1
|
Zhao X, Wang Z, Wang L, Jiang T, Dong D, Sun M. The PINK1/Parkin signaling pathway-mediated mitophagy: a forgotten protagonist in myocardial ischemia/reperfusion injury. Pharmacol Res 2024; 209:107466. [PMID: 39419133 DOI: 10.1016/j.phrs.2024.107466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/12/2024] [Accepted: 10/12/2024] [Indexed: 10/19/2024]
Abstract
Myocardial ischemia causes extensive damage, further exacerbated by reperfusion, a phenomenon called myocardial ischemia/reperfusion injury (MIRI). Nowadays, the pathological mechanisms of MIRI have received extensive attention. Oxidative stress, multiple programmed cell deaths, inflammation and others are all essential pathological mechanisms contributing to MIRI. Mitochondria are the energy supply centers of cells. Numerous studies have found that abnormal mitochondrial function is an essential "culprit" of MIRI, and mitophagy mediated by the phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1)/Parkin signaling pathway is an integral part of maintaining mitochondrial function. Therefore, exploring the association between the PINK1/Parkin signaling pathway-mediated mitophagy and MIRI is crucial. This review will mainly summarize the crucial role of the PINK1/Parkin signaling pathway-mediated mitophagy in MIR-induced several pathological mechanisms and various potential interventions that affect the PINK1/Parkin signaling pathway-mediated mitophagy, thus ameliorating MIRI.
Collapse
Affiliation(s)
- Xiaopeng Zhao
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China.
| | - Zheng Wang
- School of Medicine, Qilu Institute of Technology, Jinan 250200, China.
| | - Lijie Wang
- Department of Cardiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110033, China.
| | - Tao Jiang
- Rehabilitation Medicine Center, The Second Hospital of Shandong University, Jinan 250033, China.
| | - Dan Dong
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, China.
| |
Collapse
|
2
|
Wasim R, Singh A, Islam A, Mohammed S, Anwar A, Mahmood T. High Mobility Group Box 1 and Cardiovascular Diseases: Study of Act and Connect. Cardiovasc Toxicol 2024; 24:1268-1286. [PMID: 39242448 DOI: 10.1007/s12012-024-09919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Cardiovascular disease is the deadly disease that can result in sudden death, and inflammation plays an important role in its onset and progression. High mobility group box 1 (HMGB1) is a nuclear protein that regulates transcription, DNA replication, repair, and nucleosome assembly. HMGB1 is released passively by necrotic tissues and actively secreted by stressed cells. Extracellular HMGB1 functions as a damage associated molecular patterns molecule, producing numerous redox forms that induce a range of cellular responses by binding to distinct receptors and interactors, including tissue inflammation and regeneration. Extracellular HMGB1 inhibition reduces inflammation and is protective in experimental models of myocardial ischemia/reperfusion damage, myocarditis, cardiomyopathies caused by mechanical stress, diabetes, bacterial infection, or chemotherapeutic drugs. HMGB1 administration following a myocardial infarction followed by permanent coronary artery ligation improves cardiac function by stimulating tissue regeneration. HMGB1 inhibits contractility and produces hypertrophy and death in cardiomyocytes, while also stimulating cardiac fibroblast activity and promoting cardiac stem cell proliferation and differentiation. Maintaining normal nuclear HMGB1 levels, interestingly, protects cardiomyocytes from apoptosis by limiting DNA oxidative stress, and mice with HMGB1cardiomyocyte-specific overexpression are partially protected from cardiac injury. Finally, elevated levels of circulating HMGB1 have been linked to human heart disease. As a result, following cardiac damage, HMGB1 elicits both detrimental and helpful responses, which may be due to the formation and stability of the various redox forms, the particular activities of which in this context are mostly unknown. This review covers recent findings in HMGB1 biology and cardiac dysfunction.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacy, Integral University, Lucknow, 226026, India.
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Aditya Singh
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Anas Islam
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Saad Mohammed
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Aamir Anwar
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Tarique Mahmood
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
3
|
Ngcobo NN, Sibiya NH. The role of high mobility group box-1 on the development of diabetes complications: A plausible pharmacological target. Diab Vasc Dis Res 2024; 21:14791641241271949. [PMID: 39271468 PMCID: PMC11406611 DOI: 10.1177/14791641241271949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Diabetes mellitus has emerged as a pressing global concern, with a notable increase in recent years. Despite advancements in treatment, existing medications struggle to halt the progression of diabetes and its associated complications. Increasing evidence underscores inflammation as a significant driver in the onset of diabetes mellitus. Therefore, perspectives on new therapies must consider shifting focus from metabolic stress to inflammation. High mobility group box (HMGB-1), a nuclear protein regulating gene expression, gained attention as an endogenous danger signal capable of sparking inflammatory responses upon release into the extracellular environment in the late 1990s. PURPOSE Given the parallels between inflammatory responses and type 2 diabetes (T2D) development, this review paper explores HMGB-1's potential involvement in onset and progression of diabetes complications. Specifically, we will review and update the understanding of HMGB-1 and its inflammatory pathways in insulin resistance, diabetic nephropathy, diabetic neuropathy, and diabetic retinopathy. CONCLUSIONS HMGB-1 and its receptors i.e. receptor for advanced glycation end-products (RAGE) and toll-like receptors (TLRs) present promising targets for antidiabetic interventions. Ongoing and future projects in this realm hold promise for innovative approaches targeting HMGB-1-mediated inflammation to ameliorate diabetes and its complications.
Collapse
Affiliation(s)
- Nokwanda N Ngcobo
- Discipline of Pharmaceutical Sciences, School of Health Science, University of KwaZulu-Natal, Durban, South Africa
| | - Ntethelelo H Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa
| |
Collapse
|
4
|
He DW, Liu DZ, Luo XZ, Chen CB, Lu CH, Na N, Huang F. HMGB1-RAGE axis contributes to myocardial ischemia/reperfusion injury via regulation of cardiomyocyte autophagy and apoptosis in diabetic mice. Biol Chem 2024; 405:167-176. [PMID: 37768929 DOI: 10.1515/hsz-2023-0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/22/2023] [Indexed: 09/30/2023]
Abstract
Patients with acute myocardial infarction complicated with diabetes are more likely to develop myocardial ischemia/reperfusion (I/R) injury (MI/RI) during reperfusion therapy. Both HMGB1 and RAGE play important roles in MI/RI. However, the specific mechanisms of HMGB1 associated with RAGE are not fully clarified in diabetic MI/RI. This study aimed to investigate whether the HMGB1-RAGE axis induces diabetic MI/RI via regulating autophagy and apoptosis. A db/db mouse model of MI/RI was established, where anti-HMGB1 antibody and RAGE inhibitor (FPS-ZM1) were respectively injected after 10 min of reperfusion. The results showed that treatment with anti-HMGB1 significantly reduced the infarct size, serum LDH, and CK-MB level. Similar situations also occurred in mice administrated with FPS-ZM1, though the HMGB1 level was unchanged. Then, we found that treatment with anti-HMGB1 or FPS-ZM1 performed the same effects in suppressing the autophagy and apoptosis, as reflected by the results of lower LAMP2 and LC3B levels, increased Bcl-2 level, reduced BAX and caspase-3 levels. Moreover, the Pink1/Parkin levels were also inhibited at the same time. Collectively, this study indicates that the HMGB1-RAGE axis aggravated diabetic MI/RI via apoptosis and Pink1/Parkin mediated autophagy pathways, and inhibition of HMGB1 or RAGE contributes to alleviating those adverse situations.
Collapse
Affiliation(s)
- De-Wei He
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - De-Zhao Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Xiao-Zhi Luo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Chuan-Bin Chen
- Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, 368 Yihai Avenue, Haikou 570216, Hainan, China
| | - Chuang-Hong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Na Na
- Department of Chemistry, Scripps Research Institute, No.10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning 530021, Guangxi, China
| |
Collapse
|
5
|
Guo X, Qu FX, Zhang JD, Zheng F, Xin Y, Wang R, Li JY, Li HY, Lu CH. Amygdalin and exercise training exert a synergistic effect in improving cardiac performance and ameliorating cardiac inflammation and fibrosis in a rat model of myocardial infarction. Appl Physiol Nutr Metab 2024; 49:360-374. [PMID: 37944128 DOI: 10.1139/apnm-2023-0135] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
This study investigated the effects of amygdalin (AMY, a cyanogenic glycoside widely distributed in the fruits and seeds of Rosaceae plants) on cardiac performance and ventricular remodeling in a rat model of myocardial infarction (MI). We also investigated whether the combination of AMY with exercise training (ExT) has a beneficial synergistic effect in treating MI rats. MI was induced by the ligation of the left anterior descending coronary artery in male SD rats. ExT or AMY treatment was started 1 week after MI and continued for 1 week (short-term) or 8 weeks (long-term). Cardiac function was evaluated by echocardiographic and hemodynamic parameters. Heart tissues were harvested and subjected to 2,3,5-triphenyl-tetrazolium chloride, Masson's trichrome, hematoxylin-eosin, and immunohistochemical staining. Gene expression was determined by quantitative polymerase chain reaction. Western blot gave a qualitative assessment of protein levels. AMY or ExT improved cardiac function and reduced infarct size in MI rats. AMY or ExT also suppressed myocardial fibrosis and attenuated inflammation in the infarct border zone of hearts from MI rats, as evidenced by inhibition of collagen deposition, inflammatory cell infiltration, and pro-inflammatory markers (interleukin 1β, interleukin 6, tumor necrosis factor-α, and cyclooxygenase 2). Notably, the effects of AMY combined with ExT were superior to those of AMY alone or ExT alone. Mechanistically, these beneficial functions were correlated with the inhibition of MI-induced activation of the transforming growth factor-β/Smad pathway. Collectively, AMY and ExT exert a synergistic effect on improving cardiac performance and ameliorating cardiac inflammation and fibrosis after MI, and the effects of long-term intervention were better than short-term intervention.
Collapse
Affiliation(s)
- Xiao Guo
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao 266034, Shandong, People's Republic of China
| | - Feng-Xia Qu
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao 266034, Shandong, People's Republic of China
| | - Ji-Dong Zhang
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, People's Republic of China
| | - Fa Zheng
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao 266034, Shandong, People's Republic of China
| | - Yue Xin
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao 266034, Shandong, People's Republic of China
| | - Rong Wang
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao 266034, Shandong, People's Republic of China
| | - Jing-Yuan Li
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao 266034, Shandong, People's Republic of China
| | - Hai-Ying Li
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao 266034, Shandong, People's Republic of China
| | - Chang-Hong Lu
- Heart Center, Qingdao Fuwai Cardiovascular Hospital, Qingdao 266034, Shandong, People's Republic of China
| |
Collapse
|
6
|
Tang J, Tam E, Song E, Xu A, Sweeney G. Crosstalk between myocardial autophagy and sterile inflammation in the development of heart failure. AUTOPHAGY REPORTS 2024; 3:2320605. [PMID: 40395524 PMCID: PMC11864620 DOI: 10.1080/27694127.2024.2320605] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 05/22/2025]
Abstract
Heart failure, a leading driver of global mortality, remains a topic of intense contemporary research interest due to the prevailing unmet need in cardiometabolic therapeutics. Numerous mechanisms with the potential to influence the onset and development of heart failure remain incompletely understood. Firstly, myocardial autophagy, which involves lysosomal degradation of damaged cellular components, confers context-dependent beneficial and detrimental effects. Secondly, sterile inflammation may arise following cardiac stress and exacerbate the progression of heart failure. Inflammation changes in a temporal manner and its onset must be adequately resolved to limit progression of heart failure. Mitochondria are an important factor in contributing to sterile inflammation by releasing damage associated molecular patterns (DAMPs) including mitochondrial DNA (mtDNA). Accordingly, this is one reason why the selective autophagy of mitochondria to maintain optimal function is important in determining cardiac function. In this review, we examine the increasing evidence suggesting crosstalk between autophagy and sterile inflammation together with their role in the development of heart failure. In particular, this is exemplified in the preclinical models of ischaemia/reperfusion injury and pressure overload induced heart failure. We also highlight potential therapeutic approaches focusing on autophagy and addressing sterile inflammation, aiming to enhance outcomes in heart failure.
