1
|
Ma Z, Ma S, Chen B, Zhang Y, Zeng J, Tao J, Hu Y. Classification patterns identification of immunogenic cell death-related genes in heart failure based on deep learning. Sci Rep 2025; 15:5633. [PMID: 39955386 PMCID: PMC11829947 DOI: 10.1038/s41598-025-89333-1] [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: 08/09/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
Heart failure (HF) is a complex and prevalent condition, particularly in the elderly, presenting symptoms like chest tightness, shortness of breath, and dyspnea. The study aimed to improve the classification of HF subtypes and identify potential drug targets by exploring the role of Immunogenic Cell Death (ICD), a process known for its role in tumor immunity but underexplored in HF research. Additionally, the study sought to apply deep learning models to enhance HF classification and identify diagnosis-related genes. Various deep learning encoder models were employed to evaluate their effectiveness in clustering HF based on ICD-related genes. Identified HF subtypes were further refined using differentially expressed genes, allowing for the assessment of immune infiltration and functional enrichment. Advanced machine learning techniques were used to identify diagnosis-related genes, and these genes were used to construct nomogram models. The study also explored gene interactions with miRNA and transcription factors. Distinct HF subtypes were identified through clustering based on ICD-related genes. Differentially expressed genes revealed significant variations in immune infiltration and functional enrichment across these subtypes. The diagnostic model showed excellent performance, with an AUC exceeding 0.99 in both internal and external test sets. Diagnosis-related genes were also identified, serving as the foundation for nomogram models and further exploration of their regulatory interactions. This study provides a novel insight into HF by combining the exploration of ICD, the application of deep learning models, and the identification of diagnosis-related genes. These findings contribute to a deeper understanding of HF subtypes and highlight potential therapeutic targets for improving HF classification and treatment.
Collapse
Affiliation(s)
- Zhihui Ma
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Shixin Ma
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Bin Chen
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yongjun Zhang
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jinmei Zeng
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jianping Tao
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yu Hu
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
2
|
Son WH, Jeong WM, Park IY, Ha MS. Enhancing Inflammatory Factors, Nitric Oxide, and Arterial Stiffness Through Aquatic Walking for Amelioration and Disease Prevention: Targeting in Obese Elderly Women. Mediators Inflamm 2024; 2024:5520987. [PMID: 39742290 PMCID: PMC11685319 DOI: 10.1155/mi/5520987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 11/13/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
In elderly women, hormonal changes lead to elevated body fat content, which results in elevated levels of vascular inflammatory factors, thereby increasing the risk of cardiovascular diseases (CVDs) associated with endothelial dysfunction. Regular physical exercises tend to keep these in check and are protective to the body. Aerobic exercise has been reported to improve CVD in obese elderly women; in this regard, aquatic exercises have been demonstrated to be more efficient in energy metabolism than land-based exercise. This study aimed to examine the effect of aquatic walking exercises on the levels of inflammatory factors, nitric oxide (NO), and brachial-ankle pulse wave velocity (baPWV) in obese elderly women. We measured these in 26 obese elderly women who were randomly assigned to control (n = 12) and aquatic walking exercise (n = 14) groups. After subjecting them to aquatic walking exercises thrice a week for 12 weeks, we specifically found a significant reduction in IL-6 levels and an increase in NO levels in these obese elderly women. This was paralleled with a reduction in the right baPWV (baPWV-R). Together, these results indicate that aquatic walking exercises can help improve vascular inflammatory factors, NO levels, and arterial stiffness.
Collapse
Affiliation(s)
- Woo-Hyeon Son
- Institute of Convergence Bio-Health, Dong-A University, 26 Daesingongwon-ro, Seo-gu, Busan 49201, Republic of Korea
| | - Woo-Min Jeong
- Department of Sport and Leisure Studies, Gimcheon University, 214 Daehak-ro, Gimcheon-si, Gyeongsangbuk-do 39528, Republic of Korea
| | - In Young Park
- Undergraduate Liberal Arts College, Tongmyong University, 428 Sinseon-ro, Nam-gu, Busan 48520, Republic of Korea
| | - Min-Seong Ha
- Laboratory of Sports Conditioning: Nutrition Biochemistry and Neuroscience, Department of Sport Science, College of Arts and Sports, University of Seoul, 163 Seoulsiripdaero, Dongdaemun-gu, Seoul 02504, Republic of Korea
| |
Collapse
|
3
|
Dutka M, Garczorz W, Kosowska A, Buczek E, Godek P, Wojakowski W, Francuz T. Osteoprotegerin Is Essential for the Development of Endothelial Dysfunction Induced by Angiotensin II in Mice. Int J Mol Sci 2024; 25:6434. [PMID: 38928140 PMCID: PMC11203749 DOI: 10.3390/ijms25126434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Opinions on the effects of osteoprotegerin (OPG) have evolved over the years from a protein protecting the vasculature from calcification to a cardiovascular risk factor contributing to inflammation within the vascular wall. Nowadays, the link between OPG and angiotensin II (Ang II) appears to be particularly important. In this study, the endothelial function was investigated in OPG-knockout mice (B6.129.S4-OPG, OPG-) and wild-type (C57BL/6J, OPG+) mice under basic conditions and after Ang II exposure by assessing the endothelium-dependent diastolic response of aortic rings to acetylcholine in vitro. A further aim of the study was to compare the effect of Ang II on the expression of cytokines in the aortic wall of both groups of mice. Our study shows that rings from OPG- mice had their normal endothelial function preserved after incubation with Ang II, whereas those from OPG+ mice showed significant endothelial dysfunction. We conclude that the absence of OPG, although associated with a pro-inflammatory cytokine profile in the vascular wall, simultaneously protects against Ang II-induced increases in pro-inflammatory cytokines in the murine vascular wall. Our study also demonstrates that the absence of OPG can result in a decrease in the concentration of pro-inflammatory cytokines in the vascular wall after Ang II exposure. The presence of OPG is therefore crucial for the development of Ang II-induced inflammation in the vascular wall and for the development of Ang II-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biala, Poland
| | - Wojciech Garczorz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (W.G.); (A.K.); (T.F.)
| | - Agnieszka Kosowska
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (W.G.); (A.K.); (T.F.)
| | - Elzbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland;
| | - Piotr Godek
- Department of Cardiology and Structural Heart Disease, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (P.G.); (W.W.)
| | - Wojciech Wojakowski
- Department of Cardiology and Structural Heart Disease, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (P.G.); (W.W.)
| | - Tomasz Francuz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (W.G.); (A.K.); (T.F.)
| |
Collapse
|
4
|
Alizade E, Kahyaoglu M, Balaban I, Izci S, Guler A. Osteoprotegerin is associated with subclinical left ventricular systolic dysfunction in non-dipper hypertensive patients: a 2D speckle tracking echocardiographic study. Blood Press Monit 2024; 29:55-62. [PMID: 37937620 DOI: 10.1097/mbp.0000000000000681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
BACKGROUND Past studies have shown that non-dipper hypertensive patients have more frequent subclinical left ventricular (LV) systolic dysfunction compared to dippers. Many different parameters have been examined to predict subclinical LV dysfunction. The role of osteoprotegerin (OPG) in the pathogenesis of heart failure and LV systolic dysfunction through different mechanisms had well described. In the present study, we hypothesized that increased OPG levels could predict subclinical LV systolic dysfunction in non-dipper hypertensive patients. PATIENTS AND METHODS Hypertensive patients were divided into two groups according to the results of ambulatory blood pressure (BP) monitoring. Non-dipper patients were subsequently divided into two further groups (normal LV function and impaired LV function) according to LV global longitudinal strain (GLS). RESULTS A total of 103 hypertensive patients (51 dippers, 52 non-dippers) were included in the study. In the non-dipper group, LV GLS was normal in 21 patients and impaired in 31 patients. Based on the results of the multivariate logistic regression test, it was determined that OPG levels (OR: 2.413, 95% CI: 1.284-4.535, P = 0.006) and LVMI (OR: 1.086, 95% CI: 1.013-1.165, P = 0.021) were independently associated with impaired GLS. CONCLUSION Higher OPG values were associated with subclinical LV systolic dysfunction in non-dipper hypertensive patients. It could be used for the early diagnosis of subclinical LV systolic dysfunction, which would allow for strategies to be designed to reduce the cardiovascular event rate in this patient population.
