1
|
Singh A, Vasudevan S, Palle AR, Atchuta A, Bhadauriya S. Comparative Evaluation of Scaling and Root Planing with and without Oxygen-releasing Gel in the Treatment of Chronic Periodontitis: A Split-mouth Study. J Contemp Dent Pract 2024; 25:445-452. [PMID: 39364843 DOI: 10.5005/jp-journals-10024-3689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
AIM The study aims to compare the effectiveness of scaling root planing alone and scaling root planing with oxygen-releasing gel in the treatment of chronic periodontitis. MATERIALS AND METHODS A split-mouth randomized controlled trial was designed on 25 systemically healthy participants with 50 sites having chronic periodontitis. Two sites were selected for each patient and were randomly allocated into two groups. A total of 50 sites were selected and divided into two groups with 25 test sites in group I (Test group) and 25 control sites in group II (Control group). Group I received scaling and root planing (SRP) followed by placement of BlueM oral gel, while group II received SRP alone. Probing pocket depth (PPD), clinical attachment level (CAL), gingival index (GI), and bleeding on probing (BOP) were recorded at baseline and after 6 weeks and were compared accordingly. RESULTS The results revealed a reduction in PPD, CAL, GI, and BOP in both groups. However, the test group showed a statistically significant reduction when compared with the control group in the above-mentioned clinical parameters. CONCLUSION The results suggested that the use of oxygen-releasing gel with SRP provided additional benefits in the management of chronic periodontitis by promoting greater reductions in the clinical parameters. CLINICAL SIGNIFICANCE BlueM oral gel, i.e. the oxygen-releasing gel as a local drug delivery, may be an excellent adjunct to SRP in treating chronic periodontitis. How to cite this article: Singh A, Vasudevan S, Palle AR, et al. Comparative Evaluation of Scaling and Root Planing with and without Oxygen-releasing Gel in the Treatment of Chronic Periodontitis: A Split-mouth Study. J Contemp Dent Pract 2024;25(5):445-452.
Collapse
Affiliation(s)
- Anuradha Singh
- Department of Periodontics, Army College of Dental Sciences, Secunderabad, Telangana, India, Phone: +91 8309931210, e-mail: , Orcid: https://orcid.org/0009-0009-4652-7792
| | - Sanjay Vasudevan
- Department of Periodontics, Army College of Dental Sciences, Secunderabad, Telangana, India, Orcid: https://orcid.org/0009-0004-7733-2393
| | - Ajay Reddy Palle
- Department of Periodontics, Army College of Dental Sciences, Secunderabad, Telangana, India, Orcid: https://orcid.org/0009-0008-8948-7084
| | - Abhinav Atchuta
- Department of Periodontics, Army College of Dental Sciences, Secunderabad, Telangana, India, Orcid: https://orcid.org/0000-0001-9295-0289
| | - Surabhi Bhadauriya
- Department of Periodontics, Army College of Dental Sciences, Secunderabad, Telangana, India, Orcid: https://orcid.org/0000-0003-3132-8773
| |
Collapse
|
2
|
Packer M. SGLT2 inhibitors: role in protective reprogramming of cardiac nutrient transport and metabolism. Nat Rev Cardiol 2023; 20:443-462. [PMID: 36609604 DOI: 10.1038/s41569-022-00824-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 01/09/2023]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce heart failure events by direct action on the failing heart that is independent of changes in renal tubular function. In the failing heart, nutrient transport into cardiomyocytes is increased, but nutrient utilization is impaired, leading to deficient ATP production and the cytosolic accumulation of deleterious glucose and lipid by-products. These by-products trigger downregulation of cytoprotective nutrient-deprivation pathways, thereby promoting cellular stress and undermining cellular survival. SGLT2 inhibitors restore cellular homeostasis through three complementary mechanisms: they might bind directly to nutrient-deprivation and nutrient-surplus sensors to promote their cytoprotective actions; they can increase the synthesis of ATP by promoting mitochondrial health (mediated by increasing autophagic flux) and potentially by alleviating the cytosolic deficiency in ferrous iron; and they might directly inhibit glucose transporter type 1, thereby diminishing the cytosolic accumulation of toxic metabolic by-products and promoting the oxidation of long-chain fatty acids. The increase in autophagic flux mediated by SGLT2 inhibitors also promotes the clearance of harmful glucose and lipid by-products and the disposal of dysfunctional mitochondria, allowing for mitochondrial renewal through mitochondrial biogenesis. This Review describes the orchestrated interplay between nutrient transport and metabolism and nutrient-deprivation and nutrient-surplus signalling, to explain how SGLT2 inhibitors reverse the profound nutrient, metabolic and cellular abnormalities observed in heart failure, thereby restoring the myocardium to a healthy molecular and cellular phenotype.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Dallas, TX, USA.