Collapse
Affiliation(s)
- Jialing Tang
- Department of Biology, York University, Toronto, ON, Canada
| | - Eddie Tam
- Department of Biology, York University, Toronto, ON, Canada
| | - Erfei Song
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Metabolic and Bariatric Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aimin Xu
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
7
|
Wahid A, Wen J, Yang Q, Zhang Z, Zhao X, Tang X. Serum HMGB1 is a biomarker for acute myocardial infarction with or without heart failure. Clin Transl Sci 2023; 16:2299-2309. [PMID: 37775976 PMCID: PMC10651663 DOI: 10.1111/cts.13630] [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: 06/04/2023] [Revised: 07/25/2023] [Accepted: 08/23/2023] [Indexed: 10/01/2023] Open
Abstract
This study measured serum high mobility group box 1 (HMGB1) levels in patients with acute myocardial infarction (AMI) and/or heart failure (HF) and evaluated their relationship with peripheral inflammatory biomarkers and cardiac biomarkers, which have not been reported before. Of the patients, 55 had AMI without HF (AMI-HF ), 42 had AMI with HF (AMI+HF ), and 60 had HF without AMI (HF-AMI ) compared with 50 healthy controls. Blood samples were collected to assess serum HMGB1 levels and blood test-related inflammatory biomarkers (e.g., erythrocyte sedimentation rate [ESR], hs-CRP, uric acid, and white blood cell count) and cardiac biomarkers (e.g., MYO, cTnI, CKMB, CK, NT-proBNP, LDH, aspartate aminotransferase [AST], and alanine aminotransferase [ALT]). Compared to healthy controls, three groups of patients, especially those with AMI+HF , had significantly higher levels of serum HMGB1. All tested inflammatory biomarkers (except uric acid) were significantly positively correlated with HMGB1 in patients with AMI patients but not in patients with non-AMI. In addition, all tested cardiac biomarkers (except NT-proBNP in AMI-HF ) were significantly higher in patients with AMI than in control individuals. The levels of MYO, cTnI, CKMB, CK, AST, and ALT were not significantly changed in patients with HF-AMI compared to control individuals, but were still much lower than those in patients with AMI (except ALT). In all patients, the levels of NT-proBNP, and cTnI were significantly correlated with HMGB1 levels. Except for MYO, LDH, AST, and ALT, all cardiac biomarkers in AMI-HF and AMI+HF showed a significant correlation with HMGB1. Among risk factors, hypertension, diabetes, previous heart disease, and reduced left ventricular ejection fraction showed a significant correlation with HMGB1 in all disease groups.
Collapse
Affiliation(s)
- Abdul Wahid
- Department of Cardiology of the Third Xiang‐Ya HospitalCentral South UniversityChangshaHunanChina
| | - Juan Wen
- Department of Cardiology of the Third Xiang‐Ya HospitalCentral South UniversityChangshaHunanChina
| | - Qiong Yang
- Department of Cardiology of the Third Xiang‐Ya HospitalCentral South UniversityChangshaHunanChina
| | - Zhihui Zhang
- Department of Cardiology of the Third Xiang‐Ya HospitalCentral South UniversityChangshaHunanChina
| | - Xiexiong Zhao
- Department of Cardiology of the Third Xiang‐Ya HospitalCentral South UniversityChangshaHunanChina
| | - Xiaohong Tang
- Department of Cardiology of the Third Xiang‐Ya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
8
|
Ahmed OM, Saleh AS, Ahmed EA, Ghoneim MM, Ebrahim HA, Abdelgawad MA, Abdel-Gabbar M. Efficiency of Bone Marrow-Derived Mesenchymal Stem Cells and Hesperetin in the Treatment of Streptozotocin-Induced Type 1 Diabetes in Wistar Rats. Pharmaceuticals (Basel) 2023; 16:859. [PMID: 37375806 PMCID: PMC10303997 DOI: 10.3390/ph16060859] [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: 03/09/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) was established to be ameliorated by islet transplantation, but the shortage of the transplanted human islet tissue and the use of immunosuppressive drugs to inhibit the rejection of allogeneic grafts make this type of therapy is limited. Nowadays, therapy with stem cells is one of the most promising future treatments. This kind of therapy could have a profound impact on both replacement, as well as regenerative therapies, to improve or even cure various disorders, including diabetes mellitus. Flavonoids have also been shown to possess anti-diabetic effects. Thus, this study aims to evaluate the effectiveness of the bone marrow-derived mesenchymal stem cells (BM-MSCs) and hesperetin in the treatment of a T1DM rat model. T1DM was induced in male Wistar rats that had been starved for 16 h via intraperitoneal injection of STZ at a dose of 40 mg/kg body weight (b.wt.). After 10 days of STZ injection, the diabetic rats were allocated into four groups. The first diabetic animal group was considered a diabetic control, while the other three diabetic animal groups were treated for six weeks, respectively, with hesperetin (given orally at a dose of 20 mg/kg b.wt.), BM-MSCs (injected intravenously at a dose of 1 × 106 cells/rat/week), and their combination (hesperetin and BM-MSCs). The use of hesperetin and BM-MSCs in the treatment of STZ-induced diabetic animals significantly improved the glycemic state, serum fructosamine, insulin and C-peptide levels, liver glycogen content, glycogen phosphorylase, glucose-6-phosphatase activities, hepatic oxidative stress, and mRNA expressions of NF-κB, IL-1β, IL-10, P53, and Bcl-2 in pancreatic tissue. The study suggested the therapy with both hesperetin and BM-MSCs produced marked antihyperglycemic effects, which may be mediated via their potencies to ameliorate pancreatic islet architecture and insulin secretory response, as well as to decrease hepatic glucose output in diabetic animals. The improvement effects of hesperetin and BM-MSCs on the pancreatic islets of diabetic rats may be mediated via their antioxidant, anti-inflammatory, and antiapoptotic actions.
Collapse
Affiliation(s)
- Osama M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 62521, Egypt;
- Experimental Obesity and Diabetes Research Lab (EODRL), Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Ablaa S. Saleh
- Biochemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 62521, Egypt
| | - Eman A. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 62521, Egypt;
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, Riyadh 13713, Saudi Arabia
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| | - Mohammed Abdel-Gabbar
- Biochemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef 62521, Egypt
| |
Collapse
|
9
|
Lin Q, Kumar S, Kariyawasam U, Yang X, Yang W, Skinner JT, Gao WD, Johns RA. Human Resistin Induces Cardiac Dysfunction in Pulmonary Hypertension. J Am Heart Assoc 2023; 12:e027621. [PMID: 36927008 PMCID: PMC10111547 DOI: 10.1161/jaha.122.027621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 03/18/2023]
Abstract
Background Cardiac failure is the primary cause of death in most patients with pulmonary arterial hypertension (PH). As pleiotropic cytokines, human resistin (Hresistin) and its rodent homolog, resistin-like molecule α, are mechanistically critical to pulmonary vascular remodeling in PH. However, it is still unclear whether activation of these resistin-like molecules can directly cause PH-associated cardiac dysfunction and remodeling. Methods and Results In this study, we detected Hresistin protein in right ventricular (RV) tissue of patients with PH and elevated resistin-like molecule expression in RV tissues of rodents with RV hypertrophy and failure. In a humanized mouse model, cardiac-specific Hresistin overexpression was sufficient to cause cardiac dysfunction and remodeling. Dilated hearts exhibited reduced force development and decreased intracellular Ca2+ transients. In the RV tissues overexpressing Hresistin, the impaired contractility was associated with the suppression of protein kinase A and AMP-activated protein kinase. Mechanistically, Hresistin activation triggered the inflammation mediated by signaling of the key damage-associated molecular pattern molecule high-mobility group box 1, and subsequently induced pro-proliferative Ki67 in RV tissues of the transgenic mice. Intriguingly, an anti-Hresistin human antibody that we generated protected the myocardium from hypertrophy and failure in the rodent PH models. Conclusions Our data indicate that Hresistin is expressed in heart tissues and plays a role in the development of RV dysfunction and maladaptive remodeling through its immunoregulatory activities. Targeting this signaling to modulate cardiac inflammation may offer a promising strategy to treat PH-associated RV hypertrophy and failure in humans.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Santosh Kumar
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Udeshika Kariyawasam
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Xiaomei Yang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
- Department of AnesthesiologyQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Wei Yang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
- Department of Cardiovascular MedicineXiangya Hospital, Central South UniversityChangshaChina
| | - John T. Skinner
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Roger A. Johns
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| |
Collapse
|
10
|
Schoenfeld J, Roeh A, Holdenrieder S, von Korn P, Haller B, Krueger K, Falkai P, Halle M, Hasan A, Scherr J. High-mobility group box 1 protein, receptor for advanced glycation end products and nucleosomes increases after marathon. Front Physiol 2023; 14:1118127. [PMID: 36866178 PMCID: PMC9971726 DOI: 10.3389/fphys.2023.1118127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
Background: Prolonged and strenuous exercise has been linked to potential exercise-induced myocardial damages. One potential key to unmask the discussed underlying mechanisms of this subclinical cardiac damage could be markers of immunogenic cell damage (ICD). We investigated the kinetics of high-mobility group box 1 protein (HMGB1), soluble receptor for advanced glycation end products (sRAGE), nucleosomes, high sensitive troponin T (hs-TnT) and high sensitive C-reactive protein (hs-CRP) before and up to 12 weeks post-race and described associations with routine laboratory markers and physiological covariates. Methods: In our prospective longitudinal study, 51 adults (82% males; 43 ± 9 years) were included. All participants underwent a cardiopulmonary evaluation 10-12 weeks pre-race. HMGB1, sRAGE, nucleosomes, hs-TnT and, hs-CRP were analysed 10-12 weeks prior, 1-2 weeks before, immediately, 24 h, 72 h, and 12 weeks post-race. Results: HMGB1, sRAGE, nucleosomes and hs-TnT increased significantly from pre- to immediately post-race (0.82-2.79 ng/mL; 1132-1388 pg/mL; 9.24-56.65 ng/mL; 6-27 ng/L; p < 0.001) and returned to baseline within 24-72 h. Hs-CRP increased significantly 24 h post-race (0.88-11.5 mg/L; p < 0.001). Change in sRAGE was positively associated with change in hs-TnT (rs = 0.352, p = 0.011). Longer marathon finishing time was significantly associated with decreased levels of sRAGE [-9.2 pg/mL (β = -9.2, SE = 2.2, p < 0.001)]. Conclusion: Prolonged and strenuous exercise increases markers of ICD immediately post-race, followed by a decrease within 72 h. An acute marathon event results in transient alterations of ICD, we assume that this is not solely driven by myocyte damages.
Collapse
Affiliation(s)
- Julia Schoenfeld
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Astrid Roeh
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Bezirkskrankenhaus Augsburg, Augsburg, Germany,Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Centre Munich, Technical University Munich, Munich, Germany
| | - Pia von Korn
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and Epidemiology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kimberly Krueger
- Institute of Laboratory Medicine, German Heart Centre Munich, Technical University Munich, Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Bezirkskrankenhaus Augsburg, Augsburg, Germany,Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Johannes Scherr
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,University Center for Preventive and Sports Medicine, Balgrist University Hospital, University of Zurich, Zurich, Switzerland,*Correspondence: Johannes Scherr,
| |
Collapse
|
11
|
Starkova TY, Polyanichko AM, Artamonova TO, Tsimokha AS, Tomilin AN, Chikhirzhina EV. Structural Characteristics of High-Mobility Group Proteins HMGB1 and HMGB2 and Their Interaction with DNA. Int J Mol Sci 2023; 24:3577. [PMID: 36834988 PMCID: PMC9962726 DOI: 10.3390/ijms24043577] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Non-histone nuclear proteins HMGB1 and HMGB2 (High Mobility Group) are involved in many biological processes, such as replication, transcription, and repair. The HMGB1 and HMGB2 proteins consist of a short N-terminal region, two DNA-binding domains, A and B, and a C-terminal sequence of glutamic and aspartic acids. In this work, the structural organization of calf thymus HMGB1 and HMGB2 proteins and their complexes with DNA were studied using UV circular dichroism (CD) spectroscopy. Post-translational modifications (PTM) of HMGB1 and HMGB2 proteins were determined with MALDI mass spectrometry. We have shown that despite the similar primary structures of the HMGB1 and HMGB2 proteins, their post-translational modifications (PTMs) demonstrate quite different patterns. The HMGB1 PTMs are located predominantly in the DNA-binding A-domain and linker region connecting the A and B domains. On the contrary, HMGB2 PTMs are found mostly in the B-domain and within the linker region. It was also shown that, despite the high degree of homology between HMGB1 and HMGB2, the secondary structure of these proteins is also slightly different. We believe that the revealed structural properties might determine the difference in the functioning of the HMGB1 and HMGB2 as well as their protein partners.