Collapse
Affiliation(s)
| | | | | | | | - Ahmet Guler
- Başakşehir Cam and Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
5
|
Li Y, Hu Y, Jiang F, Chen H, Xue Y, Yu Y. Combining WGCNA and machine learning to identify mechanisms and biomarkers of ischemic heart failure development after acute myocardial infarction. Heliyon 2024; 10:e27165. [PMID: 38455553 PMCID: PMC10918227 DOI: 10.1016/j.heliyon.2024.e27165] [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: 07/10/2023] [Revised: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Background Ischemic heart failure (IHF) is a serious complication after acute myocardial infarction (AMI). Understanding the mechanism of IHF after AMI will help us conduct early diagnosis and treatment. Methods We obtained the AMI dataset GSE66360 and the IHF dataset GSE57338 from the GEO database, and screened overlapping genes common to both diseases through WGCNA analysis. Subsequently, we performed GO and KEGG enrichment analysis on overlapping genes to elucidate the common mechanism of AMI and IHF. Machine learning algorithms are also used to identify key biomarkers. Finally, we performed immune cell infiltration analysis on the dataset to further evaluate immune cell changes in AMI and IHF. Results We obtained 74 overlapping genes of AMI and IHF through WGCNA analysis, and the enrichment analysis results mainly focused on immune and inflammation-related mechanisms. Through the three machine learning algorithms of LASSO, RF and SVM-RFE, we finally obtained the four Hub genes of IL1B, TIMP2, IFIT3, and P2RY2, and verified them in the IHF dataset GSE116250, and the diagnostic model AUC = 0.907. The results of immune infiltration analysis showed that 8 types of immune cells were significantly different in AMI samples, and 6 types of immune cells were significantly different in IHF samples. Conclusion We explored the mechanism of IHF after AMI by WGCNA, enrichment analysis, and immune infiltration analysis. Four potential diagnostic candidate genes and therapeutic targets were identified by machine learning algorithms. This provides a new idea for the pathogenesis, diagnosis, and treatment of IHF after AMI.
Collapse
Affiliation(s)
- Yan Li
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Ying Hu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Feng Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Haoyu Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| |
Collapse
|
6
|
Xiang G, Guo S, Qin J, Gao H, Zhang Y, Wang S. Comprehensive insight into the pharmacology, pharmacokinetics, toxicity, detoxification and extraction of hypaconitine from Aconitum plants. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117505. [PMID: 38016573 DOI: 10.1016/j.jep.2023.117505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hypaconitine (HA), a diterpenoid alkaloid, mainly derived from Aconitum plants such as Acoitum carmichaeli Debx. And Aconitum nagarum Stapf., has recently piqued significant interest among the scientific community given its multifaceted attributes including anti-inflammatory, anticancer, analgesic, and cardio-protective properties. AIM OF THE STUDY This review presents a comprehensive exploration of the research advancements regarding the traditional uses, pharmacology, pharmacokinetics, toxicity, and toxicity reduction of HA. It aims to provide a thorough understanding of HA's multifaceted properties and its potential applications in various fields. MATERIALS AND METHODS A systematic literature search was conducted using several prominent databases including PubMed, Web of Science, NCBI, and CNKI. The search was performed using specific keywords such as "hypaconitine," "heart failure," "anti-inflammatory," "aconite decoction," "pharmacological," "pharmacokinetics," "toxicity," "detoxification or toxicity reduction," and "extraction and isolation." The inclusion of these keywords ensured a comprehensive exploration of relevant studies and enabled the retrieval of valuable information pertaining to the various aspects of HA. RESULTS Existing research has firmly established that HA possesses a range of pharmacological effects, encompassing anti-cardiac failure, anti-inflammatory, analgesic, and anti-tumor properties. The therapeutic potential of HA is promising, with potential applications in heart failure, ulcerative colitis, cancer, and other diseases. Pharmacokinetic studies suggest that HA exhibits high absorption rates, broad distribution, and rapid metabolism. However, toxic effects of HA on the nerves, heart, and embryos have also been observed. To mitigate these risks, HA needs attenuation before use, with the most common detoxification methods being processing and combined use with other drugs. Extraction methods for HA most commonly include cold maceration, soxhlet reflux extraction, and ultrasonic-assisted extraction. Despite the potential therapeutic benefits of HA, further research is warranted to elucidate its anti-heart failure effects, particularly in vivo, exploring aspects such as in vivo metabolism, distribution, and metabolites. Additionally, the therapeutic effects of HA monomers on inflammation-induced diseases and tumors should be validated in a more diverse range of experimental models, while the mechanisms underlying the therapeutic effects of HA should be investigated in greater detail. CONCLUSION This review serves to emphasize the therapeutic potential of HA and highlights the crucial need to address its toxicity concerns before considering clinical application. Further research is required to comprehensively investigate the pharmacological properties of HA, with particular emphasis on its anti-cardiac failure and anti-inflammatory activities. Such research endeavors have the potential to unveil novel treatment avenues for a broad spectrum of diseases.
Collapse
Affiliation(s)
- Gelin Xiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Sa Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jing Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Huimin Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, China.
| |
Collapse
|
7
|
Nisar A, Jagtap S, Vyavahare S, Deshpande M, Harsulkar A, Ranjekar P, Prakash O. Phytochemicals in the treatment of inflammation-associated diseases: the journey from preclinical trials to clinical practice. Front Pharmacol 2023; 14:1177050. [PMID: 37229273 PMCID: PMC10203425 DOI: 10.3389/fphar.2023.1177050] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Advances in biomedical research have demonstrated that inflammation and its related diseases are the greatest threat to public health. Inflammatory action is the pathological response of the body towards the external stimuli such as infections, environmental factors, and autoimmune conditions to reduce tissue damage and improve patient comfort. However, when detrimental signal-transduction pathways are activated and inflammatory mediators are released over an extended period of time, the inflammatory process continues and a mild but persistent pro-inflammatory state may develop. Numerous degenerative disorders and chronic health issues including arthritis, diabetes, obesity, cancer, and cardiovascular diseases, among others, are associated with the emergence of a low-grade inflammatory state. Though, anti-inflammatory steroidal, as well as non-steroidal drugs, are extensively used against different inflammatory conditions, they show undesirable side effects upon long-term exposure, at times, leading to life-threatening consequences. Thus, drugs targeting chronic inflammation need to be developed to achieve better therapeutic management without or with a fewer side effects. Plants have been well known for their medicinal use for thousands of years due to their pharmacologically active phytochemicals belonging to diverse chemical classes with a number of these demonstrating potent anti-inflammatory activity. Some typical examples include colchicine (alkaloid), escin (triterpenoid saponin), capsaicin (methoxy phenol), bicyclol (lignan), borneol (monoterpene), and quercetin (flavonoid). These phytochemicals often act via regulating molecular mechanisms that synergize the anti-inflammatory pathways such as increased production of anti-inflammatory cytokines or interfere with the inflammatory pathways such as to reduce the production of pro-inflammatory cytokines and other modulators to improve the underlying pathological condition. This review describes the anti-inflammatory properties of a number of biologically active compounds derived from medicinal plants, and their mechanisms of pharmacological intervention to alleviate inflammation-associated diseases. The emphasis is given to information on anti-inflammatory phytochemicals that have been evaluated at the preclinical and clinical levels. Recent trends and gaps in the development of phytochemical-based anti-inflammatory drugs have also been included.
