J mice were fed HFD with varied amounts of zinc (deficiency, adequate and supplementation) for 3 and 6 months. Results showed that HFD feeding induced a time-dependent aortic remodeling, demonstrated by increased vessel wall thickness, tunica cell proliferation and fibrotic responses, and inflammatory response, reflected by increased expression of inflammatory cytokines (tumor necrosis factor-α and vascular cell adhesion molecule 1). HFD feeding also caused aortic oxidative damage, reflected by 3-nitrotyrosine and 4-hydroxy-2-nonenal accumulation, and down-regulated nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression and function, shown by down-regulation of its downstream antioxidants, catalase, NAD(P)H dehydrogenase (quinone 1), and metallothionein expression. The vascular effects of obesity-induced by HFD was exacerbated by zinc deficiency but significantly improved by zinc supplementation. In addition, down-regulation of Nrf2 function and associated antioxidants expression were also worsened by zinc deficiency but improved by zinc supplementation. These results suggest that HFD induces aortic remodeling, which can be exacerbated by zinc deficiency and improved by zinc supplementation.
Collapse
Affiliation(s)
- Jun Chen
- *Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an 710061, China Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and School of Nursing, Wenzhou Medical University, Wenzhou 325035, China Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky 40202
| | - Shudong Wang
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky 40202 Center of Cardiovascular Disease, The First Hospital of Jilin University, Changchun 130000, China
| | - Manyu Luo
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky 40202 Department of Nephrology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Zhiguo Zhang
- Center of Cardiovascular Disease, The First Hospital of Jilin University, Changchun 130000, China
| | - Xiaozhen Dai
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky 40202 School of Biomedicine, Chengdu Medical College, Chengdu 610500, China
| | - Maiying Kong
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, Kentucky 40202
| | - Lu Cai
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and School of Nursing, Wenzhou Medical University, Wenzhou 325035, China Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky 40202
| | - Yuehui Wang
- Department of Geriatric Medicine, The First Hospital of Jilin University, Changchun 130000, China
| | - Bingyin Shi
- *Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an 710061, China
| | - Yi Tan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences and School of Nursing, Wenzhou Medical University, Wenzhou 325035, China Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, Kentucky 40202
| |
Collapse
|
30
|
Tashiro H, Takahashi K, Hayashi S, Kato G, Kurata K, Kimura S, Sueoka-Aragane N. Interleukin-33 from Monocytes Recruited to the Lung Contributes to House Dust Mite-Induced Airway Inflammation in a Mouse Model. PLoS One 2016; 11:e0157571. [PMID: 27310495 PMCID: PMC4910993 DOI: 10.1371/journal.pone.0157571] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 06/01/2016] [Indexed: 12/24/2022] Open
Abstract
Background Interleukin-33 (IL-33) activates group 2 innate lymphoid cells (ILC2), resulting in T-helper-2 inflammation in bronchial asthma. Airway epithelial cells were reported as sources of IL-33 during apoptosis and necrosis. However, IL-33 is known to be from sources other than airway epithelial cells such as leukocytes, and the mechanisms of IL-33 production and release are not fully understood. The aim of this study was to clarify the role of IL-33 production by monocytes in airway inflammation. Methods BALB/c mice were sensitized and challenged with a house dust mite (HDM) preparation. Airway inflammation was assessed by quantifying inflammatory cells in bronchoalveolar lavage (BAL) fluid, and IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) levels in lung. Immunohistochemistry for IL-33 in lung sections was also performed. Ly6c, CD11b, and CD11c expression was examined by flow cytometry. Clodronate liposomes were used in the HDM-airway inflammation model to deplete circulating monocytes. Results The IL-33, but not IL-25 or TSLP, level in lung homogenates was markedly increased in HDM mice compared to control mice. IL-33-positive cells in the lungs were identified using immunohistochemistry and were increased in areas surrounding bronchi and vasculature. Furthermore, IL-33 levels were increased in mononuclear cells derived from lungs of HDM mice compared to controls. The expression of Ly6c in mononuclear cells was significantly higher in HDM mice than in controls. Treatment with clodronate liposomes led to inhibition of not only inflammatory cells in BAL fluid, airway hyper reactivity and Th2 cytokines in lung, but also IL-33 in lung. Conclusion IL-33 from monocytes recruited to the lung may contribute to the pathogenesis of HDM-induced airway inflammation.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
- * E-mail:
| | - Shinichiro Hayashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Go Kato
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Keigo Kurata
- Institute of Tokyo Environmental Allergy, Tokyo, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
31
|
Sun M, Ma HF, Che YY, Cui X. Reduced IL-33 plasma levels in aplastic anemia. Cancer Cell Int 2015; 15:118. [PMID: 26677348 PMCID: PMC4681025 DOI: 10.1186/s12935-015-0270-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 12/04/2015] [Indexed: 11/17/2022] Open
Abstract
Background In this study, we aim to evaluate the balance of interleukin (IL)-33 and its soluble receptor sST2 in patients with aplastic anemia (AA). Methods Plasma IL-33, IL-17 and sST2 levels were measured in patients with active AA (n = 31), AA in remission (n = 29) and in healthy subjects (n = 30), using enzyme linked immunosorbent assays (ELISAs). Results The results showed that sST2 and IL-17 levels were significantly elevated in patients with active AA when compared to control subjects, but IL-33 levels were significantly lower in AA patients, which resulted in elevated sST2/IL-33 ratios in patients with active disease. During remission stages, the levels of these cytokines were comparable to those of healthy controls. Conclusions The elevated levels of sST2/IL-33 in the plasma during active stages of the disease suggest a possible role in the pathogenesis and course of AA.