Collapse
Affiliation(s)
| | | | | | - Anna S. Tsimokha
- Laboratory of Molecular Biology of Stem Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia
| | | | | |
Collapse
|
12
|
Boissady E, Abi Zeid Daou Y, Faucher E, Kohlhauer M, Lidouren F, El Hedjaj C, Chateau‐Joubert S, Hocini H, Hue S, Ghaleh B, Tissier R. High-Mobility Group Box 1-Signaling Inhibition With Glycyrrhizin Prevents Cerebral T-Cell Infiltration After Cardiac Arrest. J Am Heart Assoc 2023; 12:e027749. [PMID: 36734353 PMCID: PMC9973651 DOI: 10.1161/jaha.122.027749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background High-mobility group box 1 (HMGB1) is a major promotor of ischemic injuries and aseptic inflammatory responses. We tested its inhibition on neurological outcome and systemic immune response after cardiac arrest (CA) in rabbits. Methods and Results After 10 minutes of ventricular fibrillation, rabbits were resuscitated and received saline (control) or the HMGB1 inhibitor glycyrrhizin. A sham group underwent a similar procedure without CA. After resuscitation, glycyrrhizin blunted the successive rises in HMGB1, interleukin-6, and interleukin-10 blood levels as compared with control. Blood counts of the different immune cell populations were not different in glycyrrhizin versus control. After animal awakening, neurological outcome was improved by glycyrrhizin versus control, regarding both clinical recovery and histopathological damages. This was associated with reduced cerebral CD4+ and CD8+ T-cell infiltration beginning 2 hours after CA. Conversely, granulocytes' attraction or loss of microglial cells or cerebral monocytes were not modified by glycyrrhizin after CA. These modifications were not related to the blood-brain barrier preservation with glycyrrhizin versus control. Interestingly, the specific blockade of the HMGB1 receptor for advanced glycation end products by FPS-ZM1 recapitulated the neuroprotective effects of glycyrrhizin. Conclusions Our findings support that the early inhibition of HMGB1-signaling pathway prevents cerebral chemoattraction of T cells and neurological sequelae after CA. Glycyrrhizin could become a clinically relevant therapeutic target in this situation.
Collapse
Affiliation(s)
- Emilie Boissady
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Ecole Nationale Vétérinaire d’Alfort, IMRB, After ROSC NetworkMaisons‐AlfortFrance
| | - Yara Abi Zeid Daou
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Ecole Nationale Vétérinaire d’Alfort, IMRB, After ROSC NetworkMaisons‐AlfortFrance
| | - Estelle Faucher
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Ecole Nationale Vétérinaire d’Alfort, IMRB, After ROSC NetworkMaisons‐AlfortFrance
| | - Matthias Kohlhauer
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Ecole Nationale Vétérinaire d’Alfort, IMRB, After ROSC NetworkMaisons‐AlfortFrance
| | - Fanny Lidouren
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Ecole Nationale Vétérinaire d’Alfort, IMRB, After ROSC NetworkMaisons‐AlfortFrance
| | - Cynthia El Hedjaj
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Ecole Nationale Vétérinaire d’Alfort, IMRB, After ROSC NetworkMaisons‐AlfortFrance
| | | | - Hakim Hocini
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Vaccine Research InstituteUniversité Paris Est‐CréteilCréteilFrance
| | - Sophie Hue
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Vaccine Research InstituteUniversité Paris Est‐CréteilCréteilFrance
| | - Bijan Ghaleh
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Ecole Nationale Vétérinaire d’Alfort, IMRB, After ROSC NetworkMaisons‐AlfortFrance
| | - Renaud Tissier
- Université Paris Est‐Créteil, INSERM, IMRBCréteilFrance,Ecole Nationale Vétérinaire d’Alfort, IMRB, After ROSC NetworkMaisons‐AlfortFrance
| |
Collapse
|
13
|
Yang K, Cao F, Wang W, Tian Z, Yang L. The relationship between HMGB1 and autophagy in the pathogenesis of diabetes and its complications. Front Endocrinol (Lausanne) 2023; 14:1141516. [PMID: 37065747 PMCID: PMC10090453 DOI: 10.3389/fendo.2023.1141516] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Diabetes mellitus is a chronic metabolic disorder characterized by elevated blood glucose levels and has become the third leading threat to human health after cancer and cardiovascular disease. Recent studies have shown that autophagy is closely associated with diabetes. Under normal physiological conditions, autophagy promotes cellular homeostasis, reduces damage to healthy tissues and has bidirectional effects on regulating diabetes. However, under pathological conditions, unregulated autophagy activation leads to cell death and may contribute to the progression of diabetes. Therefore, restoring normal autophagy may be a key strategy to treat diabetes. High-mobility group box 1 protein (HMGB1) is a chromatin protein that is mainly present in the nucleus and can be actively secreted or passively released from necrotic, apoptotic, and inflammatory cells. HMGB1 can induce autophagy by activating various pathways. Studies have shown that HMGB1 plays an important role in insulin resistance and diabetes. In this review, we will introduce the biological and structural characteristics of HMGB1 and summarize the existing knowledge on the relationship between HMGB1, autophagy, diabetes, and diabetic complications. We will also summarize potential therapeutic strategies that may be useful for the prevention and treatment of diabetes and its complications.
Collapse
Affiliation(s)
- Kun Yang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feng Cao
- College of Acupuncture and Massage, Beijing University of Chinese Medicine, Beijing, China
- Department of Acupuncture, Haidian District Shuangyushu Community Health Service Center, Beijing, China
| | - Weili Wang
- Institute of Basic Research in Clinical Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyu Tian
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Lu Yang, ; Zhenyu Tian,
| | - Lu Yang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Lu Yang, ; Zhenyu Tian,
| |
Collapse
|
14
|
Forte M, Rodolico D, Ameri P, Catalucci D, Chimenti C, Crotti L, Schirone L, Pingitore A, Torella D, Iacovone G, Valenti V, Schiattarella GG, Perrino C, Sciarretta S. Molecular mechanisms underlying the beneficial effects of exercise and dietary interventions in the prevention of cardiometabolic diseases. J Cardiovasc Med (Hagerstown) 2022; 24:e3-e14. [PMID: 36729582 DOI: 10.2459/jcm.0000000000001397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cardiometabolic diseases still represent a major cause of mortality worldwide. In addition to pharmacological approaches, lifestyle interventions can also be adopted for the prevention of these morbid conditions. Lifestyle changes include exercise and dietary restriction protocols, such as calorie restriction and intermittent fasting, which were shown to delay cardiovascular ageing and elicit health-promoting effects in preclinical models of cardiometabolic diseases. Beneficial effects are mediated by the restoration of multiple molecular mechanisms in heart and vessels that are compromised by metabolic stress. Exercise and dietary restriction rescue mitochondrial dysfunction, oxidative stress and inflammation. They also improve autophagy. The result of these effects is a marked improvement of vascular and heart function. In this review, we provide a comprehensive overview of the molecular mechanisms involved in the beneficial effects of exercise and dietary restriction in models of diabetes and obesity. We also discuss clinical studies and gap in animal-to-human translation.
Collapse
Affiliation(s)
- Maurizio Forte
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli
| | - Daniele Rodolico
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico.,Department of Internal Medicine, University of Genova, Genova
| | - Daniele Catalucci
- Humanitas Research Hospital, IRCCS, Rozzano.,National Research Council, Institute of Genetic and Biomedical Research - UOS, Milan
| | - Cristina Chimenti
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome
| | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital.,Department of Medicine and Surgery, Università Milano-Bicocca, Milan
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina
| | - Annachiara Pingitore
- Department of General and Specialistic Surgery 'Paride Stefanini' Sapienza University of Rome
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro
| | | | | | - Gabriele G Schiattarella
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli.,Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina
| | | |
Collapse
|
15
|
Al-Hakeim HK, Al-Kaabi QJ, Maes M. High mobility group box 1 and Dickkopf-related protein 1 as biomarkers of glucose toxicity, atherogenicity, and lower β cell function in patients with type 2 diabetes mellitus. Growth Factors 2022; 40:240-253. [PMID: 36165005 DOI: 10.1080/08977194.2022.2126317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with increased atherogenicity and inflammatory responses, which may be related to high mobility group box 1 (HMGB1) and Dickkopf-related protein 1 (DKK1). The role of HMGB1 and DKK1 in T2DM is examined in association with lipid and insulin profiles. Serum HMGB1 and DKK1 were measured in T2DM with and without hypertension and compared with controls. The results showed that HMGB1 and DKK1 are higher in T2DM irrespective of hypertension. A large part of the variance in the β-cell index and glucose toxicity was explained by the combined effects of HMGB1 and DKK1. In conclusion, both HMGB1 and DKK1 may contribute to increased atherogenicity in T2DM. Moreover, both biomarkers may cause more deficits in β-cell function and increase glucose toxicity leading to the development of more inflammation and diabetic complications. HMGB1 and the Wnt pathways are other drug targets in treating T2DM.
Collapse
Affiliation(s)
| | | | - Michael Maes
- Faculty of Medicine, Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
- School of Medicine, IMPACT Strategic Research Centre, Deakin University, Geelong, Australia
| |
Collapse
|
16
|
Belmadani S, Matrougui K. Role of High Mobility Group Box 1 in Cardiovascular Diseases. Inflammation 2022; 45:1864-1874. [PMID: 35386038 PMCID: PMC11145736 DOI: 10.1007/s10753-022-01668-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/27/2022] [Accepted: 03/28/2022] [Indexed: 11/05/2022]
Abstract
High Mobility Group Box 1 (HMGB1) is a ubiquitous, highly conserved nuclear and cytosolic protein that has diverse biological roles depending on its cellular location and posttranslational modifications. The HMGB1 is localized in the nucleus but can be translocated to the cytoplasm to modulate the intracellular signaling and eventually secreted outside the cells. It is widely established that HMGB1 plays a key role in inflammation; however, the role of HMGB1 in the cardiovascular diseases is not well understood. In this review, we will discuss the latest reports on the pathophysiological link between HMGB1 and cardiovascular complications, with special emphasis on the inflammation. Thus, the understanding of the role of HMGB1 may provide new insights into developing new HMGB1-based therapies.
Collapse
Affiliation(s)
- Souad Belmadani
- Department of Physiological Sciences, EVMS, Norfolk, Virginia, 23501, USA
| | - Khalid Matrougui
- Department of Physiological Sciences, EVMS, Norfolk, Virginia, 23501, USA.
| |
Collapse
|
17
|
Chen G, Jiang H, Yao Y, Tao Z, Chen W, Huang F, Chen X. Macrophage, a potential targeted therapeutic immune cell for cardiomyopathy. Front Cell Dev Biol 2022; 10:908790. [PMID: 36247005 PMCID: PMC9561843 DOI: 10.3389/fcell.2022.908790] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiomyopathy is a major cause of heart failure, leading to systolic and diastolic dysfunction and promoting adverse cardiac remodeling. Macrophages, as key immune cells of the heart, play a crucial role in inflammation and fibrosis. Moreover, exogenous and cardiac resident macrophages are functionally and phenotypically different during cardiac injury. Although experimental evidence has shown that macrophage-targeted therapy is promising in cardiomyopathy, clinical translation remains challenging. In this article, the molecular mechanism of macrophages in cardiomyopathy has been discussed in detail based on existing literature. The issues and considerations of clinical treatment strategies for myocardial fibrosis has also been analyzed.
Collapse
Affiliation(s)
- Ganyi Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongwei Jiang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yiwei Yao
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhonghao Tao
- Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fuhua Huang
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Scavello F, Piacentini L, Castiglione S, Zeni F, Macrì F, Casaburo M, Vinci MC, Colombo GI, Raucci A. Effects of RAGE Deletion on the Cardiac Transcriptome during Aging. Int J Mol Sci 2022; 23:ijms231911130. [PMID: 36232442 PMCID: PMC9569842 DOI: 10.3390/ijms231911130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022] Open
Abstract
Cardiac aging is characterized by increased cardiomyocyte hypertrophy, myocardial stiffness, and fibrosis, which enhance cardiovascular risk. The receptor for advanced glycation end-products (RAGE) is involved in several age-related diseases. RAGE knockout (Rage−/−) mice show an acceleration of cardiac dimension changes and interstitial fibrosis with aging. This study identifies the age-associated cardiac gene expression signature induced by RAGE deletion. We analyzed the left ventricle transcriptome of 2.5-(Young), 12-(Middle age, MA), and 21-(Old) months-old female Rage−/− and C57BL/6N (WT) mice. By comparing Young, MA, and Old Rage−/− versus age-matched WT mice, we identified 122, 192, and 12 differently expressed genes, respectively. Functional inference analysis showed that RAGE deletion is associated with: (i) down-regulation of genes involved in antigen processing and presentation of exogenous antigen, adaptive immune response, and cellular responses to interferon beta and gamma in Young animals; (ii) up-regulation of genes related to fatty acid oxidation, cardiac structure remodeling and cellular response to hypoxia in MA mice; (iii) up-regulation of few genes belonging to complement activation and triglyceride biosynthetic process in Old animals. Our findings show that the age-dependent cardiac phenotype of Rage−/− mice is associated with alterations of genes related to adaptive immunity and cardiac stress pathways.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Luca Piacentini
- Bioinformatics and Artificial Intelligence Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Stefania Castiglione
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Filippo Zeni
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Federica Macrì
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Manuel Casaburo
- Animal Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Maria Cristina Vinci
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Gualtiero I. Colombo
- Unit of Immunology and Functional Genomics, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Correspondence: (G.I.C.); (A.R.); Tel.: +39-025-800-2464 (G.I.C.); +39-025-800-2802 (A.R.); Fax: +39-025-800-2342 (G.I.C. & A.R.)