- Imperial College London, London, UK.
| |
Collapse
|
3
|
Xiao W, Ahluwalia P, Wang L, Howard J, Kolhe R, Rojiani AM, Rojiani MV. TIMP-1 Dependent Modulation of Metabolic Profiles Impacts Chemoresistance in NSCLC. Cells 2022; 11:cells11193036. [PMID: 36230997 PMCID: PMC9562647 DOI: 10.3390/cells11193036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/28/2022] Open
Abstract
The development of chemoresistance remains a significant barrier to treating NSCLC. Alteration of cancer cell metabolism is an important mechanism for chemoresistance. This study explored the role of aberrant metabolism in TIMP-1-mediated chemoresistance. Bioinformatics analysis identified an association of high TIMP-1 with altered energy metabolism. We have defined the role of depolarized mitochondria through a reduction in lactate secretion, higher ROS levels in TIMP-1 KD cells and reduced GSH levels. TIMP-1 modulates the metabolic profile via acetylation of mitochondrial STAT3 and its interaction with CD44. Intriguingly, monomers of acetylated STAT3 were critical for altered metabolism, whereas STAT3 dimers abrogated this function. Further, the mitochondrial metabolic profile was also altered in a cisplatin-resistant clone of A549 cells. We also correlated the immunoexpression of CD44, STAT3 and TIMP-1 in patient samples. This study provided evidence that TIMP-1 alters the metabolic profile by modulating mitochondrial metabolism via the CD44-STAT3 axis through its effects on STAT3 acetylation. It also lent further support to the critical role of TIMP-1 in chemoresistance. Interrogation of the TCGA-LUAD dataset revealed perturbations in the critical modulator that can alter metabolic states in cancer cells. Higher expression of a five-gene signature, including TIMP-1, correlated with immunosuppressive cells and was found to be associated with overall survival. This study identified several metabolic mechanisms that could influence therapeutic options and prognosis in NSCLC patients.
Collapse
Affiliation(s)
- Wei Xiao
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Lan Wang
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - John Howard
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Amyn M. Rojiani
- Department of Pathology, Penn State College of Medicine, Hershey, PA 17033, USA
- Room T3409, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Mumtaz V. Rojiani
- Department of Pathology, Penn State College of Medicine, Hershey, PA 17033, USA
- Room T3409, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033, USA
- Correspondence: ; Tel.: +1-717-531-0003 (ext. 322422)
| |
Collapse
|
4
|
Ma X, Dong Z, Liu J, Ma L, Sun X, Gao R, Pan L, Zhang J, A D, An J, Hu K, Sun A, Ge J. β-Hydroxybutyrate Exacerbates Hypoxic Injury by Inhibiting HIF-1α-Dependent Glycolysis in Cardiomyocytes-Adding Fuel to the Fire? Cardiovasc Drugs Ther 2022; 36:383-397. [PMID: 34652582 PMCID: PMC9090701 DOI: 10.1007/s10557-021-07267-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/16/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Ketone body oxidation yields more ATP per mole of consumed oxygen than glucose. However, whether an increased ketone body supply in hypoxic cardiomyocytes and ischemic hearts is protective or not remains elusive. The goal of this study is to determine the effect of β-hydroxybutyrate (β-OHB), the main constituent of ketone bodies, on cardiomyocytes under hypoxic conditions and the effects of ketogenic diet (KD) on cardiac function in a myocardial infarction (MI) mouse model. METHODS Human peripheral blood collected from patients with acute myocardial infarction and healthy volunteers was used to detect the level of β-OHB. N-terminal proB-type natriuretic peptide (NT-proBNP) levels and left ventricular ejection fractions (LVEFs) were measured to study the relationship between plasma β-OHB and cardiac function. Adult mouse cardiomyocytes and MI mouse models fed a KD were used to research the effect of β-OHB on cardiac damage. qPCR, western blot analysis, and immunofluorescence were used to detect the interaction between β-OHB and glycolysis. Live/dead cell staining and imaging, lactate dehydrogenase, Cell Counting Kit-8 assays, echocardiography, and 2,3,5-triphenyltetrazolium chloride staining were performed to evaluate the cardiomyocyte death, cardiac function, and infarct sizes. RESULTS β-OHB level was significantly higher in acute MI patients and MI mice. Treatment with β-OHB exacerbated cardiomyocyte death and decreased glucose absorption and glycolysis under hypoxic conditions. These effects were partially ameliorated by inhibiting hypoxia-inducible factor 1α (HIF-1α) degradation via roxadustat administration in hypoxia-stimulated cardiomyocytes. Furthermore, β-OHB metabolisms were obscured in cardiomyocytes under hypoxic conditions. Additionally, MI mice fed a KD exhibited exacerbated cardiac dysfunction compared with control chow diet (CD)-fed MI mice. CONCLUSION Elevated β-OHB levels may be maladaptive to the heart under hypoxic/ischemic conditions. Administration of roxadustat can partially reverse these harmful effects by stabilizing HIF-1α and inducing a metabolic shift toward glycolysis for energy production.
Collapse
Affiliation(s)
- Xiurui Ma
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China
- Department of Cardiology, Shan Xi Cardiovascular Hospital, Taiyuan, 030024, China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- NHC Key Laboratory of Viral Heart Diseases and Key Laboratory of Viral Heart Diseases, Shanghai, China
- Academy of Medical Sciences Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jingyi Liu
- Department of Cardiology, Shan Xi Cardiovascular Hospital, Taiyuan, 030024, China
| | - Leilei Ma
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- NHC Key Laboratory of Viral Heart Diseases and Key Laboratory of Viral Heart Diseases, Shanghai, China
| | - Rifeng Gao
- Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200032, China
| | - Lihong Pan
- Academy of Medical Sciences Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jinyan Zhang
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Dilan A
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Jian An
- Department of Cardiology, Shan Xi Cardiovascular Hospital, Taiyuan, 030024, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- NHC Key Laboratory of Viral Heart Diseases and Key Laboratory of Viral Heart Diseases, Shanghai, China.