Collapse
Affiliation(s)
- Akib Nisar
- Biochemical Sciences Division, Rajiv Gandhi Institute of IT and Biotechnology, Bharati Vidyapeeth Deemed to be University, Pune, Maharashtra, India
| | - Suresh Jagtap
- Herbal Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed to be University, Pune, Maharashtra, India
| | - Suresh Vyavahare
- Shatayu Ayurved and Research Centre, Solapur, Maharashtra, India
| | - Manasi Deshpande
- Department of Dravyagun Vigyan, College of Ayurved, Bharati Vidyapeeth Deemed to be University, Pune, Maharashtra, India
| | - Abhay Harsulkar
- Herbal Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth Deemed to be University, Pune, Maharashtra, India
- Pharmaceutical Biotechnology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed to be University, Pune, Maharashtra, India
| | | | - Om Prakash
- Department of Microbiology, Immunology and Parasitology, University Health Sciences Center, New Orleans, LA, United States
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
8
|
Dutka M, Bobiński R, Wojakowski W, Francuz T, Pająk C, Zimmer K. Osteoprotegerin and RANKL-RANK-OPG-TRAIL signalling axis in heart failure and other cardiovascular diseases. Heart Fail Rev 2021; 27:1395-1411. [PMID: 34313900 PMCID: PMC9197867 DOI: 10.1007/s10741-021-10153-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 01/29/2023]
Abstract
Osteoprotegerin (OPG) is a glycoprotein involved in the regulation of bone remodelling. OPG regulates osteoclast activity by blocking the interaction between the receptor activator of nuclear factor kappa B (RANK) and its ligand (RANKL). More and more studies confirm the relationship between OPG and cardiovascular diseases. Numerous studies have confirmed that a high plasma concentration of OPG and a low concentration of tumour necrosis factor–related apoptosis inducing ligand (TRAIL) together with a high OPG/TRAIL ratio are predictors of poor prognosis in patients with myocardial infarction. A high plasma OPG concentration and a high ratio of OPG/TRAIL in the acute myocardial infarction are a prognostic indicator of adverse left ventricular remodelling and of the development of heart failure. Ever more data indicates the participation of OPG in the regulation of the function of vascular endothelial cells and the initiation of the atherosclerotic process in the arteries. Additionally, it has been shown that TRAIL has a protective effect on blood vessels and exerts an anti-atherosclerotic effect. The mechanisms of action of both OPG and TRAIL within the cells of the vascular wall are complex and remain largely unclear. However, these mechanisms of action as well as their interaction in the local vascular environment are of great interest to researchers. This article presents the current state of knowledge on the mechanisms of action of OPG and TRAIL in the circulatory system and their role in cardiovascular diseases. Understanding these mechanisms may allow their use as a therapeutic target in cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Rafał Bobiński
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Wojciech Wojakowski
- Department of Cardiology and Structural Heart Disease, Medical University of Silesia, Katowice, Poland
| | - Tomasz Francuz
- Department of Biochemistry, Medical University of Silesia, Katowice, Poland
| | - Celina Pająk
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Karolina Zimmer
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
9
|
Mao S, Zhang X, Chen M, Wang C, Chen Q, Guo L, Zhang M, Hinek A. Beneficial Effects of Baduanjin Exercise on Left Ventricular Remodelling in Patients after Acute Myocardial Infarction: an Exploratory Clinical Trial and Proteomic Analysis. Cardiovasc Drugs Ther 2020; 35:21-32. [PMID: 32761487 DOI: 10.1007/s10557-020-07047-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The beneficial effects of physical exercise on cardiac remodelling improvement after myocardial infarction have already been suggested. However, the results of previous clinical trials have not been consistent. Moreover, the putative molecular mechanisms leading to the clinically observed effects of physical exercise still remain elusive. AIM We aimed to evaluate whether the well-defined and strictly controlled traditional Chinese Qigong Baduanjin exercise (BE) would attenuate the adverse left ventricular (LV) remodelling in patients with ST-elevation myocardial infarction (STEMI). METHODS A total of 110 clinically stable STEMI patients, following successful revascularization of their infarcted coronary arteries, were randomized and enrolled in two groups: 56 were subjected to a 12-week BE-based cardiac rehabilitation programme (BE group), and the remaining 54 were exposed to the usual physical exercise (control group) for the same time period. The primary outcome was the change from baseline to 6 months in the echocardiographic LV end-diastolic volume index (ΔLVEDVi). Proteomic analysis was also performed to uncover associated mechanisms. RESULTS Compared with the control group, the BE group showed significantly lower ΔLVEDVi (-5.1 ± 1.1 vs. 0.3 ± 1.2 mL/m2, P < 0.01). Proteomic analysis revealed BE-induced variations in the expression of 80 proteins linked to regulation the of metabolic process, immune process, and extracellular matrix reorganization. Furthermore, correlation analyses between the validated serum proteomes and primary endpoint demonstrated a positive association between ΔLVEDVi and MMP-9 expression, but a negative correlation between ΔLVEDVi and CXCL1 expression. CONCLUSION This is the first study indicating that BE in STEMI patients can alleviate adverse LV remodelling associated with beneficial energy metabolism adaptation, inflammation curbing, and extracellular matrix organization adjustment.
Collapse
Affiliation(s)
- Shuai Mao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China.,Translational Medicine, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | - Xiaoxuan Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Minggui Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Chuyang Wang
- Biological Resource Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Qubo Chen
- Biological Resource Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Liheng Guo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Minzhou Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China. .,Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China.
| | - Aleksander Hinek
- Translational Medicine, Hospital for Sick Children, Toronto, M5G 0A4, Canada
| |
Collapse
|
10
|
Abstract
Myocardial infarction and post-infarction left ventricular remodelling involve a high risk of morbidity and mortality. For this reason, ongoing research is being conducted in order to learn the mechanisms of unfavourable left ventricular remodelling following a myocardial infarction. New biomarkers are also being sought that would allow for early identification of patients with a high risk of post-infarction remodelling and dysfunction of the left ventricle. In recent years, there has been ever more experimental data that confirms the significance of microRNA in cardiovascular diseases. It has been confirmed that microRNAs are stable in systemic circulation, and can be directly measured in patients' blood. It has been found that significant changes occur in the concentrations of various types of microRNA in myocardial infarction and heart failure patients. Various types of microRNA are also currently being intensively researched in terms of their usefulness as markers of cardiomyocyte necrosis, and predictors of the post-infarction heart failure development. This paper is a summary of the current knowledge on the significance of microRNA in post-infarction left ventricular remodelling and heart failure.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Faculty of Health Sciences, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Rafał Bobiński
- Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Faculty of Health Sciences, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Jan Korbecki
- Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Faculty of Health Sciences, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
11
|
Zampetti S, Lucantoni F, Pacifico L, Campagna G, Versacci P, Pierimarchi P, Buzzetti R. Association of OPG-RANKL ratio with left ventricular hypertrophy and geometric remodeling in male overweight/obese youths. J Endocrinol Invest 2019; 42:427-434. [PMID: 30132289 DOI: 10.1007/s40618-018-0932-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/01/2018] [Indexed: 01/11/2023]
Abstract
PURPOSE Receptor activator of nuclear factor kappa B ligand/receptor activator of nuclear factor kappa B/osteoprotegerin (RANKL/RANK/OPG) axis has been hypothesized as a potential mediator of left ventricular hypertrophy (LVH). The aim of the study was to assess whether circulating concentrations of RANKL, RANK, and OPG were associated with early signs of morphological cardiac changes in overweight/obese youths. METHODS We determined serum levels of RANKL, RANK and OPG by enzyme-linked immunosorbent assays in 188 overweight/obese children and adolescents. LV mass index (LVMI) and relative wall thickness (RWT) were estimated using M-mode echocardiography. RESULTS OPG and RANKL levels were higher among girls than among boys [1.73 (1.64-1.86) and 3.28 (1.90-6.37) pmol/L, respectively, vs. 1.69 (1.59-1.82) and 2.12 (1.52-3.80) pmol/L; p = 0.02 and p = 0.0001, respectively], but the OPG/RANKL ratio was lower [0.52 (0.26-0.88) vs 0.77 (0.44-1.11); p = 0.001]. In gender-specific multivariate linear regression, OPG/RANKL ratio was associated with LVMI and RWT in boys but not in girls. In multiple logistic regression, after adjustment for clinical variables, OPG/RANKL ratio was associated with concentric remodeling, eccentric and concentric LVH in boys but not in girls. CONCLUSION OPG/RANKL ratio is independently associated with LVH and patterns of LV structural remodeling in male overweight/obese children and adolescents.