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Animal Facility, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
- Correspondence: (G.I.C.); (A.R.); Tel.: +39-025-800-2464 (G.I.C.); +39-025-800-2802 (A.R.); Fax: +39-025-800-2342 (G.I.C. & A.R.)
| |
Collapse
|
19
|
Elia E, Ministrini S, Carbone F, Montecucco F. Diabetic cardiomyopathy and inflammation: development of hostile microenvironment resulting in cardiac damage. Minerva Cardiol Angiol 2022; 70:357-369. [PMID: 33427423 DOI: 10.23736/s2724-5683.20.05454-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Diabetes mellitus is emerging as a major risk factor for heart failure. Diabetic cardiomyopathy is defined as a myocardial dysfunction that is not caused by underlying hypertension or coronary artery disease. Studies about clinical features, natural history and outcomes of the disease are few and often conflicting, because a universally accepted operative definition of diabetic cardiomyopathy is still lacking. Hyperglycemia and related metabolic and endocrine disorders are the triggering factors of myocardial damage in diabetic cardiomyopathy through multiple mechanisms. Among these mechanisms, inflammation has a relevant role, similar to other chronic myocardial disease, such as hypertensive or ischemic heart disease. A balance between inflammatory damage and healing processes is fundamental for homeostasis of myocardial tissue, whereas diabetes mellitus produces an imbalance, promoting inflammation and delaying healing. Therefore, diabetes-related chronic inflammatory state can produce a progressive qualitative deterioration of myocardial tissue, which reflects on progressive left ventricular functional impairment, which can be either diastolic, with prevalent myocardial hypertrophy, or systolic, with prevalent myocardial fibrosis. The aim of this narrative review is to summarize the existing evidence about the role of inflammation in diabetic cardiomyopathy onset and development. Ultimately, potential pharmacological strategies targeting inflammatory response will be reviewed and discussed.
Collapse
Affiliation(s)
- Edoardo Elia
- Division of Cardiology, Department of Internal Medicine, Città della Salute e della Scienza, Turin, Italy
| | - Stefano Ministrini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy -
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
20
|
Jeong JH, Lee DH, Song J. HMGB1 signaling pathway in diabetes-related dementia: Blood-brain barrier breakdown, brain insulin resistance, and Aβ accumulation. Biomed Pharmacother 2022; 150:112933. [PMID: 35413600 DOI: 10.1016/j.biopha.2022.112933] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/28/2022] Open
Abstract
Diabetes contributes to the onset of various diseases, including cancer and cardiovascular and neurodegenerative diseases. Recent studies have highlighted the similarities and relationship between diabetes and dementia as an important issue for treating diabetes-related cognitive deficits. Diabetes-related dementia exhibits several features, including blood-brain barrier disruption, brain insulin resistance, and Aβ over-accumulation. High-mobility group box1 (HMGB1) is a protein known to regulate gene transcription and cellular mechanisms by binding to DNA or chromatin via receptor for advanced glycation end-products (RAGE) and toll-like receptor 4 (TLR4). Recent studies have demonstrated that the interplay between HMGB1, RAGE, and TLR4 can impact both neuropathology and diabetic alterations. Herein, we review the recent research regarding the roles of HMGB1-RAGE-TLR4 axis in diabetes-related dementia from several perspectives and emphasize the importance of the influence of HMGB1 in diabetes-related dementia.
Collapse
Affiliation(s)
- Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| | - Dong Hoon Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School, and Chonnam National University Hwasun Hospital, Hwasun 58128, Jeollanam-do, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| |
Collapse
|
21
|
Paracrine signal emanating from stressed cardiomyocytes aggravates inflammatory microenvironment in diabetic cardiomyopathy. iScience 2022; 25:103973. [PMID: 35281739 PMCID: PMC8905320 DOI: 10.1016/j.isci.2022.103973] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/17/2021] [Accepted: 02/18/2022] [Indexed: 11/21/2022] Open
Abstract
Myocardial inflammation contributes to cardiomyopathy in diabetic patients through incompletely defined underlying mechanisms. In both human and time-course experimental samples, diabetic hearts exhibited abnormal ER, with a maladaptive shift over time in rodents. Furthermore, as a cardiac ER dysfunction model, mice with cardiac-specific p21-activated kinase 2 (PAK2) deletion exhibited heightened myocardial inflammatory response in diabetes. Mechanistically, maladaptive ER stress-induced CCAAT/enhancer-binding protein homologous protein (CHOP) is a novel transcriptional regulator of cardiac high-mobility group box-1 (HMGB1). Cardiac stress-induced release of HMGB1 facilitates M1 macrophage polarization, aggravating myocardial inflammation. Therapeutically, sequestering the extracellular HMGB1 using glycyrrhizin conferred cardioprotection through its anti-inflammatory action. Our findings also indicated that an intact cardiac ER function and protective effects of the antidiabetic drug interdependently attenuated the cardiac inflammation-induced dysfunction. Collectively, we introduce an ER stress-mediated cardiomyocyte-macrophage link, altering the macrophage response, thereby providing insight into therapeutic prospects for diabetes-associated cardiac dysfunction.
Collapse
|
22
|
Wenzl FA, Ambrosini S, Mohammed SA, Kraler S, Lüscher TF, Costantino S, Paneni F. Inflammation in Metabolic Cardiomyopathy. Front Cardiovasc Med 2021; 8:742178. [PMID: 34671656 PMCID: PMC8520939 DOI: 10.3389/fcvm.2021.742178] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
Overlapping pandemics of lifestyle-related diseases pose a substantial threat to cardiovascular health. Apart from coronary artery disease, metabolic disturbances linked to obesity, insulin resistance and diabetes directly compromise myocardial structure and function through independent and shared mechanisms heavily involving inflammatory signals. Accumulating evidence indicates that metabolic dysregulation causes systemic inflammation, which in turn aggravates cardiovascular disease. Indeed, elevated systemic levels of pro-inflammatory cytokines and metabolic substrates induce an inflammatory state in different cardiac cells and lead to subcellular alterations thereby promoting maladaptive myocardial remodeling. At the cellular level, inflammation-induced oxidative stress, mitochondrial dysfunction, impaired calcium handling, and lipotoxicity contribute to cardiomyocyte hypertrophy and dysfunction, extracellular matrix accumulation and microvascular disease. In cardiometabolic patients, myocardial inflammation is maintained by innate immune cell activation mediated by pattern recognition receptors such as Toll-like receptor 4 (TLR4) and downstream activation of the NLRP3 inflammasome and NF-κB-dependent pathways. Chronic low-grade inflammation progressively alters metabolic processes in the heart, leading to a metabolic cardiomyopathy (MC) phenotype and eventually to heart failure with preserved ejection fraction (HFpEF). In accordance with preclinical data, observational studies consistently showed increased inflammatory markers and cardiometabolic features in patients with HFpEF. Future treatment approaches of MC may target inflammatory mediators as they are closely intertwined with cardiac nutrient metabolism. Here, we review current evidence on inflammatory processes involved in the development of MC and provide an overview of nutrient and cytokine-driven pro-inflammatory effects stratified by cell type.
Collapse
Affiliation(s)
- Florian A Wenzl
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Dozio E, Massaccesi L, Corsi Romanelli MM. Glycation and Glycosylation in Cardiovascular Remodeling: Focus on Advanced Glycation End Products and O-Linked Glycosylations as Glucose-Related Pathogenetic Factors and Disease Markers. J Clin Med 2021; 10:jcm10204792. [PMID: 34682915 PMCID: PMC8539574 DOI: 10.3390/jcm10204792] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 02/07/2023] Open
Abstract
Glycation and glycosylation are non-enzymatic and enzymatic reactions, respectively, of glucose, glucose metabolites, and other reducing sugars with different substrates, such as proteins, lipids, and nucleic acids. Increased availability of glucose is a recognized risk factor for the onset and progression of diabetes-mellitus-associated disorders, among which cardiovascular diseases have a great impact on patient mortality. Both advanced glycation end products, the result of non-enzymatic glycation of substrates, and O-linked-N-Acetylglucosaminylation, a glycosylation reaction that is controlled by O-N-AcetylGlucosamine (GlcNAc) transferase (OGT) and O-GlcNAcase (OGA), have been shown to play a role in cardiovascular remodeling. In this review, we aim (1) to summarize the most recent data regarding the role of glycation and O-linked-N-Acetylglucosaminylation as glucose-related pathogenetic factors and disease markers in cardiovascular remodeling, and (2) to discuss potential common mechanisms linking these pathways to the dysregulation and/or loss of function of different biomolecules involved in this field.
Collapse
Affiliation(s)
- Elena Dozio
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (L.M.); (M.M.C.R.)
- Correspondence: ; Tel.: +39-02-50-315-342
| | - Luca Massaccesi
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (L.M.); (M.M.C.R.)
| | - Massimiliano Marco Corsi Romanelli
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (L.M.); (M.M.C.R.)
- Service of Laboratory Medicine1-Clinical Pathology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| |
Collapse
|
24
|
Xu Z, Jin Y, Gao Z, Zeng Y, Du J, Yan H, Chen X, Ping L, Lin N, Yang B, He Q, Luo P. Autophagic degradation of CCN2 (cellular communication network factor 2) causes cardiotoxicity of sunitinib. Autophagy 2021; 18:1152-1173. [PMID: 34432562 DOI: 10.1080/15548627.2021.1965712] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Excessive macroautophagy/autophagy is one of the causes of cardiomyocyte death induced by cardiovascular diseases or cancer therapy, yet the underlying mechanism remains unknown. We and other groups previously reported that autophagy might contribute to cardiomyocyte death caused by sunitinib, a tumor angiogenesis inhibitor that is widely used in clinic, which may help to understand the mechanism of autophagy-induced cardiomyocyte death. Here, we found that sunitinib-induced autophagy leads to apoptosis of cardiomyocyte and cardiac dysfunction as the cardiomyocyte-specific Atg7-/+ heterozygous mice are resistant to sunitinib. Sunitinib-induced maladaptive autophagy selectively degrades the cardiomyocyte survival mediator CCN2 (cellular communication network factor 2) through the TOLLIP (toll interacting protein)-mediated endosome-related pathway and cardiomyocyte-specific knockdown of Ccn2 through adeno-associated virus serotype 9 (AAV9) mimics sunitinib-induced cardiac dysfunction in vivo, suggesting that the autophagic degradation of CCN2 is one of the causes of sunitinib-induced cardiotoxicity and death of cardiomyocytes. Remarkably, deletion of Hmgb1 (high mobility group box 1) inhibited sunitinib-induced cardiomyocyte autophagy and apoptosis, and the HMGB1-specific inhibitor glycyrrhizic acid (GA) significantly mitigated sunitinib-induced autophagy, cardiomyocyte death and cardiotoxicity. Our study reveals a novel target protein of autophagic degradation in the regulation of cardiomyocyte death and highlights the pharmacological inhibitor of HMGB1 as an attractive approach for improving the safety of sunitinib-based cancer therapy.