- Academy of Medical Sciences Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Human Phenome Institute, Fudan University, Shanghai, 201203, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- NHC Key Laboratory of Viral Heart Diseases and Key Laboratory of Viral Heart Diseases, Shanghai, China
- Academy of Medical Sciences Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| |
Collapse
|
5
|
Jiang M, Xie X, Cao F, Wang Y. Mitochondrial Metabolism in Myocardial Remodeling and Mechanical Unloading: Implications for Ischemic Heart Disease. Front Cardiovasc Med 2021; 8:789267. [PMID: 34957264 PMCID: PMC8695728 DOI: 10.3389/fcvm.2021.789267] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Ischemic heart disease refers to myocardial degeneration, necrosis, and fibrosis caused by coronary artery disease. It can lead to severe left ventricular dysfunction (LVEF ≤ 35–40%) and is a major cause of heart failure (HF). In each contraction, myocardium is subjected to a variety of mechanical forces, such as stretch, afterload, and shear stress, and these mechanical stresses are clinically associated with myocardial remodeling and, eventually, cardiac outcomes. Mitochondria produce 90% of ATP in the heart and participate in metabolic pathways that regulate the balance of glucose and fatty acid oxidative phosphorylation. However, altered energetics and metabolic reprogramming are proved to aggravate HF development and progression by disturbing substrate utilization. This review briefly summarizes the current insights into the adaptations of cardiomyocytes to mechanical stimuli and underlying mechanisms in ischemic heart disease, with focusing on mitochondrial metabolism. We also discuss how mechanical circulatory support (MCS) alters myocardial energy metabolism and affects the detrimental metabolic adaptations of the dysfunctional myocardium.
Collapse
Affiliation(s)
- Min Jiang
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaoye Xie
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Cadre Ward, The 960 Hospital of Chinese People's Liberation Army, Jinan, China
| | - Feng Cao
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yabin Wang
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
6
|
Ou W, Liang Y, Qin Y, Wu W, Xie M, Zhang Y, Zhang Y, Ji L, Yu H, Li T. Hypoxic acclimation improves cardiac redox homeostasis and protects heart against ischemia-reperfusion injury through upregulation of O-GlcNAcylation. Redox Biol 2021; 43:101994. [PMID: 33964586 PMCID: PMC8121980 DOI: 10.1016/j.redox.2021.101994] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/11/2021] [Accepted: 04/22/2021] [Indexed: 02/05/2023] Open
Abstract
Ischemia-reperfusion (I/R) injury is detrimental to cardiovascular system. Alteration in glucose metabolism has been recognized as an important adaptive response under hypoxic conditions. However, the biological benefits underlying this metabolic phenotype remain to be elucidated. This study was designed to investigate the impact of hypoxic acclimation (HA) on cardiac I/R injury and the antioxidative mechanism(s). Male adult mice were acclimated in a hypoxic chamber (10% oxygen [O2]) for 8 h/day for 14 days, and then subjected to cardiac I/R injury by ligation of left anterior descending coronary artery for 30 min and reperfusion for 24 h or 7 days. Our results showed that HA attenuated oxidative stress and reduced infarct size in the I/R hearts. This cardioprotective effect is coupled with an elevation of protein O-linked N-acetylglucosamine (O-GlcNAc) modification partially due to inflammatory stimulation. Hyperglycosylation activated glucose-6-phosphate dehydrogenase (G6PDH), the rate-limiting enzyme in the pentose phosphate pathway, resulting in an upregulation of NADPH/NADP+ and GSH/GSSG couples and enhancement of redox homeostasis in the heart. Pharmacological suppression of O-GlcNAcylation totally abolished the influence of HA on the G6PDH activity, redox balance and post-I/R damage in the hearts and cultured cardiomyocytes, whereby augmentation of O-GlcNAcylation further enhanced the benefits, suggesting a central role of O-GlcNAcylation in HA-initiated antioxidative and cardioprotective effects. These findings, therefore, identified HA as a promising anti-I/R strategy for the heart and proposed O-GlcNAc modification of G6PDH as a therapeutic target in ischemic heart disease.