Collapse
Affiliation(s)
- Ming Sun
- Department of Hematology, Linzi District People's Hospital, Zibo, 255400 China
| | - Hai-Feng Ma
- Department of Hematology, Linzi District People's Hospital, Zibo, 255400 China
| | - Ye-Yun Che
- Department of Hematology, Linzi District People's Hospital, Zibo, 255400 China
| | - Xin Cui
- Department of Internal Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021 China
| |
Collapse
|
32
|
Wagner A, Köhm M, Nordin A, Svenungsson E, Pfeilschifter JM, Radeke HH. Increased Serum Levels of the IL-33 Neutralizing sST2 in Limited Cutaneous Systemic Sclerosis. Scand J Immunol 2015; 82:269-74. [PMID: 26095613 DOI: 10.1111/sji.12317] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 06/07/2015] [Indexed: 01/31/2023]
Abstract
The pathophysiology of both limited cutaneous systemic sclerosis (lcSSc) and diffuse cutaneous SSc (dcSSc), representing two subtypes of an autoimmune disease of the connective tissue, is still enigmatic. Life-limiting, progressive fibrotic changes as a consequence of vasculopathy and autoimmunity are characteristic in varying extent for lcSSc and dcSSc. Previously, an increased IL-33 serum concentration in early phase SSc patients and an elevated tissue expression of its receptor, ST2L, on endothelial cells (EC) were described. While suggested as a biomarker for fibrotic diseases, for example liver fibrosis, the role of soluble ST2 (sST2) in the pathological processes and its contribution to vascular fibrosis in SSc has not been investigated. Here, we showed that sST2 is elevated in late phase limited cutaneous SSc (lcSSc) as compared to patients with shorter disease duration or with the diffuse subtype of SSc. We demonstrated that sST2, not IL-33, is significantly increased in serum of lcSSc patients with disease duration over 9 years. Soluble ST2 was not elevated in healthy controls or in SSc patients with early skin involvement or disease duration shorter than 9 years. Furthermore, we observed that sST2 serum levels were lowered by iloprost (prostacyclin) treatment. After 5 days of iloprost infusion, sST2 serum levels fell in 6 of 7 patients. Therefore, we not only like to propose sST2 as a biomarker for progressive vascular fibrosis, but moreover, suggest that the involvement of sST2 in the pathogenesis of lcSSc may be exploited therapeutically.