Collapse
Affiliation(s)
- S Zampetti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - F Lucantoni
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - L Pacifico
- Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - G Campagna
- Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - P Versacci
- Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - P Pierimarchi
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - R Buzzetti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
12
|
Hage C, Michaëlsson E, Linde C, Donal E, Daubert JC, Gan LM, Lund LH. Inflammatory Biomarkers Predict Heart Failure Severity and Prognosis in Patients With Heart Failure With Preserved Ejection Fraction: A Holistic Proteomic Approach. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.116.001633. [PMID: 28100627 DOI: 10.1161/circgenetics.116.001633] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 11/29/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Underlying mechanisms in heart failure (HF) with preserved ejection fraction remain unknown. We investigated cardiovascular plasma biomarkers in HF with preserved ejection fraction and their correlation to diastolic dysfunction, functional class, pathophysiological processes, and prognosis. METHODS AND RESULTS In 86 stable patients with HF and EF ≥45% in the Karolinska Rennes (KaRen) biomarker substudy, biomarkers were quantified by a multiplex immunoassay. Orthogonal projection to latent structures by partial least square analysis was performed on 87 biomarkers and 240 clinical variables, ranking biomarkers associated with New York Heart Association (NYHA) Functional class and the composite outcome (all-cause mortality and HF hospitalization). Biomarkers significantly correlated with outcome were analyzed by multivariable Cox regression and correlations with echocardiographic measurements performed. The orthogonal partial least square outcome-predicting biomarker pattern was run against the Ingenuity Pathway Analysis (IPA) database, containing annotated data from the public domain. The orthogonal partial least square analyses identified 32 biomarkers correlated with NYHA class and 28 predicting outcomes. Among outcome-predicting biomarkers, growth/differentiation factor-15 was the strongest and an additional 7 were also significant in Cox regression analyses when adjusted for age, sex, and N-terminal probrain natriuretic peptide: adrenomedullin (hazard ratio per log increase 2.53), agouti-related protein; (1.48), chitinase-3-like protein 1 (1.35), C-C motif chemokine 20 (1.35), fatty acid-binding protein (1.33), tumor necrosis factor receptor 1 (2.29), and TNF-related apoptosis-inducing ligand (0.34). Twenty-three of them correlated with diastolic dysfunction (E/e') and 5 with left atrial volume index. The IPA suggested that increased inflammation, immune activation with decreased necrosis and apoptosis preceded poor outcome. CONCLUSIONS In HF with preserved ejection fraction, novel biomarkers of inflammation predict HF severity and prognosis that may complement or even outperform traditional markers, such as N-terminal probrain natriuretic peptide. These findings lend support to a hypothesis implicating global systemic inflammation in HF with preserved ejection fraction. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov; Unique identifier: NCT00774709.
Collapse
Affiliation(s)
- Camilla Hage
- From the Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (C.H., C.L., L.H.L.); Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden (C.H., C.L., L.H.L.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal, Sweden (E.M., L.-M.G.); Département de Cardiologie and CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France (E.D., J.-C.D.); and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Sweden (L.-M.G.).
| | - Erik Michaëlsson
- From the Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (C.H., C.L., L.H.L.); Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden (C.H., C.L., L.H.L.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal, Sweden (E.M., L.-M.G.); Département de Cardiologie and CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France (E.D., J.-C.D.); and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Sweden (L.-M.G.)
| | - Cecilia Linde
- From the Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (C.H., C.L., L.H.L.); Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden (C.H., C.L., L.H.L.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal, Sweden (E.M., L.-M.G.); Département de Cardiologie and CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France (E.D., J.-C.D.); and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Sweden (L.-M.G.)
| | - Erwan Donal
- From the Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (C.H., C.L., L.H.L.); Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden (C.H., C.L., L.H.L.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal, Sweden (E.M., L.-M.G.); Département de Cardiologie and CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France (E.D., J.-C.D.); and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Sweden (L.-M.G.)
| | - Jean-Claude Daubert
- From the Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (C.H., C.L., L.H.L.); Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden (C.H., C.L., L.H.L.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal, Sweden (E.M., L.-M.G.); Département de Cardiologie and CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France (E.D., J.-C.D.); and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Sweden (L.-M.G.)
| | - Li-Ming Gan
- From the Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (C.H., C.L., L.H.L.); Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden (C.H., C.L., L.H.L.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal, Sweden (E.M., L.-M.G.); Département de Cardiologie and CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France (E.D., J.-C.D.); and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Sweden (L.-M.G.)
| | - Lars H Lund
- From the Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (C.H., C.L., L.H.L.); Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden (C.H., C.L., L.H.L.); Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal, Sweden (E.M., L.-M.G.); Département de Cardiologie and CIC-IT U 804, Centre Hospitalier Universitaire de Rennes, France (E.D., J.-C.D.); and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Sweden (L.-M.G.)
| |
Collapse
|
13
|
Arisi MF, Chirico EN, Sebeny R, Muthukumaran G, Mu A, De Jonghe BC, Margulies KB, Libonati JR. Myocardial apoptosis and mesenchymal stem cells with acute exercise. Physiol Rep 2017; 5:5/11/e13297. [PMID: 28576853 PMCID: PMC5471436 DOI: 10.14814/phy2.13297] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/01/2023] Open
Abstract
Aerobic exercise confers many health benefits. However, numerous reports have shown that acute aerobic exercise can injure the heart. We tested the general hypothesis that acute moderate‐intensity exercise in rodents induces cardiomyocyte damage and stimulates mesenchymal stem cells (MSCs) to increase paracrine‐mediated protective effects on cardiomyocytes. A single session of treadmill running (13 m/min, 0% grade, for 45 min) in untrained C57BL/6 male mice (n = 18) increased cleaved poly ADP‐ribose polymerase (PARP), a marker of apoptosis, in the myocardium 24 h postexercise. Microarray analysis of mouse myocardium identified 11 relevant apoptotic genes and several shifts in matrix remodeling transcripts over the postexercise window. Postexercise cardiomyocyte death was recapitulated in neonatal rat cardiomyocytes (NRCMs) by culturing cells in 2% plasma harvested from exercised rats. The increased cell death observed in exercise‐treated NRCMs was attenuated by β‐adrenergic blockade, but not antioxidant treatment. MSC survival, proliferation, and chemotaxis showed no significant differences between sedentary and exercise plasma conditions, despite increased IL‐6, TNF‐α, IL‐1β, and IFN‐γ secretions from MSCs treated with exercise plasma. NRCM survival was increased nearly 500% when cocultured with MSCs, but this effect was not altered under exercise plasma culture conditions. Our results suggest acute moderate‐intensity aerobic treadmill running in exercise‐naïve rodents induces temporal cardiomyocyte death due to plasma‐borne factors, namely, catecholaminergic stress. Even though exercise conditions prompt an inflammatory response in MSCs, the exercise milieu does not alter the MSC‐protective phenotype on cardiomyocytes.