Collapse
Affiliation(s)
- Wei Ou
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China; Department of Anesthesiology, Nanchong Central Hospital, Nanchong, 637000, China
| | - Yu Liang
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yu Qin
- Department of Anesthesiology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Wei Wu
- Department of Anesthesiology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Maodi Xie
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yabing Zhang
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yarong Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Liwei Ji
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Haiyang Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tao Li
- Laboratory of Mitochondria and Metabolism, Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Anesthesia and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Bekhite M, González-Delgado A, Hübner S, Haxhikadrija P, Kretzschmar T, Müller T, Wu JMF, Bekfani T, Franz M, Wartenberg M, Gräler M, Greber B, Schulze PC. The role of ceramide accumulation in human induced pluripotent stem cell-derived cardiomyocytes on mitochondrial oxidative stress and mitophagy. Free Radic Biol Med 2021; 167:66-80. [PMID: 33705961 DOI: 10.1016/j.freeradbiomed.2021.02.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/19/2021] [Accepted: 02/09/2021] [Indexed: 11/23/2022]
Abstract
Oversupply of fatty acids (FAs) to cardiomyocytes (CMs) is associated with increased ceramide content and elevated the risk of lipotoxic cardiomyopathy. Here we investigate the role of ceramide accumulation on mitochondrial function and mitophagy in cardiac lipotoxicity using CMs derived from human induced pluripotent stem cell (hiPSC). Mature CMs derived from hiPSC exposed to the diabetic-like environment or transfected with plasmids overexpressing serine-palmitoyltransferase long chain base subunit 1 (SPTLC1), a subunit of the serine-palmitoyltransferase (SPT) complex, resulted in increased intracellular ceramide levels. Accumulation of ceramides impaired insulin-dependent phosphorylation of Akt through activating protein phosphatase 2A (PP2A) and disturbed gene and protein levels of key metabolic enzymes including GLUT4, AMPK, PGC-1α, PPARα, CD36, PDK4, and PPARγ compared to controls. Analysis of CMs oxidative metabolism using a Seahorse analyzer showed a significant reduction in ATP synthesis-related O2 consumption, mitochondrial β-oxidation and respiratory capacity, indicating an impaired mitochondrial function under diabetic-like conditions or SPTLC1-overexpression. Further, ceramide accumulation increased mitochondrial fission regulators such as dynamin-related protein 1 (DRP1) and mitochondrial fission factor (MFF) as well as auto/mitophagic proteins LC3B and PINK-1 compared to control. Incubation of CMs with the specific SPT inhibitor (myriocin) showed a significant increase in mitochondrial fusion regulators the mitofusin 2 (MFN2) and optic atrophy 1 (OPA1) as well as p-Akt, PGC-1 α, GLUT-4, and ATP production. In addition, a significant decrease in auto/mitophagy and apoptosis was found in CMs treated with myriocin. Our results suggest that ceramide accumulation has important implications in driving insulin resistance, oxidative stress, increased auto/mitophagy, and mitochondrial dysfunction in the setting of lipotoxic cardiomyopathy. Therefore, modulation of the de novo ceramide synthesis pathway may serve as a novel therapeutic target to treat metabolic cardiomyopathy.
Collapse
Affiliation(s)
- Mohamed Bekhite
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany.
| | - Andres González-Delgado
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Sascha Hübner
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Pëllumb Haxhikadrija
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Tom Kretzschmar
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Tina Müller
- Clinic for Anesthesiology and Intensive Care Medicine, University Hospital Jena, FSU, Jena, Germany
| | - Jasmine M F Wu
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Tarek Bekfani
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Marcus Franz
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Maria Wartenberg
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Markus Gräler
- Clinic for Anesthesiology and Intensive Care Medicine, University Hospital Jena, FSU, Jena, Germany
| | - Boris Greber
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| |
Collapse
|
8
|
Liu XS, Zeng J, Yang YX, Qi CL, Xiong T, Wu GZ, Zeng CY, Wang DX. DRD4 Mitigates Myocardial Ischemia/Reperfusion Injury in Association With PI3K/AKT Mediated Glucose Metabolism. Front Pharmacol 2021; 11:619426. [PMID: 33584304 PMCID: PMC7873565 DOI: 10.3389/fphar.2020.619426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
Ischemia-reperfusion (I/R) could cause heart irreversible damage, which is tightly combined with glucose metabolism disorder. It is demonstrated that GLUT4 (glucose transporter 4) translocation is critical for glucose metabolism in the cardiomyocytes under I/R injury. Moreover, DRD4 (dopamine receptor D4) modulate glucose metabolism, and protect neurocytes from anoxia/reoxygenation (A/R) injury. Thus, DRD4 might regulate myocardial I/R injury in association with GLUT4-mediated glucose metabolism. However, the effects and mechanisms are largely unknown. In the present study, the effect of DRD4 in heart I/R injury were studied ex vivo and in vitro. For I/R injury ex vivo, DRD4 agonist (PD168077) was perfused by Langendorff system in the isolated rat heart. DRD4 activated by PD168077 improved cardiac function in the I/R-injured heart as determined by the left ventricular developed pressure (LVDP), +dp/dt, and left ventricular end diastolic pressure (LVEDP), and reduced heart damage evidenced by infarct size, the release of troponin T (TNT) and lactate dehydrogenase (LDH). DRD4 activation diminished I/R injury induced apoptosis and enhanced cell viability impaired by I/R injury in cardiomyocyte, showed by TUNEL staining, flow cytometer and CCK8 assay. Furthermore, DRD4 activation did not change total GULT4 protein expression level but increased the membrane GULT4 localization determined by western blot. In terms of mechanism, DRD4 activation increased pPI3K/p-AKT but not the total PI3K/AKT during anoxia/reoxygenation (A/R) injury in vitro. Interestingly, PI3K inhibitor, Wortmannin, blocked PI3K/AKT pathway and depleted the membrane GULT4, and further promoted apoptosis showed by TUNEL staining, flow cytometer, western blot of cleaved caspase 3, BAX and BCL2 expression. Thus, DRD4 activation exerted a protective effect against I/R injury by promoting GLUT4 translocation depended on PI3K/AKT pathway, which enhanced the ability of glucose uptake, and ultimately reduced the apoptosis in cardiomyocytes.