Collapse
Affiliation(s)
- A Wagner
- pharmazentrum frankfurt/ZAFES, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - M Köhm
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Frankfurt am Main, Germany
| | - A Nordin
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - E Svenungsson
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - J M Pfeilschifter
- pharmazentrum frankfurt/ZAFES, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - H H Radeke
- pharmazentrum frankfurt/ZAFES, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
33
|
Martínez-Martínez E, López-Ándres N, Jurado-López R, Rousseau E, Bartolomé MV, Fernández-Celis A, Rossignol P, Islas F, Antequera A, Prieto S, Luaces M, Cachofeiro V. Galectin-3 Participates in Cardiovascular Remodeling Associated With Obesity. Hypertension 2015; 66:961-9. [DOI: 10.1161/hypertensionaha.115.06032] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/10/2015] [Indexed: 02/07/2023]
Abstract
Remodeling, diastolic dysfunction, and arterial stiffness are some of the alterations through which obesity affects the cardiovascular system. Fibrosis and inflammation are important mechanisms underlying cardiovascular remodeling, although the precise promoters involved in these processes are still unclear. Galectin-3 (Gal-3) induces inflammation and fibrosis in the cardiovascular system. We have investigated the potential role of Gal-3 in cardiac damage in morbidly obese patients, and we have evaluated the protective effect of the Gal-3 inhibition in the occurrence of cardiovascular fibrosis and inflammation in an experimental model of obesity. Morbid obesity is associated with alterations in cardiac remodeling, mainly left ventricular hypertrophy and diastolic dysfunction. Obesity and hypertension are the main determinants of left ventricular hypertrophy. Insulin resistance, left ventricular hypertrophy, and circulating levels of C-reactive protein and Gal-3 are associated with a worsening of diastolic function in morbidly obese patients. Obesity upregulates Gal-3 production in the cardiovascular system in a normotensive animal model of diet-induced obesity by feeding for 6 weeks a high-fat diet (33.5% fat). Gal-3 inhibition with modified citrus pectin (100 mg/kg per day) reduced cardiovascular levels of Gal-3, total collagen, collagen I, transforming and connective growth factors, osteopontin, and monocyte chemoattractant protein-1 in the heart and aorta of obese animals without changes in body weight or blood pressure. In morbidly obese patients, Gal-3 levels are associated with diastolic dysfunction. In obese animals, Gal-3 blockade decreases cardiovascular fibrosis and inflammation. These data suggest that Gal-3 could be a novel therapeutic target in cardiac fibrosis and inflammation associated with obesity.
Collapse
Affiliation(s)
- Ernesto Martínez-Martínez
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Natalia López-Ándres
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Raquel Jurado-López
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Elodie Rousseau
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Mará Visitación Bartolomé
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Amaya Fernández-Celis
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Patrick Rossignol
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Fabian Islas
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Alfonso Antequera
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Santiago Prieto
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - María Luaces
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| | - Victoria Cachofeiro
- From the Cardiovascular Translational Research, Navarrabiomed (Fundación Miguel Servet), Pamplona, Spain (E.M.-M., N.L.-A., A.F.-C.); INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116 Université de Lorraine, CHU de Nancy, and INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France (N.L.-A., E.T., P.R.); Department of Physiology, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid,
| |
Collapse
|
34
|
Lancellotti P, Dulgheru R, Magne J, Henri C, Servais L, Bouznad N, Ancion A, Martinez C, Davin L, Le Goff C, Nchimi A, Piérard L, Oury C. Elevated Plasma Soluble ST2 Is Associated with Heart Failure Symptoms and Outcome in Aortic Stenosis. PLoS One 2015; 10:e0138940. [PMID: 26390433 PMCID: PMC4577123 DOI: 10.1371/journal.pone.0138940] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/06/2015] [Indexed: 01/27/2023] Open
Abstract
B-type natriuretic peptide (BNP) is often used as a complementary finding in the diagnostic work-up of patients with aortic stenosis (AS). Whether soluble ST2, a new biomarker of cardiac stretch, is associated with symptomatic status and outcome in asymptomatic AS is unknown. sST2 and BNP levels were measured in 86 patients (74±13 years; 59 asymptomatic, 69%) with AS (<1.5 cm2) and preserved left ventricular ejection fraction who were followed-up for 26±16 months. Both BNP and sST2 were associated with NYHA class but sST2 (>23 ng/mL, AUC = 0.68, p<0.01) was more accurate to identify asymptomatic patients or those who developed symptoms during follow-up. sST2 was independently related to left atrial index (p<0.0001) and aortic valve area (p = 0.004; model R2 = 0.32). A modest correlation was found with BNP (r = 0.4, p<0.01). During follow-up, 29 asymptomatic patients (34%) developed heart failure symptoms. With multivariable analysis, peak aortic jet velocity (HR = 2.7, p = 0.007) and sST2 level (HR = 1.04, p = 0.03) were independent predictors of cardiovascular events. In AS, sST2 levels could provide complementary information regarding symptomatic status, new onset heart failure symptoms and outcome. It might become a promising biomarker in these patients.