Collapse
Affiliation(s)
- Maria F Arisi
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Roxanne Sebeny
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Anbin Mu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bart C De Jonghe
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Joseph R Libonati
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
14
|
Sasso GRDS, Florencio-Silva R, Simões RS, Baracat MCP, Soares Júnior JM, Baracat EC. Elevated serum osteoprotegerin levels in women: friend or foe? Rev Assoc Med Bras (1992) 2016; 61:524-9. [PMID: 26841162 DOI: 10.1590/1806-9282.61.06.524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 07/08/2015] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION osteoprotegerin has emerged as a new candidate for the treatment of osteoporosis. However, high levels of osteoprotegerin have been linked to vascular calcification, an independent and well-defined risk factor for cardiovascular disease (CVD) and mortality. Thus, the action of osteoprotegerin in these situations has been questioned. OBJECTIVE to evaluate the effect of osteoprotegerin (OPG) on the human body, especially in bone tissue and in vascular diseases. METHODS the scientific databases consulted were PubMed-Medline and Cochrane, using keywords (MeSH terms) grouped into the following syntaxes: (Osteoprotegerin OR Osteoclastogenesis Inhibitory Factor OR Receptors, Tumor Necrosis Factor, Member 11b OR Tumor Necrosis Factor Receptor Superfamily, Member 11b OR FDCR-1 Protein OR FDCR 1 Protein OR OCIF Protein OR Follicular Dendritic Cell-Derived Receptor-1) AND (Bones AND Bone OR Bones AND Bone Tissue OR Bones OR Bone Tissue OR Cardiovascular Diseases). RESULTS Osteoprotegerin is present in various organs and binds to two ligands: nuclear factor kB (RANKL) related to the differentiation of osteoclasts, and tumor necrosis factor related to the apoptosis-inducing ligand (TRAIL). OPG inhibits the regulation effects of nuclear factor kB on inflammation and on the skeletal and vascular systems, preventing the apoptosis induced by TRAIL, being related to the preservation of bone tissue. CONCLUSION a deeper knowledge of the mechanisms involved in the association between OPG serum levels, bone integrity and cardiovascular disease can provide important data for future therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Ricardo Santos Simões
- Department of Obstetrics and Gynecology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - José Maria Soares Júnior
- Department of Obstetrics and Gynecology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
15
|
From bones to blood pressure, developing novel biologic approaches targeting the osteoprotegein pathway for pulmonary vascular disease. Pharmacol Ther 2016; 169:78-82. [PMID: 27373854 PMCID: PMC5243145 DOI: 10.1016/j.pharmthera.2016.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Abstract
Osteoprotegerin (tnfsf11b, OPG) is a soluble member of the TNF superfamily originally described as an important regulator of osteoclastogenesis almost 20 years ago. OPG is a heparin-binding secreted glycoprotein that exists as a 55–62 kDa monomer or a 110–120 kDa disulphide-linked homodimer. Acting as a soluble decoy receptor for RANKL, OPG actively regulates RANK signalling, and thereby osteoclastogenesis. OPG has subsequently been shown to also be a decoy receptor TNF related apoptosis inducing-ligand (tnfsf10, TRAIL, Apo2L). TRAIL is a type II transmembrane protein that is widely expressed in a variety of human tissues, including the spleen, lung, and prostate. Through binding to TRAIL, OPG can inhibit TRAIL-induced apoptosis of cancer cells. More recently, OPG has been demonstrated to be secreted by, and influence, vascular smooth muscle cells phenotype particularly related to vascular calcification and pulmonary vascular remodelling. In pulmonary artery smooth muscle cell (PASMC) suppression of BMP, induction of 5-HT and IL-1 signalling have been shown to stimulate the release of OPG in vitro, which causes cell migration and proliferation. Patients with idiopathic PAH (IPAH) demonstrate increased circulating and tissue levels of OPG, and circulating serum levels predict survival. In pre-clinical models, OPG levels correlate with disease severity. Since OPG is a naturally circulating protein, we are investigating the potential of novel biologic antibody therapies to rescue PAH phenotype in disease models. Further pre-clinical and mechanistic data are forthcoming, but we believe current published data identify OPG as an exciting and novel therapeutic target in PAH.
Collapse
|
16
|
Lu J, Liu F, Liu D, Du H, Hao J, Yang X, Cui W. Amlodipine and atorvastatin improved hypertensive cardiac hypertrophy through regulation of receptor activator of nuclear factor kappa B ligand/receptor activator of nuclear factor kappa B/osteoprotegerin system in spontaneous hypertension rats. Exp Biol Med (Maywood) 2016; 241:1237-49. [PMID: 26908571 DOI: 10.1177/1535370216630180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 12/23/2015] [Indexed: 11/16/2022] Open
Abstract
The present study aims to study the role of receptor activator of nuclear factor kappa B ligand/receptor activator of nuclear factor kappa B/osteoprotegerin (RANKL/RANK/OPG) system in cardiac hypertrophy in a spontaneous hypertension rat (SHR) model and the effects of amlodipine and atorvastatin intervention. Thirty-six-week-old male SHRs were randomly divided into four groups: 1) SHR control group; 2) amlodipine alone (10 mg/kg/d) group, 3) atorvastatin alone (10 mg/kg/d) group, 4) combination of amlodinpine and atorvastatin (10 mg/kg/d for each) group. Same gender, weight, and age of Wistar-Kyoto (WKY) rats with normal blood pressure were used as normal control. Drugs were administered by oral gavage over 12 weeks. The thicknesses of left ventricle walls, left ventricle weight, and cardiac function were measured by transthoracic echocardiography. Left ventricular pressure and function were assessed by hemodynamic examination. Cardiomyocyte hypertrophy and collagen accumulation in cardiac tissue were measured by hematoxylin and eosin (HE) and Masson staining, respectively. The hydroxyproline content of cardiac tissue was examined by biochemistry technique. RANKL, RANK and OPG mRNA, protein expression and tissue localization were studied by RT-PCR, Immunohistochemistry and Western blot. Treatment with amlodipine or atorvastatin alone significantly decreased left ventricular mass index, cardiomyocyte cross-sectional area and interstitial fibrosis in SHR (each P < 0.05). Moreover, combined amlodipine and atorvastatin treatment induced significant reversal of left ventricular hypertrophy and decreased cardiomyocyte cross-sectional area and interstitial fibrosis in SHR to a greater extent than each agent alone (P < 0.05). Compared with WKY rats, the myocardial expression of RANKL, RANK, and OPG was increased. Both amlodipine and atorvastatin reduced RANKL, RANK, and OPG expression, with the best effects seen with the combination. Based on our results, activation of the RANKL/RANK/OPG system may be an important factor leading to ventricular remodeling in SHR rats. Amlodipine and atorvastatin could improve ventricular remodeling in SHR rats through intervention with the RANKL/RANK/OPG system.