Collapse
Affiliation(s)
- Xue-Song Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jing Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Yu-Xue Yang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Chun-Lei Qi
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ting Xiong
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Geng-Ze Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Chun-Yu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Da-Xin Wang
- The Hospital Affiliated to Medical School of Yangzhou University (Taizhou people's Hospital), Taizhou, China
| |
Collapse
|
9
|
Naryzhnaya NV, Ma HJ, Maslov LN. The involvement of protein kinases in the cardioprotective effect of chronic hypoxia. Physiol Res 2020; 69:933-945. [PMID: 33129243 DOI: 10.33549/physiolres.934439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The purpose of this review is to analyze the involvement of protein kinases in the cardioprotective mechanism induced by chronic hypoxia. It has been reported that chronic intermittent hypoxia contributes to increased expression of the following kinases in the myocardium: PKCdelta, PKCalpha, p-PKCepsilon, p-PKCalpha, AMPK, p-AMPK, CaMKII, p-ERK1/2, p-Akt, PI3-kinase, p-p38, HK-1, and HK-2; whereas, chronic normobaric hypoxia promotes increased expression of the following kinases in the myocardium: PKCepsilon, PKCbetaII, PKCeta, CaMKII, p-ERK1/2, p-Akt, p-p38, HK-1, and HK-2. However, CNH does not promote enhanced expression of the AMPK and JNK kinases. Adaptation to hypoxia enhances HK-2 association with mitochondria and causes translocation of PKCdelta, PKCbetaII, and PKCeta to the mitochondria. It has been shown that PKCdelta, PKCepsilon, ERK1/2, and MEK1/2 are involved in the cardioprotective effect of chronic hypoxia. The role of other kinases in the cardioprotective effect of adaptation to hypoxia requires further research.
Collapse
Affiliation(s)
- N V Naryzhnaya
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia.
| | | | | |
Collapse
|
10
|
Hu K, Deng W, Yang J, Wei Y, Wen C, Li X, Chen Q, Ke D, Li G. Intermittent hypoxia reduces infarct size in rats with acute myocardial infarction: a systematic review and meta-analysis. BMC Cardiovasc Disord 2020; 20:422. [PMID: 32962654 PMCID: PMC7507284 DOI: 10.1186/s12872-020-01702-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 09/09/2020] [Indexed: 11/20/2022] Open
Abstract
Background To determine whether intermittent hypoxia (IH) can reduce the infarct size (IS) after acute myocardial infarction (AMI) in rats. Methods Articles were identified in PubMed, EMBASE and the Web of Science and were included if they evaluated the effect of IH on the changes in the infarcted area after AMI in rats. Results A preliminary search identified 3633 articles and 29 data sets from 23 articles (12 in vivo, 16 in vitro). The IS decreased after AMI in IH rats both in vitro (SMD -1.46, 95% CI [− 2.37, − 0.55]; I2 = 85.6%, P = 0.000) and in vivo (SMD -1.43, 95% CI [− 2.05, − 0.82], I2 = 73.6%, P = 0.000). Sensitivity analysis indicated that IH had a strong protective effect against myocardial infarction, and the hypoxia concentration was significantly correlated with the change in IS after AMI. Conclusion IH can reduce IS after AMI in rats. This effect of IH may be related to the dose of hypoxia, and the oxygen concentration may be one of the most important influencing factors.
Collapse
Affiliation(s)
- Ke Hu
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Wei Deng
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Jing Yang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Yu Wei
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Chaolin Wen
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Xingsheng Li
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Qingwei Chen
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Dazhi Ke
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Guiqiong Li
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| |
Collapse
|
11
|
Zhang J, Ren D, Fedorova J, He Z, Li J. SIRT1/SIRT3 Modulates Redox Homeostasis during Ischemia/Reperfusion in the Aging Heart. Antioxidants (Basel) 2020; 9:antiox9090858. [PMID: 32933202 PMCID: PMC7556005 DOI: 10.3390/antiox9090858] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemia/reperfusion (I/R) injury is the central cause of global death in cardiovascular diseases, which is characterized by disorders such as angina, stroke, and peripheral vascular disease, finally causing severe debilitating diseases and death. The increased rates of morbidity and mortality caused by I/R are parallel with aging. Aging-associated cardiac physiological structural and functional deterioration were found to contribute to abnormal reactive oxygen species (ROS) production during I/R stress. Disturbed redox homeostasis could further trigger the related signaling pathways that lead to cardiac irreversible damages with mitochondria dysfunction and cell death. It is notable that sirtuin proteins are impaired in aged hearts and are critical to maintaining redox homeostasis via regulating substrate metabolism and inflammation and thus preserving cardiac function under stress. This review discussed the cellular and functional alterations upon I/R especially in aging hearts. We propose that mitochondria are the primary source of reactive oxygen species (ROS) that contribute to I/R injury in aged hearts. Then, we highlight the cardiomyocyte protection of the age-related proteins Sirtuin1 (SIRT1) and Sirtuin1 (SIRT3) in response to I/R injury, and we discuss their modulation of cardiac metabolism and the inflammatory reaction that is involved in ROS formation.
Collapse
Affiliation(s)
- Jingwen Zhang
- College of Life Sciences, Shandong Normal University, Jinan 250014, China;
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (D.R.); (J.F.); (Z.H.)
| | - Di Ren
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (D.R.); (J.F.); (Z.H.)
| | - Julia Fedorova
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (D.R.); (J.F.); (Z.H.)
| | - Zhibin He
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (D.R.); (J.F.); (Z.H.)