Collapse
Affiliation(s)
- Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Ying Jin
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Zizheng Gao
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Yan Zeng
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Jiangxia Du
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Xueqin Chen
- Department of Oncology, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Li Ping
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Nengming Lin
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R.China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, P.R.China.,Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China.,Department of Pharmacology and Toxicology, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
25
|
Coppola A, Capuani B, Pacifici F, Pastore D, Arriga R, Bellia A, Andreadi A, Di Daniele N, Lauro R, Della-Morte D, Sconocchia G, Lauro D. Activation of Peripheral Blood Mononuclear Cells and Leptin Secretion: New Potential Role of Interleukin-2 and High Mobility Group Box (HMGB)1. Int J Mol Sci 2021; 22:7988. [PMID: 34360753 PMCID: PMC8347813 DOI: 10.3390/ijms22157988] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/18/2021] [Accepted: 07/22/2021] [Indexed: 01/15/2023] Open
Abstract
Activation of innate immunity and low-grade inflammation contributes to hyperglycemia and an onset of Type 2 Diabetes Mellitus (T2DM). Interleukin-2 (IL-2), leptin, High Mobility Group Box-1 (HMGB-1), and increased glucose concentrations are mediators of these processes also by modulating peripheral blood mononuclear cells (PBMCs) response. The aim of this study was to investigate if HMGB-1 and IL-2 turn on PBMCs and their leptin secretion. In isolated human PBMCs and their subpopulations from healthy individuals and naïve T2DM patients, leptin release, pro-inflammatory response and Toll-like Receptors (TLRs) activation was measured. After treatment with IL-2 and HMGB1, NK (Natural Killer) have the highest amount of leptin secretion, whilst NK-T have the maximal release in basal conditions. TLR4 (TAK242) and/or TLR2 (TLR2-IgA) inhibitors decreased leptin secretion after IL-2 and HMGB1 treatment. A further non-significant increase in leptin secretion was reported in PBMCs of naive T2DM patients in response to IL-2 and HMGB-1 stimulation. Finally, hyperglycemia or hyperinsulinemia might stimulate leptin secretion from PBMCs. The amount of leptin released from PBMCs after the different treatments was enough to stimulate the secretion of IL-1β from monocytes. Targeting leptin sera levels and secretion from PBMCs could represent a new therapeutic strategy to counteract metabolic diseases such as T2DM.
Collapse
Affiliation(s)
- Andrea Coppola
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
| | - Barbara Capuani
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
| | - Francesca Pacifici
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
| | - Donatella Pastore
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
| | - Roberto Arriga
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
| | - Alfonso Bellia
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
- Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| | - Aikaterini Andreadi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
- Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
- Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| | - Renato Lauro
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
| | - David Della-Morte
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Rome Open University, 00166 Rome, Italy
| | - Giuseppe Sconocchia
- Institute of Translational Pharmacology, National Research Council Rome, 00133 Rome, Italy;
| | - Davide Lauro
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.C.); (B.C.); (F.P.); (D.P.); (R.A.); (A.B.); (A.A.); (N.D.D.); (R.L.); (D.D.-M.)
- Department of Medical Sciences, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
26
|
Kaur N, Guan Y, Raja R, Ruiz-Velasco A, Liu W. Mechanisms and Therapeutic Prospects of Diabetic Cardiomyopathy Through the Inflammatory Response. Front Physiol 2021; 12:694864. [PMID: 34234695 PMCID: PMC8257042 DOI: 10.3389/fphys.2021.694864] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
The incidence of heart failure (HF) continues to increase rapidly in patients with diabetes. It is marked by myocardial remodeling, including fibrosis, hypertrophy, and cell death, leading to diastolic dysfunction with or without systolic dysfunction. Diabetic cardiomyopathy (DCM) is a distinct myocardial disease in the absence of coronary artery disease. DCM is partially induced by chronic systemic inflammation, underpinned by a hostile environment due to hyperglycemia, hyperlipidemia, hyperinsulinemia, and insulin resistance. The detrimental role of leukocytes, cytokines, and chemokines is evident in the diabetic heart, yet the precise role of inflammation as a cause or consequence of DCM remains incompletely understood. Here, we provide a concise review of the inflammatory signaling mechanisms contributing to the clinical complications of diabetes-associated HF. Overall, the impact of inflammation on the onset and development of DCM suggests the potential benefits of targeting inflammatory cascades to prevent DCM. This review is tailored to outline the known effects of the current anti-diabetic drugs, anti-inflammatory therapies, and natural compounds on inflammation, which mitigate HF progression in diabetic populations.
Collapse
Affiliation(s)
| | | | | | | | - Wei Liu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
27
|
Scavello F, Zeni F, Milano G, Macrì F, Castiglione S, Zuccolo E, Scopece A, Pezone G, Tedesco CC, Nigro P, Degani G, Gambini E, Veglia F, Popolo L, Pompilio G, Colombo GI, Bianchi ME, Raucci A. Soluble Receptor for Advanced Glycation End-products regulates age-associated Cardiac Fibrosis. Int J Biol Sci 2021; 17:2399-2416. [PMID: 34326683 PMCID: PMC8315019 DOI: 10.7150/ijbs.56379] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/20/2021] [Indexed: 12/21/2022] Open
Abstract
Myocardial aging increases the cardiovascular risk in the elderly. The Receptor for Advanced Glycation End-products (RAGE) is involved in age-related disorders. The soluble isoform (sRAGE) acts as a scavenger blocking the membrane-bound receptor activation. This study aims at investigating RAGE contribution to age-related cardiac remodeling. We analyzed the cardiac function of three different age groups of female Rage-/- and C57BL/6N (WT) mice: 2.5- (Young), 12- (Middle-age, MA) and 21-months (Old) old. While aging, Rage-/- mice displayed an increase in left ventricle (LV) dimensions compared to age-matched WT animals, with the main differences observed in the MA groups. Rage-/- mice showed higher fibrosis and a larger number of α-Smooth Muscle Actin (SMA)+ cells with age, along with increased expression of pro-fibrotic Transforming Growth Factor (TGF)-β1 pathway components. RAGE isoforms were undetectable in LV of WT mice, nevertheless, circulating sRAGE declined with aging and inversely associated with LV diastolic dimensions. Human cardiac fibroblasts stimulated with sRAGE exhibited a reduction in proliferation, pro-fibrotic proteins and TGF-beta Receptor 1 (TGFbR1) expression and Smad2-3 activation. Finally, sRAGE administration to MA WT animals reduced cardiac fibrosis. Hence, our work shows that RAGE associates with age-dependent myocardial changes and indicates sRAGE as an inhibitor of cardiac fibroblasts differentiation and age-dependent cardiac fibrosis.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Filippo Zeni
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Giuseppina Milano
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Federica Macrì
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Stefania Castiglione
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Estella Zuccolo
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Alessandro Scopece
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Giovanni Pezone
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | - Patrizia Nigro
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Genny Degani
- Department of Biosciences, University of Milan, Milan, Italy
| | - Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Laura Popolo
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Gualtiero I. Colombo
- Unit of Immunology and Functional Genomics, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Marco E. Bianchi
- Chromatin Dynamics Unit, San Raffaele University and IRCCS San Raffaele Hospital, Milan, Italy
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| |
Collapse
|
28
|
MiR-216a-5p ameliorates learning-memory deficits and neuroinflammatory response of Alzheimer's disease mice via regulation of HMGB1/NF-κB signaling. Brain Res 2021; 1766:147511. [PMID: 33957091 DOI: 10.1016/j.brainres.2021.147511] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 01/25/2023]
Abstract
OBJECTIVE The objective of this study was to explore whether miR-216a-5p could affect the learning-memory ability and inflammatory response of Alzheimer's disease (AD) mice via regulation of the HMGB1/NF-κB pathway. METHODS Mice were divided into the normal (wild-type C57BL/6 mice), AD (APP/PS1 double-transgenic mice), AD + miR-216a-5p, and AD + vector groups. The Morris water maze test was used to examine learning and memory ability. Nissl staining and TUNEL staining were performed to observe the survival and apoptosis of hippocampal neurons. In addition, Aβ deposition and the expression of inflammatory cytokines were determined, while miR-216a-5p expression and HMGB1/NF-κB pathway-related proteins were detected by qRT-PCR and Western blotting, respectively. RESULTS AD mice exhibited decreased miR-216a-5p expression but increased HMGB-1 protein expression in the hippocampus, and these mice had a prolonged escape latency, fewer number of times crossing the platform location and shortened time in the target quadrant compared to those in normal mice. AD mice also had an elevated number of TUNEL-positive cells, increased deposition of Aβ, increased expression of inflammatory cytokines and decreased number of Nissl-positive cells. In addition, AD mice presented with downregulated expression of cytoplasmic NF-κB p65 protein but upregulated expression of nuclear NF-κB p65 protein. However, AD mice treated with miR-216a-5p exhibited significant improvements of the abovementioned parameters. The dual-luciferase reporter assay confirmed that HMGB1 is a target gene of miR-216a-5p. CONCLUSION MiR-216a-5p can improve learning-memory ability and attenuate the inflammatory response of AD mice through targeted inhibition of the HMGB1/NF-κB pathway.
Collapse
|
29
|
High-mobility group box 1 serves as an inflammation driver of cardiovascular disease. Biomed Pharmacother 2021; 139:111555. [PMID: 33865014 DOI: 10.1016/j.biopha.2021.111555] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 03/15/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is the most deadly disease, which can cause sudden death, in which inflammation is a key factor in its occurrence and development. High-mobility group box 1 (HMGB1) is a novel nuclear DNA-binding protein that activates innate immunity to induce inflammation in CVD. HMGB1 exists in the cytoplasm and nucleus of different cell types, including those in the heart. By binding to its receptors, HMGB1 triggers a variety of signaling cascades, leading to inflammation and CVD. To help develop HMGB1-targeted therapies, here we discuss HMGB1 and its biological functions, receptors, signaling pathways, and pathophysiology related to inflammation and CVD, including cardiac remodeling, cardiac hypertrophy, myocardial infarction, heart failure, pulmonary hypertension, atherosclerosis, and cardiomyopathy.
Collapse
|
30
|
Burr SD, Stewart JA. Rap1a Overlaps the AGE/RAGE Signaling Cascade to Alter Expression of α-SMA, p-NF-κB, and p-PKC-ζ in Cardiac Fibroblasts Isolated from Type 2 Diabetic Mice. Cells 2021; 10:cells10030557. [PMID: 33806572 PMCID: PMC8000763 DOI: 10.3390/cells10030557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/28/2021] [Accepted: 02/28/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease, specifically heart failure, is a common complication for individuals with type 2 diabetes mellitus. Heart failure can arise with stiffening of the left ventricle, which can be caused by “active” cardiac fibroblasts (i.e., myofibroblasts) remodeling the extracellular matrix (ECM). Differentiation of fibroblasts to myofibroblasts has been demonstrated to be an outcome of AGE/RAGE signaling. Hyperglycemia causes advanced glycated end products (AGEs) to accumulate within the body, and this process is greatly accelerated under chronic diabetic conditions. AGEs can bind and activate their receptor (RAGE) to trigger multiple downstream outcomes, such as altering ECM remodeling, inflammation, and oxidative stress. Previously, our lab has identified a small GTPase, Rap1a, that possibly overlaps the AGE/RAGE signaling cascade to affect the downstream outcomes. Rap1a acts as a molecular switch connecting extracellular signals to intracellular responses. Therefore, we hypothesized that Rap1a crosses the AGE/RAGE cascade to alter the expression of AGE/RAGE associated signaling proteins in cardiac fibroblasts in type 2 diabetic mice. To delineate this cascade, we used genetically different cardiac fibroblasts from non-diabetic, diabetic, non-diabetic RAGE knockout, diabetic RAGE knockout, and Rap1a knockout mice and treated them with pharmacological modifiers (exogenous AGEs, EPAC, Rap1a siRNA, and pseudosubstrate PKC-ζ). We examined changes in expression of proteins implicated as markers for myofibroblasts (α-SMA) and inflammation/oxidative stress (NF-κB and SOD-1). In addition, oxidative stress was also assessed by measuring hydrogen peroxide concentration. Our results indicated that Rap1a connects to the AGE/RAGE cascade to promote and maintain α-SMA expression in cardiac fibroblasts. Moreover, Rap1a, in conjunction with activation of the AGE/RAGE cascade, increased NF-κB expression as well as hydrogen peroxide concentration, indicating a possible oxidative stress response. Additionally, knocking down Rap1a expression resulted in an increase in SOD-1 expression suggesting that Rap1a can affect oxidative stress markers independently of the AGE/RAGE signaling cascade. These results demonstrated that Rap1a contributes to the myofibroblast population within the heart via AGE/RAGE signaling as well as promotes possible oxidative stress. This study offers a new potential therapeutic target that could possibly reduce the risk for developing diabetic cardiovascular complications attributed to AGE/RAGE signaling.