Collapse
Affiliation(s)
- Jingchao Lu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Hebei 050000, China
| | - Fan Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Hebei 050000, China
| | - Demin Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Hebei 050000, China
| | - Hong Du
- Department of Cardiology, The Second Hospital of Hebei Medical University, Hebei 050000, China
| | - Jie Hao
- Department of Cardiology, The Second Hospital of Hebei Medical University, Hebei 050000, China
| | - Xiuchun Yang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Hebei 050000, China
| | - Wei Cui
- Department of Cardiology, The Second Hospital of Hebei Medical University, Hebei 050000, China
| |
Collapse
|
17
|
Hao Y, Tsuruda T, Sekita-Hatakeyama Y, Kurogi S, Kubo K, Sakamoto S, Nakamura M, Udagawa N, Sekimoto T, Hatakeyama K, Chosa E, Asada Y, Kitamura K. Cardiac hypertrophy is exacerbated in aged mice lacking the osteoprotegerin gene. Cardiovasc Res 2016; 110:62-72. [PMID: 26825553 DOI: 10.1093/cvr/cvw025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/22/2016] [Indexed: 02/06/2023] Open
Abstract
AIMS Osteoprotegerin (OPG) may play a role in the progression of cardiac hypertrophy and heart failure. However, its pathophysiological role in changes in cardiac structure and function with ageing remains to be elucidated. METHODS AND RESULTS We conducted experiments using 2.5- and 12-month-old OPG(-/-) mice and age-matched wild-type (WT) mice and compared the morphology and function of the left ventricle (LV). Both 2.5- and 12-month-old OPG(-/-) mice showed a higher systolic blood pressure and a greater heart weight/body weight ratio than age-matched WT mice. Twelve-month-old OPG(-/-) mice had a significantly larger LV chamber and reduced wall thickness compared with age-matched WT mice, and contractile function was decreased. The morphological differences were accompanied by an increase in the number of apoptotic cells and activation of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) in the LV of 12-month-old OPG(-/-) mice. Correspondingly, OPG small interfering RNA induced the expressions of TRAIL and cleaved caspase-3 in cultured cardiac myocytes. In addition, these mice revealed a decrease in interstitial fibrosis, activation of matrix metalloproteinase (MMP)-2 and tissue inhibitors of MMP-1 and -2, and inactivation of procollagen α1 synthesis. Moreover, intraperitoneal administration of recombinant OPG to either 2.5- or 12-month-old OPG(-/-) mice for 28 days led to partial improvement of LV structure and function without affecting systolic blood pressure. CONCLUSION These results suggest that OPG plays a role in preserving myocardial structure and function with ageing through a reduction in apoptosis and preservation of the matrix structure. In addition, this appears to be independent of effects on the vasculature.
Collapse
Affiliation(s)
- Yilin Hao
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Toshihiro Tsuruda
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yoko Sekita-Hatakeyama
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Syuji Kurogi
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Keishi Kubo
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Sumiharu Sakamoto
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Midori Nakamura
- Department of Biochemistry, Matsumoto Dental University, Nagano 399-0781, Japan
| | - Nobuyuki Udagawa
- Department of Biochemistry, Matsumoto Dental University, Nagano 399-0781, Japan
| | - Tomohisa Sekimoto
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Kinta Hatakeyama
- Department of Diagnostic Pathology, Nara Medical University, Nara 634-0813, Japan
| | - Etsuo Chosa
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yujiro Asada
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Kazuo Kitamura
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, University of Miyazaki, Miyazaki 889-1692, Japan
| |
Collapse
|
18
|
Friões F, Laszczynska O, Almeida PB, Silva N, Guimarães JT, Omland T, Azevedo A, Bettencourt P. Prognostic Value of Osteoprotegerin in Acute Heart Failure. Can J Cardiol 2015; 31:1266-71. [DOI: 10.1016/j.cjca.2015.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 04/03/2015] [Accepted: 04/03/2015] [Indexed: 12/20/2022] Open
|
19
|
Sleep-disordered breathing is associated with depletion of circulating endothelial progenitor cells and elevation in pulmonary arterial pressure in patients with decompensated systolic heart failure. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2015; 12:424-30. [PMID: 26346897 PMCID: PMC4554780 DOI: 10.11909/j.issn.1671-5411.2015.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 01/13/2015] [Accepted: 04/03/2015] [Indexed: 11/25/2022]
Abstract
Background Sleep-disordered breathing (SDB) is known to occur frequently in and may predict worsening progression of patients with congestive heart failure (CHF). SDB is also known to play an important role in the development of idiopathic pulmonary arterial hypertension (PAH) via inducing endothelial dysfunction and vascular remodeling, a pathological process that can be significantly influenced by factors such as osteoprotegerin (OPG) and endothelial progenitor cells (EPCs). The objective of this study is to determine if CHF with SDB is associated with changes in OPG, EPCs, and PAH. Methods EPCs were isolated, cultured, and quantified from CHF patients with SDB (n = 52), or without SDB (n = 68). OPG and N-terminal pro-brain natriuretic peptide (NT-proBNP) from each group was analyzed and correlated with EPCs and the mean pulmonary artery pressure (mPAP) measured by right heart catheterization. Results A significant decrease in circulating EPCs (29.30 ± 9.01 vs. 45.17 ± 10.51 EPCs/× 200 field; P < 0.05) was found in CHF patients with SDB compared to those without SDB. Both OPG (789.83 ± 89.38 vs. 551.29 ± 42.12 pg/mL; P < 0.05) and NT-proBNP (5946.50 ± 1434.50 vs. 3028.60 ± 811.90 ng/mL; P < 0.05) were also significantly elevated in SDB CHF patients who also had significantly elevated mPAP (50.2 ± 9.5 vs. 36.4 ± 4.1 mm Hg; P < 0.05). EPC numbers correlated inversely with the episodes of apnea and hypopnea per hour (RDI, r = –0.45, P = 0.037) and blood level of OPG (r = –0.53, P = 0.011). Although NT-proBNP was also increased significantly in patients with SDB, it had no correlation with either EPCs or RDI. Conclusions SDB due to hypoxemia from decompensated CHF is associated with (1) OPG elevation, (2) EPC depletion, and (3) mPAP elevation. The inverse relationship of circulating OPG with EPCs suggests a likely mechanism for hypoxemia and OPG in the development of pulmonary vascular dysfunction via depleting EPCs, thus worsening prognosis of CHF.
Collapse
|
20
|
Zahid W, Bergestuen D, Haugaa KH, Ueland T, Thiis-Evensen E, Aukrust P, Fosse E, Edvardsen T. Myocardial Function by Two-Dimensional Speckle Tracking Echocardiography and Activin A May Predict Mortality in Patients with Carcinoid Intestinal Disease. Cardiology 2015; 132:81-90. [PMID: 26111973 DOI: 10.1159/000431076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/04/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Myocardial fibrosis causes deterioration of myocardial function in carcinoid intestinal disease (CID). We assessed the ability of myocardial function and various biomarkers to predict mortality in patients with CID. METHODS A total of 71 patients with small intestinal carcinoid tumours were included, and underwent echocardiography at baseline. Systolic function was assessed by two-dimensional speckle tracking echocardiography as left ventricular (LV) and right ventricular (RV) strain, and as mitral annular displacement (MAD), by tissue Doppler imaging. Parameters of diastolic function, the amount of liver metastases, and various biomarkers were also analysed. RESULTS During 1,274 ± 368 days of follow-up, 18 patients (25%) died. Of the 53 survivors, 46 patients (87%) were available for follow-up echocardiography. Baseline LV strain and MAD was reduced in those who died compared to those who survived (p < 0.001). Baseline plasma levels of activin A were markedly higher in patients who died during follow-up than those who survived (p = 0.001). In multivariate Cox hazard models (A, B, C and D), LV strain, age, the amount of liver metastases, MAD, and activin A were independent predictors of mortality. CONCLUSION Assessment of myocardial function by echocardiography, and the biomarker activin A, can be very useful in the risk stratification of patients with CID. © 2015 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Wasim Zahid
- Department of Cardiology and Center for Cardiological Innovation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Ueland T, Gullestad L, Nymo SH, Yndestad A, Aukrust P, Askevold ET. Inflammatory cytokines as biomarkers in heart failure. Clin Chim Acta 2014; 443:71-7. [PMID: 25199849 DOI: 10.1016/j.cca.2014.09.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/31/2014] [Accepted: 09/01/2014] [Indexed: 12/18/2022]
Abstract
Inflammation has been implicated in the pathogenesis of heart failure (HF). In addition to their direct involvement as mediators in the pathogenesis of HF, inflammatory cytokines and related mediators could also be suitable markers for risk stratification and prognostication in HF patients. Many reports have suggested that inflammatory cytokines may predict adverse outcome in these patients. However, most studies have been limited in sample size and lacking full adjustment with the most recent and strongest biochemical predictor such as NT-proBNP and high sensitivity troponins. Furthermore, a number of pre-analytical and analytical aspects of cytokine measurements may limit their use as biomarkers. This review focuses on technical, informative and practical considerations concerning the clinical use of inflammatory cytokines as prognostic biomarkers in HF. We focus on the predictive value of tumor necrosis factor (TNF) α, the TNF family receptors sTNFR1 and osteoprotegerin, interleukin (IL)-6 and its receptor gp130, the chemokines MCP-1, IL-8, CXCL16 and CCL21 and the pentraxin PTX-3 in larger prospective fully adjusted studies. No single inflammatory cytokine provides sufficient discrimination to justify the transition to everyday clinical use as a prognosticator in HF. However, while subjecting potential new HF markers to rigorous comparisons with "gold-standard" markers, such as NT-proBNP, using receiver operating characteristics (ROCs) and HF risk models, makes sense from a clinical standpoint, it may pose a threat to a broadening of mechanistic insight if the new markers are dismissed solely on account of lower statistical power.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Faculty of Medicine, University of Oslo, Norway; K.G. Jebsen Inflammatory Research Center, University of Oslo, Norway; KG Jebsen Thrombosis Research and Expertise Center, N-9037 Tromsø, Norway.