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (D.R.); (J.F.); (Z.H.)
- Correspondence: ; Tel.: +1-813-974-4917
| |
Collapse
|
12
|
Optimizing the Use of iPSC-CMs for Cardiac Regeneration in Animal Models. Animals (Basel) 2020; 10:ani10091561. [PMID: 32887495 PMCID: PMC7552322 DOI: 10.3390/ani10091561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In 2006, the first induced pluripotent stem cells were generated by reprogramming skin cells. Induced pluripotent stem cells undergo fast cell division, can differentiate into many different cell types, can be patient-specific, and do not raise ethical issues. Thus, they offer great promise as in vitro disease models, drug toxicity testing platforms, and for autologous tissue regeneration. Heart failure is one of the major causes of death worldwide. It occurs when the heart cannot meet the body’s metabolic demands. Induced pluripotent stem cells can be differentiated into cardiac myocytes, can form patches resembling native cardiac tissue, and can engraft to the damaged heart. However, despite correct host/graft coupling, most animal studies demonstrate an arrhythmogenicity of the engrafted tissue and variable survival. This is partially because of the heterogeneity and immaturity of the cells. New evidence suggests that by modulating induced pluripotent stem cells-cardiac myocytes (iPSC-CM) metabolism by switching substrates and changing metabolic pathways, you can decrease iPSC-CM heterogeneity and arrhythmogenicity. Novel culture methods and tissue engineering along with animal models of heart failure are needed to fully unlock the potential of cardiac myocytes derived from induced pluripotent stem cells for cardiac regeneration. Abstract Heart failure (HF) is a common disease in which the heart cannot meet the metabolic demands of the body. It mostly occurs in individuals 65 years or older. Cardiac transplantation is the best option for patients with advanced HF. High numbers of patient-specific cardiac myocytes (CMs) can be generated from induced pluripotent stem cells (iPSCs) and can possibly be used to treat HF. While some studies found iPSC-CMS can couple efficiently to the damaged heart and restore cardiac contractility, almost all found iPSC-CM transplantation is arrhythmogenic, thus hampering the use of iPSC-CMs for cardiac regeneration. Studies show that iPSC-CM cultures are highly heterogeneous containing atrial-, ventricular- and nodal-like CMs. Furthermore, they have an immature phenotype, resembling more fetal than adult CMs. There is an urgent need to overcome these issues. To this end, a novel and interesting avenue to increase CM maturation consists of modulating their metabolism. Combined with careful engineering and animal models of HF, iPSC-CMs can be assessed for their potential for cardiac regeneration and a cure for HF.
Collapse
|
13
|
Cheng W, Wang L, Yang T, Wu A, Wang B, Li T, Lu Z, Yang J, Li Y, Jiang Y, Wu X, Meng H, Zhao M. Qiliqiangxin Capsules Optimize Cardiac Metabolism Flexibility in Rats With Heart Failure After Myocardial Infarction. Front Physiol 2020; 11:805. [PMID: 32848816 PMCID: PMC7396640 DOI: 10.3389/fphys.2020.00805] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 06/17/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic modulation is a promising therapy for ischemic heart disease and heart failure. This study aimed to clarify the regional modulatory effect of Qiliqiangxin capsules (QLQX), a traditional Chinese medicine, on cardiac metabolic phenotypes. Sprague-Dawley rats underwent left anterior descending coronary artery ligation and were treated with QLQX and enalapril. Striking global left ventricular dysfunction and left ventricular remodeling were significantly improved by QLQX. In addition to the posterior wall, QLQX also had a unique beneficial effect on the anterior wall subject to a severe oxygen deficit. Cardiac tissues in the border and remote areas were separated for detection. QLQX enhanced the cardiac 18F-fluorodeoxyglucose uptake and the levels and translocation of glucose transport 4 (GLUT4) in the border area. Meanwhile, it also suppressed glucose transport 1 (GLUT1) in both areas, indicating that QLQX encouraged border myocytes to use more glucose in a GLUT4-dependent manner. It was inferred that QLQX promoted a shift from glucose oxidation to anaerobic glycolysis in the border area by the augmentation of phosphorylated pyruvate dehydrogenase, pyruvate dehydrogenase kinases 4, and lactic dehydrogenase A. QLQX also upregulated the protein expression of fatty acid translocase and carnitine palmitoyl transferase-1 in the remote area to possibly normalize fatty acid (FA) uptake and oxidation similar to that in healthy hearts. QLQX protected global viable cardiomyocytes and promoted metabolic flexibility by modulating metabolic proteins regionally, indicating its potential for driving the border myocardium into an anaerobic glycolytic pathway against hypoxia injuries and urging the remote myocardium to oxidize FA to maximize energy production.