Collapse
Affiliation(s)
- Patrizio Lancellotti
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
- Gruppo Villa Maria Care and Research, Lugo (RA), Italy
- * E-mail: (PL); (CO); (AN)
| | - Raluca Dulgheru
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Julien Magne
- Department of Cardiology, CHU Dupuytren, and INSERM 1094, Faculté de Médecine de Limoges, Limoges, France
| | - Christine Henri
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Laurence Servais
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Nassim Bouznad
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Arnaud Ancion
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Christophe Martinez
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Laurent Davin
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Caroline Le Goff
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Alain Nchimi
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
- * E-mail: (PL); (CO); (AN)
| | - Luc Piérard
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
| | - Cécile Oury
- GIGA Cardiovascular Sciences, Heart Valve Clinic, Department of Cardiology and Radiology, University of Liège Hospital, and University of Liège, Liège, Belgium
- * E-mail: (PL); (CO); (AN)
| |
Collapse
|
35
|
Martínez-Martínez E, Cachofeiro V, Rousseau E, Álvarez V, Calvier L, Fernández-Celis A, Leroy C, Miana M, Jurado-López R, Briones AM, Jaisser F, Zannad F, Rossignol P, López-Andrés N. Interleukin-33/ST2 system attenuates aldosterone-induced adipogenesis and inflammation. Mol Cell Endocrinol 2015; 411:20-7. [PMID: 25896545 DOI: 10.1016/j.mce.2015.04.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/13/2015] [Accepted: 04/08/2015] [Indexed: 01/01/2023]
Abstract
Interleukin-33 (IL-33) but not soluble ST2 (sST2) exerts anti-inflammatory and protective effects in several tissues. Aldosterone, a proinflammatory mediator which promotes adipogenesis, is elevated in obese patients. The aim of this study was to investigate the interactions between IL-33/ST2 system and Aldosterone in adipose tissue. Rats fed a high fat diet presented increased sST2 expression, diminished IL-33/sST2 ratio and enhanced levels of differentiation and inflammation in adipose tissue as compared to controls. A similar pattern was observed in adipose tissue from C57BL/6 Aldosterone-treated mice. In both animal models, Aldosterone was correlated with sST2. Treatment of 3T3-L1 adipocytes with IL-33 delayed adipocyte differentiation diminished lipid accumulation and decreased inflammation. Aldosterone decreased IL-33 and increased sST2 expressions in differentiated adipocytes. Aldosterone-induced adipocyte differentiation and inflammation were blocked by IL-33 treatment, but sST2 did not exert any effects. The crosstalk between IL-33/ST2 and Aldosterone could be relevant in the metabolic consequences of obesity.
Collapse
Affiliation(s)
| | - Victoria Cachofeiro
- Department of Physiology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Elodie Rousseau
- INSERM, Centre d'Investigations Cliniques- Plurithématique 1433, UMR 1116 Université de Lorraine and CHU de Nancy, Vandoeuvre-lès-Nancy, France
| | - Virginia Álvarez
- Cardiovascular Translational Research, NavarraBiomed (Fundación Miguel Servet), Pamplona, Spain; Heart Area, Hospital de Navarra, Pamplona, Spain
| | - Laurent Calvier
- INSERM, Centre d'Investigations Cliniques- Plurithématique 1433, UMR 1116 Université de Lorraine and CHU de Nancy, Vandoeuvre-lès-Nancy, France
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, NavarraBiomed (Fundación Miguel Servet), Pamplona, Spain
| | - Céline Leroy
- INSERM, Centre d'Investigations Cliniques- Plurithématique 1433, UMR 1116 Université de Lorraine and CHU de Nancy, Vandoeuvre-lès-Nancy, France
| | - María Miana
- Department of Physiology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Raquel Jurado-López
- Department of Physiology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Ana M Briones
- Department of Pharmacology, Universidad Autónoma de Madrid, Spain
| | - Frederic Jaisser
- INSERM, Centre d'Investigations Cliniques- Plurithématique 1433, UMR 1116 Université de Lorraine and CHU de Nancy, Vandoeuvre-lès-Nancy, France; INSERM UMR 872 Team 1, Centre de Recherche des Cordeliers, University Pierre and Marie Curie, Paris, France
| | - Faiez Zannad
- INSERM, Centre d'Investigations Cliniques- Plurithématique 1433, UMR 1116 Université de Lorraine and CHU de Nancy, Vandoeuvre-lès-Nancy, France
| | - Patrick Rossignol
- INSERM, Centre d'Investigations Cliniques- Plurithématique 1433, UMR 1116 Université de Lorraine and CHU de Nancy, Vandoeuvre-lès-Nancy, France
| | - Natalia López-Andrés
- Cardiovascular Translational Research, NavarraBiomed (Fundación Miguel Servet), Pamplona, Spain; INSERM, Centre d'Investigations Cliniques- Plurithématique 1433, UMR 1116 Université de Lorraine and CHU de Nancy, Vandoeuvre-lès-Nancy, France.