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Norway; Faculty of Medicine, University of Oslo, Norway; KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Norway
| | - Ståle H Nymo
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Faculty of Medicine, University of Oslo, Norway; K.G. Jebsen Inflammatory Research Center, University of Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Norway; Faculty of Medicine, University of Oslo, Norway; K.G. Jebsen Inflammatory Research Center, University of Oslo, Norway; KG Jebsen Thrombosis Research and Expertise Center, N-9037 Tromsø, Norway
| | - Erik T Askevold
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Norway; Clinic for Internal Medicine, Lovisenberg Diakonale Hospital, N-0027 Oslo, Norway
| |
Collapse
|
22
|
Kalaycıoğlu E, Gökdeniz T, Aykan AÇ, Hatem E, Gürsoy MO, Ören A, Yaman H, Karadeniz AG, Çelik Ş. Osteoprotegerin is associated with subclinical left ventricular systolic dysfunction in diabetic hypertensive patients: a speckle tracking study. Can J Cardiol 2014; 30:1529-34. [PMID: 25442456 DOI: 10.1016/j.cjca.2014.08.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 08/11/2014] [Accepted: 08/19/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Recently, the role of osteoprotegerin (OPG) in the pathogenesis of heart failure through different mechanisms has received much attention. Subclinical changes in left ventricular (LV) function can be identified using quantification of myocardial strain, and global longitudinal strain (GLS) is a superior predictor of outcomes than ejection fraction. We hypothesized that increased OPG levels could predict subclinical LV systolic dysfunction in treated diabetic hypertensive patients with preserved LV ejection fraction. METHODS The study was composed of 86 diabetic hypertensive and 30 nondiabetic hypertensive patients. All patients underwent echocardiography and venous blood samples were taken for determination of OPG. The relation between OPG levels and LV GLS was investigated using 2-dimensional speckle tracking echocardiography. RESULTS Diabetic hypertensive patients had higher diastolic peak early/early diastolic tissue velocity and lower systolic tissue velocity, GLS, GLS rate systolic, and GLS rate early diastolic than nondiabetic hypertensive patients (P = 0.009, P = 0.049, P < 0.001, P = 0.004, and P < 0.001, respectively). Diabetic hypertensive patients were divided into 2 groups according to median GLS value (> 18.5 and ≤ 18.5). The patients with GLS ≤ 18.5 had higher diastolic blood pressure (mm Hg; P = 0.048), OPG (pmol/L; P < 0.001), and hemoglobin A1c (%; P = 0.042) values than those with GLS > 18.5. In multivariate logistic regression analysis, OPG was found to be an independent predictor of impaired GLS (P = 0.001). Receiver operating characteristic curve analysis revealed that OPG values of > 6.45 (pmol/L) identified the patients with GLS ≤ 18.5. CONCLUSIONS Plasma OPG values could predict subclinical LV systolic dysfunction in diabetic hypertensive patients.
Collapse
Affiliation(s)
- Ezgi Kalaycıoğlu
- Department of Cardiology, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey.
| | - Tayyar Gökdeniz
- Department of Cardiology, Kafkas University, Faculty of Medicine, Kars, Turkey
| | - Ahmet Çağrı Aykan
- Department of Cardiology, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey
| | - Engin Hatem
- Department of Cardiology, Erzurum Regional Training and Research Hospital, Erzurum, Turkey
| | - Mustafa Ozan Gürsoy
- Department of Cardiology, Gaziemir Salih Nevvar İşgören State Hospital, İzmir, Turkey
| | - Asım Ören
- Department of Medical Biochemistry, Karadeniz Technical University Faculty of Medicine, Trabzon, Turkey
| | - Hüseyin Yaman
- Department of Medical Biochemistry, Karadeniz Technical University Faculty of Medicine, Trabzon, Turkey
| | - Ayşe Gül Karadeniz
- Department of Radiology, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey
| | - Şükrü Çelik
- Department of Cardiology, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey
| |
Collapse
|
23
|
Sluijter JPG, Condorelli G, Davidson SM, Engel FB, Ferdinandy P, Hausenloy DJ, Lecour S, Madonna R, Ovize M, Ruiz-Meana M, Schulz R, Van Laake LW. Novel therapeutic strategies for cardioprotection. Pharmacol Ther 2014; 144:60-70. [PMID: 24837132 DOI: 10.1016/j.pharmthera.2014.05.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 12/12/2022]
Abstract
The morbidity and mortality from ischemic heart disease (IHD) remain significant worldwide. The treatment for acute myocardial infarction has improved over the past decades, including early reperfusion of occluded coronary arteries. Although it is essential to re-open the artery as soon as possible, paradoxically this leads to additional myocardial injury, called acute ischemia-reperfusion injury (IRI), for which currently no effective therapy is available. Therefore, novel therapeutic strategies are required to protect the heart from acute IRI in order to reduce myocardial infarction size, preserve cardiac function and improve clinical outcomes in patients with IHD. In this review article, we will first outline the pathophysiology of acute IRI and review promising therapeutic strategies for cardioprotection. These include novel aspects of mitochondrial function, epigenetics, circadian clocks, the immune system, microvesicles, growth factors, stem cell therapy and gene therapy. We discuss the therapeutic potential of these novel cardioprotective strategies in terms of pharmacological targeting and clinical application.