Collapse
Affiliation(s)
- Wenkun Cheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Yang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aiming Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tong Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingjing Yang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yangyang Jiang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxiao Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Meng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Sun H, Wang J, Que J, Peng Y, Yu Y, Wang L, Ye H, Huang K, Xue Y, Zhou Y, Ji K. RNA sequencing revealing the role of AMP-activated protein kinase signaling in mice myocardial ischemia reperfusion injury. Gene 2019; 703:91-101. [PMID: 30974198 DOI: 10.1016/j.gene.2019.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/23/2019] [Accepted: 04/02/2019] [Indexed: 12/26/2022]
Abstract
Long non-coding RNAs (lncRNA) and circular RNAs (circRNA) that sponge miRNAs could indirectly regulate gene expression, contributing to certain biological processes. This study aimed to investigate the role of non-coding RNAs in the pathogenesis of myocardial ischemia reperfusion-injury (MIRI). MIRI in male C57B/6J mice was induced by left anterior descending coronary artery ligation occlusion for 30 min, and 4 h of reperfusion. RNA sequencing was performed to obtain the mRNA and non-coding RNA expression profiles of the MIRI and sham groups. Bioinformatic methods were used to analyze the co-expression RNAs, miRNA binding sites and competitive endogenous RNA (ceRNA) pairs. Differentially expressed RNAs were identified with a cutoff fold change > 2 and p < 0.05. A total of 64 mRNAs were upregulated and 98 mRNAs were downregulated, and 10 lncRNAs were upregulated and 10 lncRNAs were downregulated. All altered (p < 0.05) mRNAs were selected for gene ontology and pathway analysis. The AMP-activated protein kinase (AMPK) signaling pathway was enriched in the downregulated genes, and the activation of AMPK was confirmed by western blotting. The lncRNA co-expression network and ceRNA network base on genes in AMPK signaling pathway were then constructed, revealing that ENSMUST00000147762.7 and TUCP_000184 might be key regulators in MIRI induced AMPK activation. The expression levels of AMPK signaling-related RNAs and those involved in the ceRNA network were validated using qRT-PCR. Overall, this study identified potential new targets on AMPK signaling in MIRI.
Collapse
Affiliation(s)
- Huankun Sun
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Jiaoni Wang
- Department of Diagnostic Ultrasound and Echocardiography, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Jiaqun Que
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yangpei Peng
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yongwei Yu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Lei Wang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Haihao Ye
- Department of Cardiology, Wenzhou Hospital of Traditional Chinese Medicine, Wenzhou 325000, Zhejiang, China
| | - Kaiyu Huang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yangjing Xue
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Yingying Zhou
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| | - Kangting Ji
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| |
Collapse
|
15
|
Cheng WJ, Liu X, Zhang L, Guo XQ, Wang FW, Zhang Y, Tian YM. Chronic intermittent hypobaric hypoxia attenuates skeletal muscle ischemia-reperfusion injury in mice. Life Sci 2019; 231:116533. [PMID: 31173783 DOI: 10.1016/j.lfs.2019.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/25/2019] [Accepted: 06/03/2019] [Indexed: 10/26/2022]
Abstract
AIM The aim of this study was to investigate the protective effect of chronic intermittent hypobaric hypoxia (CIHH) against skeletal muscle ischemia-reperfusion (IR) injury and to determine the underlying mechanism. MAIN METHODS C57BL/6 mice were randomly divided into 3 groups: skeletal muscle IR injury group (IR), CIHH pretreatment following IR group (IR + CIHH), and sham operation group (Sham). The skeletal muscle IR injury model was induced by the unilateral application of a tourniquet on a hind limb for 3 h and then releasing it for 24 h. CIHH pretreatment simulating a 5000-m altitude was applied 6 h per day for 28 days. The functional and morphological performance of IR-injured gastrocnemius muscle was evaluated using contraction force, H&E staining, and transmission electron microscopy. IR injury-induced CD68+ macrophage infiltration was assessed by immunofluorescence. TNFα levels in serum and muscle were measured by ELISA and western blotting, respectively. Apoptosis was examined by TUNEL staining and Cleaved Caspase-3 protein expression. KEY FINDINGS Acute IR injury resulted in reduced contraction tension, morphological destruction, macrophage infiltration, increased TNFα levels, and apoptosis in gastrocnemius muscle. CIHH pretreatment significantly ameliorated contraction function and morphological performance in IR-injured skeletal muscle. In addition, CIHH pretreatment resulted in marked decreases in CD68+ macrophage infiltration, TNFα levels, and apoptosis. SIGNIFICANCE These data demonstrated that CIHH has a protective effect against acute IR injury in skeletal muscle via inhibition of inflammation and apoptosis.
Collapse
Affiliation(s)
- Wen-Jie Cheng
- Department of Anesthesiology, Tianjin Hospital, Tianjin 300000, China; Graduate school, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Xin Liu
- Department of Neurology, Second Hospital of Xi'an Medical University, Xi'an, Shanxi 710038, China
| | - Li Zhang
- Department of Orthopedics, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Xin-Qi Guo
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Fu-Wei Wang
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Yi Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Yan-Ming Tian
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei 050017, China; Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China.
| |
Collapse
|
16
|
Protection of Myocardial Ischemia-Reperfusion by Therapeutic Hypercapnia: a Mechanism Involving Improvements in Mitochondrial Biogenesis and Function. J Cardiovasc Transl Res 2019; 12:467-477. [PMID: 30980235 DOI: 10.1007/s12265-019-09886-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
Previous studies proposed that acidic reperfusion may be a protective strategy for myocardial ischemia-reperfusion therapy with potential of clinical transformation. In this study, we investigated whether therapeutic hypercapnia could mimic acidosis postconditioning in isolated hearts with a 30-min left coronary artery ligation-reperfusion model in rats. Therapeutic hypercapnia (inhalation 20% CO2 for 10 min) is cardioprotective with a strict therapeutic time window and acidity: it reduced the infarct ratio and serum myocardial enzyme and increased the myocardial ATP content. Furthermore, mitochondrial morphology damage, the loss of mitochondrial membrane potential, and the formation of mitochondrial permeability transition pore were effectively inhibited, indicating the improvements in mitochondrial function. The expression of the mitochondrial biogenesis regulators was upregulated simultaneously. These findings indicated therapeutic hypercapnia in animals can mimic ex vivo acidosis postconditioning to alleviate myocardial ischemia-reperfusion injury. The effect is related to improvement in mitochondrial function and regulation of the mitochondrial biogenesis pathway.