Collapse
|
31
|
Ling S, Xu JW. NETosis as a Pathogenic Factor for Heart Failure. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6687096. [PMID: 33680285 PMCID: PMC7929675 DOI: 10.1155/2021/6687096] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/07/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
Heart failure threatens the lives of patients and reduces their quality of life. Heart failure, especially heart failure with preserved ejection fraction, is closely related to systemic and local cardiac persistent chronic low-grade aseptic inflammation, microvascular damage characterized by endothelial dysfunction, oxidative stress, myocardial remodeling, and fibrosis. However, the initiation and development of persistent chronic low-grade aseptic inflammation is unexplored. Oxidative stress-mediated neutrophil extracellular traps (NETs) are the main immune defense mechanism against external bacterial infections. Furthermore, NETs play important roles in noninfectious diseases. After the onset of myocardial infarction, atrial fibrillation, or myocarditis, neutrophils infiltrate the damaged tissue and aggravate inflammation. In tissue injury, damage-related molecular patterns (DAMPs) may induce pattern recognition receptors (PRRs) to cause NETs, but whether NETs are directly involved in the pathogenesis and development of heart failure and the mechanism is still unclear. In this review, we analyzed the markers of heart failure and heart failure-related diseases and comorbidities, such as mitochondrial DNA, high mobility box group box 1, fibronectin extra domain A, and galectin-3, to explore their role in inducing NETs and to investigate the mechanism of PRRs, such as Toll-like receptors, receptor for advanced glycation end products, cGAS-STING, and C-X-C motif chemokine receptor 2, in activating NETosis. Furthermore, we discussed oxidative stress, especially the possibility that imbalance of thiol redox and MPO-derived HOCl promotes the production of 2-chlorofatty acid and induces NETosis, and analyzed the possibility of NETs triggering coronary microvascular thrombosis. In some heart diseases, the deletion or blocking of neutrophil-specific myeloperoxidase and peptidylarginine deiminase 4 has shown effectiveness. According to the results of current pharmacological studies, MPO and PAD4 inhibitors are effective at least for myocardial infarction, atherosclerosis, and certain autoimmune diseases, whose deterioration can lead to heart failure. This is essential for understanding NETosis as a therapeutic factor of heart failure and the related new pathophysiology and therapeutics of heart failure.
Collapse
Affiliation(s)
- Shuang Ling
- Institute of Interdisciplinary Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jin-Wen Xu
- Institute of Interdisciplinary Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
32
|
Behl T, Sharma E, Sehgal A, Kaur I, Kumar A, Arora R, Pal G, Kakkar M, Kumar R, Bungau S. Expatiating the molecular approaches of HMGB1 in diabetes mellitus: Highlighting signalling pathways via RAGE and TLRs. Mol Biol Rep 2021; 48:1869-1881. [PMID: 33479829 DOI: 10.1007/s11033-020-06130-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/24/2020] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus (DM) has become one of the major healthcare challenges worldwide in the recent times and inflammation being one of its key pathogenic process/mechanism affect several body parts including the peripheral and central nervous system. High-mobility group box 1 (HMGB1) is one of the major non-histone proteins that plays a key role in triggering the inflammatory response. Upon its release into the extracellular milieu, HMGB1 acts as an "alarmin" for the immune system to initiate tissue repair as a component of the host defense system. Furthermore, HMGB1 along with its downstream receptors like Toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE) serve as the suitable target for DM. The forthcoming research in the field of diabetes would potentially focus on the development of alternative approaches to target the centre of inflammation that is primarily mediated by HMGB1 to improve diabetic-related complications. This review covers the therapeutic actions of HMGB1 protein, which acts by activating the RAGE and TLR molecules to constitute a functional tripod system, in turn activating NF-κB pathway that contributes to the production of mediators for pro-inflammatory cytokines associated with DM. The interaction between TLR2 and TLR4 with ligands present in the host and the activation of RAGE stimulates various immune and metabolic responses that contribute to diabetes. This review emphasizes to elucidate the role of HMGB1 in the initiation and progression of DM and control over the inflammatory tripod as a promising therapeutic approach in the management of DM.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Eshita Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Giridhari Pal
- Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Munish Kakkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ravinder Kumar
- Cardiovascular Research Institute, Icahn School of Medicine, New York, USA
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
33
|
Battault S, Renguet E, Van Steenbergen A, Horman S, Beauloye C, Bertrand L. Myocardial glucotoxicity: Mechanisms and potential therapeutic targets. Arch Cardiovasc Dis 2020; 113:736-748. [PMID: 33189592 DOI: 10.1016/j.acvd.2020.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022]
Abstract
Besides coronary artery disease, which remains the main cause of heart failure in patients with diabetes, factors independent of coronary artery disease are involved in the development of heart failure in the onset of what is called diabetic cardiomyopathy. Among them, hyperglycaemia - a hallmark of type 2 diabetes - has both acute and chronic deleterious effects on myocardial function, and clearly participates in the establishment of diabetic cardiomyopathy. In the present review, we summarize the cellular and tissular events that occur in a heart exposed to hyperglycaemia, and depict the complex molecular mechanisms proposed to be involved in glucotoxicity. Finally, from a more translational perspective, different therapeutic strategies targeting hyperglycaemia-mediated molecular mechanisms will be detailed.
Collapse
Affiliation(s)
- Sylvain Battault
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Edith Renguet
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Anne Van Steenbergen
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Sandrine Horman
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Christophe Beauloye
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium; Division of cardiology, Cliniques Universitaires Saint-Luc, B-1200 Brussels, Belgium.
| | - Luc Bertrand
- Pole of cardiovascular research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium; WELBIO, B-1300 Wavre, Belgium.
| |
Collapse
|
34
|
Kim YH, Kwak MS, Lee B, Shin JM, Aum S, Park IH, Lee MG, Shin JS. Secretory autophagy machinery and vesicular trafficking are involved in HMGB1 secretion. Autophagy 2020; 17:2345-2362. [PMID: 33017561 PMCID: PMC8496717 DOI: 10.1080/15548627.2020.1826690] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Nuclear protein HMGB1 is secreted in response to various stimuli and functions as a danger-associated molecular pattern. Extracellular HMGB1 induces inflammation, cytokine production, and immune cell recruitment via activation of various receptors. As HMGB1 does not contain an endoplasmic reticulum-targeting signal peptide, HMGB1 is secreted via the endoplasmic reticulum-Golgi independently via an unconventional secretion pathway. However, the mechanism underlying HMGB1 secretion remains largely unknown. Here, we investigated the role of secretory autophagy machinery and vesicular trafficking in HMGB1 secretion. We observed that HSP90AA1 (heat shock protein 90 alpha family class A member 1), a stress-inducible protein, regulates the translocation of HMGB1 from the nucleus to the cytoplasm and its secretion through direct interaction. Additionally, geldanamycin, an HSP90AA1 inhibitor, reduced HMGB1 secretion. GORASP2/GRASP55 (golgi reassembly stacking protein 2), ARF1Q71L (ADP ribosylation factor 1), and SAR1AT39N (secretion associated Ras related GTPase 1A), which promoted unconventional protein secretion, increased HMGB1 secretion. HMGB1 secretion was inhibited by an early autophagy inhibitor and diminished in ATG5-deficient cells even when GORASP2 was overexpressed. In contrast, a late autophagy inhibitor increased HMGB1 secretion under the same conditions. The multivesicular body formation inhibitor GW4869 dramatically decreased HMGB1 secretion under HMGB1 secretion-inducing conditions. Thus, we demonstrated that secretory autophagy and multivesicular body formation mediate HMGB1 secretion.
Collapse
Affiliation(s)
- Young Hun Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Man Sup Kwak
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Bin Lee
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Min Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sowon Aum
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - In Ho Park
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Min Goo Lee
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, Korea
| |
Collapse
|
35
|
Tan Y, Zhang Z, Zheng C, Wintergerst KA, Keller BB, Cai L. Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence. Nat Rev Cardiol 2020; 17:585-607. [PMID: 32080423 PMCID: PMC7849055 DOI: 10.1038/s41569-020-0339-2] [Citation(s) in RCA: 476] [Impact Index Per Article: 95.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
The pathogenesis and clinical features of diabetic cardiomyopathy have been well-studied in the past decade, but effective approaches to prevent and treat this disease are limited. Diabetic cardiomyopathy occurs as a result of the dysregulated glucose and lipid metabolism associated with diabetes mellitus, which leads to increased oxidative stress and the activation of multiple inflammatory pathways that mediate cellular and extracellular injury, pathological cardiac remodelling, and diastolic and systolic dysfunction. Preclinical studies in animal models of diabetes have identified multiple intracellular pathways involved in the pathogenesis of diabetic cardiomyopathy and potential cardioprotective strategies to prevent and treat the disease, including antifibrotic agents, anti-inflammatory agents and antioxidants. Some of these interventions have been tested in clinical trials and have shown favourable initial results. In this Review, we discuss the mechanisms underlying the development of diabetic cardiomyopathy and heart failure in type 1 and type 2 diabetes mellitus, and we summarize the evidence from preclinical and clinical studies that might provide guidance for the development of targeted strategies. We also highlight some of the novel pharmacological therapeutic strategies for the treatment and prevention of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Zhiguo Zhang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Chao Zheng
- The Second Affiliated Hospital Center of Chinese-American Research Institute for Diabetic Complications, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kupper A Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA
- Division of Endocrinology, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Bradley B Keller
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.
- Wendy Novak Diabetes Center, University of Louisville, Norton Children's Hospital, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.
- Department of Radiation Oncology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
36
|
Costa GC, Montagnoli TL, Da Silva JS, de Alencar AKN, Reina Gamba LE, Alves BEO, da Silva MMC, Trachez MM, do Nascimento JHM, Pimentel-Coelho PM, Mendez-Otero R, Lima LM, Barreiro EJ, Sudo RT, Zapata-Sudo G. New Benzofuran N-Acylhydrazone Reduces Cardiovascular Dysfunction in Obese Rats by Blocking TNF-Alpha Synthesis. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3337-3350. [PMID: 32884238 PMCID: PMC7443037 DOI: 10.2147/dddt.s258459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022]
Abstract
Introduction Diabetic obese patients are susceptible to the development of cardiovascular disease, including hypertension and cardiac dysfunction culminating in diabetic cardiomyopathy (DC), which represents a life-threatening health problem with increased rates of morbidity and mortality. The aim of the study is to characterize the effects of a new benzofuran N-acylhydrazone compound, LASSBio-2090, on metabolic and cardiovascular alterations in Zucker diabetic fatty (ZDF) rats presenting DC. Methods Male non-diabetic lean Zucker rats (ZL) and ZDF rats treated with vehicle (dimethylsulfoxide) or LASSBio-2090 were used in this study. Metabolic parameters, cardiovascular function, left ventricle histology and inflammatory protein expression were analyzed in the experimental groups. Results LASSBio-2090 administration in ZDF rats reduced glucose levels to 85.0 ± 1.7 mg/dL (p < 0.05). LASSBio-2090 also lowered the cholesterol and triglyceride levels from 177.8 ± 31.2 to 104.8 ± 5.3 mg/dL and from 123.0 ± 11.4 to 90.9 ± 4.8 mg/dL, respectively, in obese diabetic rats (p < 0.05). LASSBio-2090 normalized plasma insulin, insulin sensitivity and endothelial function in aortas from diabetic animals (p < 0.05). It also enhanced systolic and diastolic left-ventricular function and reverted myocardial remodeling by blocking the threefold elevation of TNF-α levels in hearts from ZDF rats. Conclusion LASSBio-2090 alleviates metabolic disturbance and cardiomyopathy in an obese and diabetic rat model, thus representing a novel strategy for the treatment of cardiovascular complications in obesity-associated type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Gizele Cabral Costa
- Programa de Pós-Graduação em Medicina (Cardiologia), Instituto do Coração Edson Saad, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Tadeu Lima Montagnoli
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Jaqueline Soares Da Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Allan Kardec Nogueira de Alencar
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Luis Eduardo Reina Gamba
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Bryelle Eccard Oliveira Alves
- Programa de Pós-Graduação em Medicina (Cardiologia), Instituto do Coração Edson Saad, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.,Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Marina Moraes Carvalho da Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Margarete Manhães Trachez
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - José Hamilton M do Nascimento
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.,Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Pedro Moreno Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Rosália Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Lidia Moreira Lima
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Eliezer J Barreiro
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Roberto Takashi Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Gisele Zapata-Sudo
- Programa de Pós-Graduação em Medicina (Cardiologia), Instituto do Coração Edson Saad, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.,Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
37
|
Late Peaks of HMGB1 and Sepsis Outcome: Evidence For Synergy With Chronic Inflammatory Disorders. Shock 2020; 52:334-339. [PMID: 30239421 DOI: 10.1097/shk.0000000000001265] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
High mobility group box 1 (HMGB1) is released from macrophages as a late biomarker of sepsis. Conditions associated with pre-existing macrophage activation may modify HMGB1 expression. This study aimed to assess the impact of HMGB1 kinetics on 28-day mortality. In a sub-study of a previous randomized clinical trial among patients with systemic inflammatory response syndrome and gram-negative infections, patients were classified in early and late HMGB1 peak groups. Serial measurements of HMGB1, ferritin and interferon-gamma (IFNγ) were performed in all available sera. Two hundred ten patients were included; 118 (46.5%) had at least one inflammatory disease (diabetes, chronic obstructive pulmonary disease, chronic heart failure, or chronic renal disease). Mortality after 28 days was higher among patients with a late peak of HMGB1 (OR 2.640; P = 0.026). Co-existence of late peak and inflammatory disease synergistically impacted mortality (odds ratio of logistic regression analysis 3.17; P: 0.027). Late peak was concomitantly associated with higher values of ferritin (P = 0.035), and IFNγ (P = 0.002) among patients with hyperferritinemia. It is concluded that late HMGB1 peak was associated with worse prognosis, especially in patients with underlying chronic inflammatory conditions.