Collapse
Affiliation(s)
- Joost P G Sluijter
- Department of Cardiology, University Medical Center Utrecht, The Netherlands; ICIN, Netherlands Heart Institute, Utrecht, The Netherlands
| | | | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Derek J Hausenloy
- Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, South Africa
| | - Rosalinda Madonna
- Department of Neurosciences and Imaging, Institute of Cardiology, University of Chieti, Chieti, Italy
| | - Michel Ovize
- Service d'Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, France; Inserm U1060-CarMeN, CIC de Lyon, Université Claude Bernard Lyon, Lyon, France
| | - Marisol Ruiz-Meana
- Laboratori Cardiologia, Vall d'Hebron Institut de Recerca, Universitat Autonoma de Barcelona, Spain
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig Universität, Gießen, Germany
| | - Linda W Van Laake
- Department of Cardiology, University Medical Center Utrecht, The Netherlands.
| | | |
Collapse
|
24
|
The NO/ONOO-cycle as the central cause of heart failure. Int J Mol Sci 2013; 14:22274-330. [PMID: 24232452 PMCID: PMC3856065 DOI: 10.3390/ijms141122274] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 01/08/2023] Open
Abstract
The NO/ONOO-cycle is a primarily local, biochemical vicious cycle mechanism, centered on elevated peroxynitrite and oxidative stress, but also involving 10 additional elements: NF-κB, inflammatory cytokines, iNOS, nitric oxide (NO), superoxide, mitochondrial dysfunction (lowered energy charge, ATP), NMDA activity, intracellular Ca(2+), TRP receptors and tetrahydrobiopterin depletion. All 12 of these elements have causal roles in heart failure (HF) and each is linked through a total of 87 studies to specific correlates of HF. Two apparent causal factors of HF, RhoA and endothelin-1, each act as tissue-limited cycle elements. Nineteen stressors that initiate cases of HF, each act to raise multiple cycle elements, potentially initiating the cycle in this way. Different types of HF, left vs. right ventricular HF, with or without arrhythmia, etc., may differ from one another in the regions of the myocardium most impacted by the cycle. None of the elements of the cycle or the mechanisms linking them are original, but they collectively produce the robust nature of the NO/ONOO-cycle which creates a major challenge for treatment of HF or other proposed NO/ONOO-cycle diseases. Elevated peroxynitrite/NO ratio and consequent oxidative stress are essential to both HF and the NO/ONOO-cycle.
Collapse
|
25
|
Li C, Chen S, Song J, Liu H, Gu W, Ai H, Zhao B, Zhang S. Molecular cloning and characterization of TNFSF14 (LIGHT) and its receptor TNFRSF14 (HVEM) in guinea pig (Cavia porcellus). Gene 2013; 526:374-84. [PMID: 23732292 DOI: 10.1016/j.gene.2013.05.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 05/06/2013] [Accepted: 05/15/2013] [Indexed: 01/05/2023]
Abstract
LIGHT (lymphotoxin-related inducible ligand that competes with herpes simplex virus (HSV) glycoprotein D for herpesvirus entry mediator on T cells) is a member of the tumor necrosis factor (TNF) ligand superfamily, which plays important roles in inflammatory and immune responses. In the present study, the cDNAs of guinea pig (Cavia porcellus) LIGHT (designated as gpLIGHT) and its receptor herpes virus entry mediator (designated as gpHVEM) were amplified from spleen by reverse transcription polymerase chain reaction (RT-PCR). The ORFs of gpLIGHT and gpHVEM cover 726 and 861 bp, encoding predicted proteins with 241 and 286 aas, respectively. The three-dimensional (3D) structure, phylogenetic relationships, and characterization of both genes were also analyzed. We also generated a 3D model to verify interaction between the two proteins. Real-time quantitative PCR (qPCR) analysis revealed that both LIGHT and HVEM are constitutively expressed in guinea pig various tissues. A fusion protein SUMO (Small Ubiquitin-like Modifier)-gpsLIGHT (the soluble mature part of gpLIGHT) was efficiently expressed in Escherichia coli BL21 (DE3) and purified using metal chelate affinity chromatography (Ni-NTA). Laser scanning confocal microscopy (LSCM) showed that gpsLIGHT can bind its receptors on T cells. The LIGHT-HVEM signaling pathway plays an important role in the immune system, and our results might provide a platform for further research into the effects of LIGHT and HVEM.
Collapse
Affiliation(s)
- Chunlan Li
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, Life Sciences College, Nanjing Normal University, Nanjing 210046, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Matsuo J, Nakamura S, Ito A, Yamazaki T, Ishida K, Hayashi Y, Yoshida M, Takahashi K, Sekizuka T, Takeuchi F, Kuroda M, Nagai H, Hayashida K, Sugimoto C, Yamaguchi H. Protochlamydia induces apoptosis of human HEp-2 cells through mitochondrial dysfunction mediated by chlamydial protease-like activity factor. PLoS One 2013; 8:e56005. [PMID: 23409113 PMCID: PMC3569409 DOI: 10.1371/journal.pone.0056005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 01/04/2013] [Indexed: 11/22/2022] Open
Abstract
Obligate amoebal endosymbiotic bacterium Protochlamydia with ancestral pathogenic chlamydial features evolved to survive within protist hosts, such as Acanthamoba, 0.7–1.4 billion years ago, but not within vertebrates including humans. This observation raises the possibility that interactions between Protochlamydia and human cells may result in a novel cytopathic effect, leading to new insights into host-parasite relationships. Previously, we reported that Protochlamydia induces apoptosis of the immortalized human cell line, HEp-2. In this study, we attempted to elucidate the molecular mechanism underlying this apoptosis. We first confirmed that, upon stimulation with the bacteria, poly (ADP-ribose) polymerase (PARP) was cleaved at an early stage in HEp-2 cells, which was dependent on the amount of bacteria. A pan-caspase inhibitor and both caspase-3 and -9 inhibitors similarly inhibited the apoptosis of HEp-2 cells. A decrease of the mitochondrial membrane potential was also confirmed. Furthermore, lactacystin, an inhibitor of chlamydial protease-like activity factor (CPAF), blocked the apoptosis. Cytochalasin D also inhibited the apoptosis, which was dependent on the drug concentration, indicating that bacterial entry into cells was required to induce apoptosis. Interestingly, Yersinia type III inhibitors (ME0052, ME0053, and ME0054) did not have any effect on the apoptosis. We also confirmed that the Protochlamydia used in this study possessed a homologue of the cpaf gene and that two critical residues, histidine-101 and serine-499 of C. trachomatis CPAF in the active center, were conserved. Thus, our results indicate that after entry, Protochlamydia-secreted CPAF induces mitochondrial dysfunction with a decrease of the membrane potential, followed by caspase-9, caspase-3 and PARP cleavages for apoptosis. More interestingly, because C. trachomatis infection can block the apoptosis, our finding implies unique features of CPAF between pathogenic and primitive chlamydiae.
Collapse
Affiliation(s)
- Junji Matsuo
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shinji Nakamura
- Division of Biomedical Imaging Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Ito
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Tomohiro Yamazaki
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kasumi Ishida
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yasuhiro Hayashi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mitsutaka Yoshida
- Division of Ultrastructural Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kaori Takahashi
- Division of Ultrastructural Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tsuyoshi Sekizuka
- Pathogen Genomics Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Fumihiko Takeuchi
- Pathogen Genomics Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Makoto Kuroda
- Pathogen Genomics Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Hiroki Nagai
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kyoko Hayashida
- Research Center for Zoonosis Control, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Chihiro Sugimoto
- Research Center for Zoonosis Control, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Hiroyuki Yamaguchi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
- * E-mail:
| |
Collapse
|
27
|
Ruiz-Esparza GU, Flores-Arredondo JH, Segura-Ibarra V, Torre-Amione G, Ferrari M, Blanco E, Serda RE. The physiology of cardiovascular disease and innovative liposomal platforms for therapy. Int J Nanomedicine 2013; 8:629-40. [PMID: 23413209 PMCID: PMC3572823 DOI: 10.2147/ijn.s30599] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Heart disease remains the major cause of death in males and females, emphasizing the need for novel strategies to improve patient treatment and survival. A therapeutic approach, still in its infancy, is the development of site-specific drug-delivery systems. Nanoparticle-based delivery systems, such as liposomes, have evolved into robust platforms for site-specific delivery of therapeutics. In this review, the clinical impact of cardiovascular disease and the pathophysiology of different subsets of the disease are described. Potential pathological targets for therapy are introduced, and promising advances in nanotherapeutic cardiovascular applications involving liposomal platforms are presented.
Collapse
|