| |
Collapse
|
36
|
Baseline body mass index among children and adults undergoing allogeneic hematopoietic cell transplantation: clinical characteristics and outcomes. Bone Marrow Transplant 2014; 50:402-10. [PMID: 25531283 DOI: 10.1038/bmt.2014.280] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/14/2014] [Accepted: 11/03/2014] [Indexed: 01/14/2023]
Abstract
Obesity is an important public health problem that may influence the outcomes of hematopoietic cell transplantation (HCT). We studied 898 children and adults receiving first-time allogeneic hematopoietic SCTs between 2004 and 2012. Pretransplant body mass index (BMI) was classified as underweight, normal weight, overweight or obese using the WHO classification or age-adjusted BMI percentiles for children. The study population was predominantly Caucasian, and the median age was 51 years (5 months-73 years). The cumulative 3-year incidence of nonrelapse mortality (NRM) in underweight, normal weight, overweight and obese patients was 20%, 19%, 20% and 33%, respectively. Major causes of NRM were acute and chronic GVHD. The corresponding incidence of relapse was 30%, 41%, 37% and 30%, respectively. Three-year OS was 59%, 48%, 47% and 43%, respectively. Multivariate analysis showed that obesity was associated with higher NRM (hazard ratio (HR) 1.43, P=0.04) and lower relapse (HR 0.65, P=0.002). Pretransplant plasma levels of ST2 and TNFR1 biomarkers were significantly higher in obese compared with normal weight patients (P=0.04 and P=0.05, respectively). The increase in NRM observed in obese patients was partially offset by a lower incidence of relapse with no difference in OS.
Collapse
|
37
|
Abreu AR, de Abreu AR, Santos LT, de Souza AA, da Silva LG, Chianca DA, de Menezes RC. Blunted GABA-mediated inhibition within the dorsomedial hypothalamus potentiates the cardiovascular response to emotional stress in rats fed a high-fat diet. Neuroscience 2014; 262:21-30. [PMID: 24397951 DOI: 10.1016/j.neuroscience.2013.12.053] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/23/2013] [Accepted: 12/24/2013] [Indexed: 11/25/2022]
Abstract
Rats fed a high-fat diet (HFD) present an exaggerated endocrine response to stress conditions, which, like obesity, show a high correlation with cardiovascular diseases. Meanwhile the GABAergic neurotransmission within the dorsomedial hypothalamus (DMH) is involved in the regulation of the physiological responses during emotional stress. Here we evaluated the influence of obesity, induced by a HFD, on the cardiovascular responses induced by air jet stress in rats, and the role of the GABAergic tonus within the DMH in these changes. Our results showed that consumption of a HFD (45% w/w fat) for 9 weeks induced obesity and increases in baseline mean arterial pressure (MAP) and heart rate (HR). Moreover, obesity potentiated stress responsiveness, evidenced by the greater changes in MAP and HR induced by stress in obese rats. The injection of muscimol into the DMH reduced the maximal increases in HR and MAP induced by stress in both groups; however, the reduction in the maximal increases in MAP in the HFD group was less pronounced. Moreover, the injection of muscimol into the DMH of obese rats was less effective in reducing the stress-induced tachycardia, since the HR attained the same levels at the end of the stress paradigm as after the vehicle injection. Injection of bicuculline into DMH induced increases in MAP and HR in both groups. Nevertheless, obesity shortened the tachycardic response to bicuculline injection. These data show that obesity potentiates the cardiovascular response to stress in rats due to an inefficient GABAA-mediated inhibition within the DMH.
Collapse
Affiliation(s)
- A R Abreu
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| | - A R de Abreu
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| | - L T Santos
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| | - A A de Souza
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| | - L G da Silva
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| | - D A Chianca
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| | - R C de Menezes
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, MG, Brazil.
| |
Collapse
|