Collapse
|
38
|
Scavello F, Zeni F, Tedesco CC, Mensà E, Veglia F, Procopio AD, Bonfigli AR, Olivieri F, Raucci A. Modulation of soluble receptor for advanced glycation end-products (RAGE) isoforms and their ligands in healthy aging. Aging (Albany NY) 2020; 11:1648-1663. [PMID: 30903794 PMCID: PMC6461165 DOI: 10.18632/aging.101860] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/06/2019] [Indexed: 02/07/2023]
Abstract
The receptor for advanced glycation end-products (RAGE) recognizes several ligands involved in inflammatory diseases. Two circulating soluble isoforms exist: esRAGE derived from alternative splicing and cRAGE generated by the membrane-bound RAGE (FL-RAGE) proteolysis. Together, esRAGE and cRAGE constitute sRAGE and function as decoy receptors preventing FL-RAGE/ligands binding. We determined serum concentration of both, esRAGE and cRAGE, and their ligands AGEs, HMGB1 and S100A8/A9 in a healthy population of 169 subjects aged 20-90 years. cRAGE showed a negative (r=-0.375, P<0.0001) while AGEs (r=0.160, P=0.0384) and S100A8/A9 (r=0.207, P=0.0091) a positive correlation with age. esRAGE did not change during aging and inversely correlated with Hemoglobin, ALT, insulin, HOMA index, Waist-Hip ratio (W/H), Waist Circumference (WC) and positively with AGEs. cRAGE exhibited also an inverse correlation with WC, W/H, PAI-1, HMGB1, AGEs and S100A8/A9. Age, W/H, HMGB1, S100A8/A9 and AGEs are independent predictors of cRAGE, whereas W/H and AGEs associate with esRAGE. Treatment of cells with glycated albumin reduced cRAGE production and upregulated FL-RAGE. These results indicate that in a healthy population cRAGE is a biomarker of aging while esRAGE represents a more reliable marker of obesity and insulin resistance. Hence, sRAGE isoforms levels could be differentially associated with age-related diseases risk factors.
Collapse
Affiliation(s)
- Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Filippo Zeni
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | - Emanuela Mensà
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | | | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| |
Collapse
|
39
|
Wen Y, Sun HY, Tan Z, Liu RH, Huang SQ, Chen GY, Qi H, Tang LJ. Abdominal paracentesis drainage ameliorates myocardial injury in severe experimental pancreatitis rats through suppressing oxidative stress. World J Gastroenterol 2020; 26:35-54. [PMID: 31933513 PMCID: PMC6952299 DOI: 10.3748/wjg.v26.i1.35] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/06/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Abdominal paracentesis drainage (APD) is a safe and effective strategy for severe acute pancreatitis (SAP) patients. However, the effects of APD treatment on SAP-associated cardiac injury remain unknown.
AIM To investigate the protective effects of APD on SAP-associated cardiac injury and the underlying mechanisms.
METHODS SAP was induced by 5% sodium taurocholate retrograde injection in Sprague-Dawley rats. APD was performed by inserting a drainage tube with a vacuum ball into the lower right abdomen of the rats immediately after SAP induction. Morphological staining, serum amylase and inflammatory mediators, serum and ascites high mobility group box (HMGB) 1, cardiac-related enzymes indexes and cardiac function, oxidative stress markers and apoptosis and associated proteins were assessed in the myocardium in SAP rats. Nicotinamide adenine dinucleotide phosphate oxidase activity and mRNA and protein expression were also examined.
RESULTS APD treatment improved cardiac morphological changes, inhibited cardiac dysfunction, decreased cardiac enzymes and reduced cardiomyocyte apoptosis, proapoptotic Bax and cleaved caspase-3 protein levels. APD significantly decreased serum levels of HMGB1, inhibited nicotinamide adenine dinucleotide phosphate oxidase expression and ultimately alleviated cardiac oxidative injury. Furthermore, the activation of cardiac nicotinamide adenine dinucleotide phosphate oxidase by pancreatitis-associated ascitic fluid intraperitoneal injection was effectively inhibited by adding anti-HMGB1 neutralizing antibody in rats with mild acute pancreatitis.
CONCLUSION APD treatment could exert cardioprotective effects on SAP-associated cardiac injury through suppressing HMGB1-mediated oxidative stress, which may be a novel mechanism behind the effectiveness of APD on SAP.
Collapse
Affiliation(s)
- Yi Wen
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu 610083, Sichuan Province, China
| | - Hong-Yu Sun
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu 610083, Sichuan Province, China
| | - Zhen Tan
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu 610083, Sichuan Province, China
| | - Ruo-Hong Liu
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu 610083, Sichuan Province, China
| | - Shang-Qing Huang
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu 610083, Sichuan Province, China
| | - Guang-Yu Chen
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu 610083, Sichuan Province, China
| | - Hao Qi
- Department of Dermatology, The Air Force Hospital of Western Theater Command, Chengdu 610083, Sichuan Province, China
| | - Li-Jun Tang
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu 610083, Sichuan Province, China
| |
Collapse
|
40
|
Biscetti F, Rando MM, Nardella E, Cecchini AL, Pecorini G, Landolfi R, Flex A. High Mobility Group Box-1 and Diabetes Mellitus Complications: State of the Art and Future Perspectives. Int J Mol Sci 2019; 20:ijms20246258. [PMID: 31835864 PMCID: PMC6940913 DOI: 10.3390/ijms20246258] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 12/17/2022] Open
Abstract
Diabetes mellitus (DM) is an endemic disease, with growing health and social costs. The complications of diabetes can affect potentially all parts of the human body, from the heart to the kidneys, peripheral and central nervous system, and the vascular bed. Although many mechanisms have been studied, not all players responsible for these complications have been defined yet. High Mobility Group Box-1 (HMGB1) is a non-histone nuclear protein that has been implicated in many pathological processes, from sepsis to ischemia. The purpose of this review is to take stock of all the most recent data available on the role of HMGB1 in the complications of DM.
Collapse
Affiliation(s)
- Federico Biscetti
- U.O.C. Clinica Medica e Malattie Vascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (G.P.); (R.L.); (A.F.)
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Correspondence: ; Tel.: +39-06-3015-4335; Fax: +39-06-3550-7232
| | | | - Elisabetta Nardella
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.M.R.); (E.N.); (A.L.C.)
| | | | - Giovanni Pecorini
- U.O.C. Clinica Medica e Malattie Vascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (G.P.); (R.L.); (A.F.)
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.M.R.); (E.N.); (A.L.C.)
| | - Raffaele Landolfi
- U.O.C. Clinica Medica e Malattie Vascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (G.P.); (R.L.); (A.F.)
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.M.R.); (E.N.); (A.L.C.)
| | - Andrea Flex
- U.O.C. Clinica Medica e Malattie Vascolari, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (G.P.); (R.L.); (A.F.)
- Laboratory of Vascular Biology and Genetics, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (M.M.R.); (E.N.); (A.L.C.)
| |
Collapse
|
41
|
Gao W, Cui H, Li Q, Zhong H, Yu J, Li P, He X. Upregulation of microRNA-218 reduces cardiac microvascular endothelial cells injury induced by coronary artery disease through the inhibition of HMGB1. J Cell Physiol 2019; 235:3079-3095. [PMID: 31566720 DOI: 10.1002/jcp.29214] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 08/23/2019] [Indexed: 12/19/2022]
Abstract
This study is performed to examine the impacts of microRNA-218 (miR-218) on cardiac microvascular endothelial cells (CMECs) injury induced by coronary artery disease (CAD). Reverse-transcription quantitative polymerase chain reaction (RT-qPCR) was applied for detecting miR-218 expression in serum of patients with CAD and healthy controls, and the correlation between miR-218 expression and the clinical indexes such as creatine kinase, creatine kinase-myocardial band, cardiac troponin I, and coronary Gensini score was analyzed. CMECs were coincubated with homocysteine for 24 hr for CMECs injury, and the cells were transfected with miR-218 mimics or miR-218 inhibitors. Besides, we used oxidized low density lipoprotein as an inducer to incubate with CMECs for 24 hr, and the model of CMECs injury was established to be transfected with miR-218 mimics. RT-qPCR and western blot analysis were used to detect miR-218 and HMGB1 expression in CMECs. A series of experiments were used to determine cell proliferation, apoptosis, migration, and angiogenesis ability of CMECs. Vascular endothelial growth factor expression and inflammatory factor contents were measured. The obtained results suggested that miR-218 expression in peripheral blood of patients with CAD descended substantially versus that of healthy controls. Low miR-218 expression was found in CAD-induced CMECs injury. Overexpressed miR-218 promoted the proliferation, migration, angiogenesis ability, induced apoptosis, and alleviated the inflammatory injury of CAD-induced CMECs. miR-218 may negatively regulate the expression of HMGB1 in CAD. This study demonstrates that upregulation of miR-218 reduces CMECs injury induced by CAD through the inhibition of HMGB1.
Collapse
Affiliation(s)
- Wenhui Gao
- Department of Cardiovascular, Hangzhouwan Hospital, Ningbo, Zhejiang Province, China
| | - Hanbin Cui
- Department of Cardiovascular, No. 1 Hospital, Ningbo, Zhejiang Province, China
| | - Qianjun Li
- Department of Respiratory, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| | - Hai Zhong
- Department of Thoracic Surgery, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| | - Jingjing Yu
- Department of Pathology, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| | - Ping Li
- Department of Anesthesiology, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| | - Xijie He
- Department of Cardiology, No. 2 Hospital Yinzhou County, Ningbo, Zhejiang Province, China
| |
Collapse
|
42
|
Raucci A, Di Maggio S, Scavello F, D'Ambrosio A, Bianchi ME, Capogrossi MC. The Janus face of HMGB1 in heart disease: a necessary update. Cell Mol Life Sci 2019; 76:211-229. [PMID: 30306212 PMCID: PMC6339675 DOI: 10.1007/s00018-018-2930-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 12/23/2022]
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein involved in transcription regulation, DNA replication and repair and nucleosome assembly. HMGB1 is passively released by necrotic tissues or actively secreted by stressed cells. Extracellular HMGB1 acts as a damage-associated molecular pattern (DAMPs) molecule and gives rise to several redox forms that by binding to different receptors and interactors promote a variety of cellular responses, including tissue inflammation or regeneration. Inhibition of extracellular HMGB1 in experimental models of myocardial ischemia/reperfusion injury, myocarditis, cardiomyopathies induced by mechanical stress, diabetes, bacterial infection or chemotherapeutic drugs reduces inflammation and is protective. In contrast, administration of HMGB1 after myocardial infarction induced by permanent coronary artery ligation ameliorates cardiac performance by promoting tissue regeneration. HMGB1 decreases contractility and induces hypertrophy and apoptosis in cardiomyocytes, stimulates cardiac fibroblast activities, and promotes cardiac stem cell proliferation and differentiation. Interestingly, maintenance of appropriate nuclear HMGB1 levels protects cardiomyocytes from apoptosis by preventing DNA oxidative stress, and mice with HMGB1cardiomyocyte-specific overexpression are partially protected from cardiac damage. Finally, higher levels of circulating HMGB1 are associated to human heart diseases. Hence, during cardiac injury, HMGB1 elicits both harmful and beneficial responses that may in part depend on the generation and stability of the diverse redox forms, whose specific functions in this context remain mostly unexplored. This review summarizes recent findings on HMGB1 biology and heart dysfunctions and discusses the therapeutic potential of modulating its expression, localization, and oxidative-dependent activities.
Collapse
Affiliation(s)
- Angela Raucci
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy.
| | - Stefania Di Maggio
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
| | - Francesco Scavello
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
| | - Alessandro D'Ambrosio
- Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Marco E Bianchi
- Chromatin Dynamics Unit, Università Vita-Salute San Raffaele, Milan, Italy
| | - Maurizio C Capogrossi
- Department of Cardiology, Ochsner Medical Center, New Orleans, USA
- Division of Cardiology, Johns Hopkins Bayview Medical Center, Baltimore, USA
| |
Collapse
|
43
|
Inhibition of HMGB1 ameliorates cardiac fibrosis through regulation of endothelial-to-mesenchymal transition. Int J Cardiol 2019; 274:282. [DOI: 10.1016/j.ijcard.2018.07.148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 07/28/2018] [Accepted: 07/31/2018] [Indexed: 11/19/2022]
|
44
|
Yadav SK, Kambis TN, Mishra PK. Regulating Inflammatory Cytokines in the Diabetic Heart. OXIDATIVE STRESS IN HEART DISEASES 2019:427-436. [DOI: 10.1007/978-981-13-8273-4_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
45
|
Abstract
High-mobility group box 1 (HMGB1) is one of the most abundant proteins in eukaryotes and the best characterized damage-associated molecular pattern (DAMP). The biological activities of HMGB1 depend on its subcellular location, context and post-translational modifications. Inside the nucleus, HMGB1 is engaged in many DNA events such as DNA repair, transcription regulation and genome stability; in the cytoplasm, its main function is to regulate the autophagic flux while in the extracellular environment, it possesses more complicated functions and it is involved in a large variety of different processes such as inflammation, migration, invasion, proliferation, differentiation and tissue regeneration. Due to this pleiotropy, the role of HMGB1 has been vastly investigated in various pathological diseases and a large number of studies have explored its function in cardiovascular pathologies. However, in this contest, the precise mechanism of action of HMGB1 and its therapeutic potential are still very controversial since is debated whether HMGB1 is involved in tissue damage or plays a role in tissue repair and regeneration. The main focus of this review is to provide an overview of the effects of HMGB1 in different ischemic heart diseases and to discuss its functions in these pathological conditions.
Collapse
|
46
|
Zhang H, Lu X, Liu Z, Du K. Rosuvastatin reduces the pro-inflammatory effects of adriamycin on the expression of HMGB1 and RAGE in rats. Int J Mol Med 2018; 42:3415-3423. [PMID: 30320373 PMCID: PMC6202077 DOI: 10.3892/ijmm.2018.3928] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 10/03/2018] [Indexed: 12/22/2022] Open
Abstract
Rosuvastatin has cardiac protective effects through its anti‑inflammatory effects. The nuclear protein high‑mobility group box 1 (HMGB1) can activate inflammatory pathways when released from dying cells. The present study aimed to investigate the effects of rosuvastatin in adriamycin (ADR)‑treated rats. Adult male rats were randomized to three groups: i) Control group, ii) ADR group, and iii) ADR+rosuvastatin group. Serum biochemical indices were measured using an enzyme‑linked immunosorbent assay. Cardiac function was assessed by echocardiography. The expression of HMGB1 and receptors for advanced glycation end products (RAGE) were assessed by reverse transcription‑quantitative polymerase chain reaction analysis, western blot analysis, and immunohistochemistry. Cytokines were measured using flow cytometry. Rosuvastatin improved the biochemical indices and cardiac morphology and alleviated the pathological lesions. In the ADR+rosuvastatin group, the mRNA and protein levels of HMGB1 and RAGE in the myocardium were significantly lower compared with those in the ADR group (both P<0.05). The results showed that rosuvastatin significantly reduced the levels of HMGB1 and RAGE in the myocardium of the ADR‑treated rats. These results suggest that the protective effects of rosuvastatin may be associated with attenuation of the HMGB1/RAGE‑mediated inflammatory response in ADR‑treated rats. Despite this protective effect of rosuvastatin in the present study, it did not improve cardiac function in terms of the diastolic left ventricular internal dimension, systolic left ventricular internal dimension, left ventricular ejection fraction and left ventricular fractional shortening; this may be due the observation duration being insufficient.
Collapse
Affiliation(s)
- Haiyan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiang Lu
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhengxia Liu
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Kang Du
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
47
|
Yu SY, Dong B, Fang ZF, Hu XQ, Tang L, Zhou SH. Knockdown of lncRNA AK139328 alleviates myocardial ischaemia/reperfusion injury in diabetic mice via modulating miR-204-3p and inhibiting autophagy. J Cell Mol Med 2018; 22:4886-4898. [PMID: 30047214 PMCID: PMC6156366 DOI: 10.1111/jcmm.13754] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/10/2018] [Indexed: 01/13/2023] Open
Abstract
This study was aimed at investigating the effects of lncRNA AK139328 on myocardial ischaemia/reperfusion injury (MIRI) in diabetic mice. Ischaemia/reperfusion (I/R) model was constructed in normal mice (NM) and diabetic mice (DM). Microarray analysis was utilized to identify lncRNA AK139328 overexpressed in DM after myocardial ischaemia/reperfusion (MI/R). RT‐qPCR assay was utilized to investigate the expressions of lncRNA AK139328 and miR‐204‐3p in cardiomyocyte and tissues. Left ventricular end diastolic diameter (LVEDD), left ventricular end systolic diameter (LVESD), left ventricular ejection fraction (LVEF) and fractioning shortening (FS) were obtained by transthoracic echocardiography. Haematoxylin‐eosin (HE) staining and Masson staining were utilized to detect the damage of myocardial tissues degradation of myocardial fibres and integrity of myocardial collagen fibres. Evans Blue/TTC staining was used to determine the myocardial infarct size. TUNEL staining was utilized to investigate cardiomyocyte apoptosis. The targeted relationship between lncRNA AK139328 and miR‐204‐3p was confirmed by dual‐luciferase reporter gene assay. MTT assay was used for analysis of cardiomyocyte proliferation. Western blot was utilized to investigate the expression of alpha smooth muscle actin (α‐SMA), Atg7, Atg5, LC3‐II/LC3‐I and p62 marking autophagy. Knockdown of lncRNA AK139328 relieved myocardial ischaemia/reperfusion injury in DM and inhibited cardiomyocyte autophagy as well as apoptosis of DM. LncRNA AK139328 modulated miR‐204‐3p directly. MiR‐204‐3p and knockdown of lncRNA AK139328 relieved hypoxia/reoxygenation injury via inhibiting cardiomyocyte autophagy. Silencing lncRNA AK139328 significantly increased miR‐204‐3p expression and inhibited cardiomyocyte autophagy, thereby attenuating MIRI in DM.
Collapse
Affiliation(s)
- Si-Yang Yu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bo Dong
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhen-Fei Fang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin-Qun Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liang Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Sheng-Hua Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
48
|
Oikonomou E, Mourouzis K, Fountoulakis P, Papamikroulis GA, Siasos G, Antonopoulos A, Vogiatzi G, Tsalamadris S, Vavuranakis M, Tousoulis D. Interrelationship between diabetes mellitus and heart failure: the role of peroxisome proliferator-activated receptors in left ventricle performance. Heart Fail Rev 2018; 23:389-408. [PMID: 29453696 DOI: 10.1007/s10741-018-9682-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heart failure (HF) is a common cardiac syndrome, whose pathophysiology involves complex mechanisms, some of which remain unknown. Diabetes mellitus (DM) constitutes not only a glucose metabolic disorder accompanied by insulin resistance but also a risk factor for cardiovascular disease and HF. During the last years though emerging data set up, a bidirectional interrelationship between these two entities. In the case of DM impaired calcium homeostasis, free fatty acid metabolism, redox state, and advance glycation end products may accelerate cardiac dysfunction. On the other hand, when HF exists, hypoperfusion of the liver and pancreas, b-blocker and diuretic treatment, and autonomic nervous system dysfunction may cause impairment of glucose metabolism. These molecular pathways may be used as therapeutic targets for novel antidiabetic agents. Peroxisome proliferator-activated receptors (PPARs) not only improve insulin resistance and glucose and lipid metabolism but also manifest a diversity of actions directly or indirectly associated with systolic or diastolic performance of left ventricle and symptoms of HF. Interestingly, they may beneficially affect remodeling of the left ventricle, fibrosis, and diastolic performance but they may cause impaired water handing, sodium retention, and decompensation of HF which should be taken into consideration in the management of patients with DM. In this review article, we present the pathophysiological data linking HF with DM and we focus on the molecular mechanisms of PPARs agonists in left ventricle systolic and diastolic performance providing useful insights in the molecular mechanism of this class of metabolically active regiments.
Collapse
Affiliation(s)
- Evangelos Oikonomou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece.
| | - Konstantinos Mourouzis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| | - Petros Fountoulakis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| | - Georgios Angelos Papamikroulis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| | - Gerasimos Siasos
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| | - Alexis Antonopoulos
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| | - Georgia Vogiatzi
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| | - Sotiris Tsalamadris
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| | - Manolis Vavuranakis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Vasilissis Sofias 114, TK, 115 28, Athens, Greece
| |
Collapse
|
49
|
Palomer X, Pizarro-Delgado J, Vázquez-Carrera M. Emerging Actors in Diabetic Cardiomyopathy: Heartbreaker Biomarkers or Therapeutic Targets? Trends Pharmacol Sci 2018; 39:452-467. [PMID: 29605388 DOI: 10.1016/j.tips.2018.02.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/20/2018] [Accepted: 02/27/2018] [Indexed: 12/14/2022]
Abstract
The diabetic heart is characterized by metabolic disturbances that are often accompanied by local inflammation, oxidative stress, myocardial fibrosis, and cardiomyocyte apoptosis. Overall changes result in contractile dysfunction, concentric left ventricular (LV) hypertrophy, and dilated cardiomyopathy, that together affect cardiac output and eventually lead to heart failure, the foremost cause of death in diabetic patients. There are currently several validated biomarkers for the diagnosis and risk assessment of cardiac diseases, but none is capable of discriminating patients with diabetic cardiomyopathy (DCM). In this review we point to several novel candidate biomarkers from new activated molecular pathways (including microRNAs) with the potential to detect or prevent DCM in its early stages, or even to treat it once established. The prospective use of selected biomarkers that integrate inflammation, oxidative stress, fibrosis, and metabolic dysregulation is widely discussed.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Javier Pizarro-Delgado
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain; Research Institute, Hospital Sant Joan de Déu, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
50
|
Sanmarco LM, Eberhardt N, Ponce NE, Cano RC, Bonacci G, Aoki MP. New Insights into the Immunobiology of Mononuclear Phagocytic Cells and Their Relevance to the Pathogenesis of Cardiovascular Diseases. Front Immunol 2018; 8:1921. [PMID: 29375564 PMCID: PMC5767236 DOI: 10.3389/fimmu.2017.01921] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022] Open
Abstract
Macrophages are the primary immune cells that reside within the myocardium, suggesting that these mononuclear phagocytes are essential in the orchestration of cardiac immunity and homeostasis. Independent of the nature of the injury, the heart triggers leukocyte activation and recruitment. However, inflammation is harmful to this vital terminally differentiated organ with extremely poor regenerative capacity. As such, cardiac tissue has evolved particular strategies to increase the stress tolerance and minimize the impact of inflammation. In this sense, growing evidences show that mononuclear phagocytic cells are particularly dynamic during cardiac inflammation or infection and would actively participate in tissue repair and functional recovery. They respond to soluble mediators such as metabolites or cytokines, which play central roles in the timing of the intrinsic cardiac stress response. During myocardial infarction two distinct phases of monocyte influx have been identified. Upon infarction, the heart modulates its chemokine expression profile that sequentially and actively recruits inflammatory monocytes, first, and healing monocytes, later. In the same way, a sudden switch from inflammatory macrophages (with microbicidal effectors) toward anti-inflammatory macrophages occurs within the myocardium very shortly after infection with Trypanosoma cruzi, the causal agent of Chagas cardiomyopathy. While in sterile injury, healing response is necessary to stop tissue damage; during an intracellular infection, the anti-inflammatory milieu in infected hearts would promote microbial persistence. The balance of mononuclear phagocytic cells seems to be also dynamic in atherosclerosis influencing plaque initiation and fate. This review summarizes the participation of mononuclear phagocyte system in cardiovascular diseases, keeping in mind that the immune system evolved to promote the reestablishment of tissue homeostasis following infection/injury, and that the effects of different mediators could modulate the magnitude and quality of the immune response. The knowledge of the effects triggered by diverse mediators would serve to identify new therapeutic targets in different cardiovascular pathologies.
Collapse
Affiliation(s)
- Liliana Maria Sanmarco
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Natalia Eberhardt
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Nicolás Eric Ponce
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Roxana Carolina Cano
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Universidad Católica de Córdoba, Unidad Asociada Área Ciencias Agrarias, Ingeniería, Ciencias Biológicas y de la Salud, Facultad de Ciencias Químicas, Córdoba, Argentina
| | - Gustavo Bonacci
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Maria Pilar Aoki
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| |
Collapse
|