Collapse
|
17
|
Tao L, Wang L, Yang X, Jiang X, Hua F. Recombinant human glucagon-like peptide-1 protects against chronic intermittent hypoxia by improving myocardial energy metabolism and mitochondrial biogenesis. Mol Cell Endocrinol 2019; 481:95-103. [PMID: 30503377 DOI: 10.1016/j.mce.2018.11.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Obstructive sleep apnea syndrome is a chronic disease associated with intermittent hypoxia (IH) and is an important risk factor for cardiovascular disease. Glucagon-like peptide (GLP-1) is a naturally occurring incretin used as a promising therapeutic agent in the treatment of acute myocardial infarction, dilated cardiomyopathy, and advanced heart failure. However, whether GLP-1 can protect against IH-induced cardiac injury is still unclear. Accordingly, in this study, we evaluated the effects of recombinant human GLP-1 (rhGLP-1) on cardiac health in mice. METHODS Mice were subjected to repetitive 5% O2 for 30 s and 21% O2 for 30 s, for a total of 8 h/day for 4 weeks. Subsequently, mice received subcutaneous injection of saline or rhGLP-1 (100 μg/kg, three times per day). Cardiac function, myocardial apoptosis and fibrosis, energy metabolism, and mitochondrial biogenesis were examined for evaluation of cardiac injury. RESULTS A reduction in diastolic function (E/A ratio) in mice exposed to IH was significantly reversed by rhGLP-1. IH induced marked cardiomyocyte apoptosis and myocardial fibrosis. Additionally, IH resulted in a shift from fatty acid to glucose metabolism in the myocardium with downregulation of peroxisome proliferator-activated receptor (PPAR) α and PPARγ. Moreover, IH caused a reduction in mitochondrial DNA (mtDNA) replication and transcription, together with reduced mtDNA content and impaired mitochondrial ultrastructure. These changes were abolished by rhGLP-1 via activation of PGC-1α and Akt signaling. CONCLUSIONS rhGLP-1 protects against IH-induced cardiac injury by improving myocardial energy metabolism and enhancing the early adaptive changes of mitochondrial biogenesis.
Collapse
Affiliation(s)
- Lichan Tao
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Long Wang
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Xiaoyu Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Xiaohong Jiang
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Fei Hua
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China.
| |
Collapse
|
18
|
Klos M, Morgenstern S, Hicks K, Suresh S, Devaney EJ. The effects of the ketone body β-hydroxybutyrate on isolated rat ventricular myocyte excitation-contraction coupling. Arch Biochem Biophys 2018; 662:143-150. [PMID: 30543786 DOI: 10.1016/j.abb.2018.11.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/06/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023]
Abstract
β-hydroxybutyrate is the primary ketone body produced by the body during ketosis and is used to meet its metabolic demands. The healthy adult heart derives most of its energy from fatty acid oxidation. However, in certain diseases, the heart alters its substrate preference and increases its ketone body metabolism. Little is known about the effects of βOHB on ventricular myocyte excitation-contraction coupling. Therefore, we examined the effects of ketone body metabolism on single cell excitation-contraction coupling during normoxic and hypoxic conditions. Myocytes were isolated from adult rats, cultured for 18 h in RPMI 1640, RPMI 1640 no glucose, and M199, HEPES with/without various amount of βOHB added. To simulate hypoxia, myocytes were incubated at 1%O2, 5% CO2 for 1 h followed by incubation at atmospheric oxygen (21%O2,5% CO2) for 30 min before recordings. Recordings were obtained using an IonOptix system at 36±1ᵒ C. Myocytes were paced at 0.5, 1, 2, 3, and 4 Hz. We found that exposure to βOHB had no effect on excitation-contraction coupling. However, culturing cells with βOHB results in a significant increase in both contraction and calcium in RPMI 1640 media. Dose response experiments demonstrated 0.5 mM βOHB is enough to increase myocyte contraction in the absence of glucose. However, βOHB has no measurable effects on myocytes cultured in a nutrient rich media, M199, HEPES. Therefore, βOHB improves single cell excitation-contraction coupling, is protective against hypoxia, and may be a beneficial adaptation for the heart during periods of nutrient scarcity and or metabolic dysregulation.
Collapse
Affiliation(s)
- Matthew Klos
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH, 44106, USA; Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Sherry Morgenstern
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH, 44106, USA
| | - Kayla Hicks
- Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Shreyas Suresh
- Case Western Reserve University School of Graduate Studies, Cleveland, OH, 44106, USA
| | - Eric J Devaney
- Department of Pediatric Cardiac Surgery, UH Hospitals Cleveland, Cleveland, OH, 44106, USA; Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
19
|
Siques P, Brito J, Flores K, Ordenes S, Arriaza K, Pena E, León-Velarde F, López de Pablo ÁL, Gonzalez MC, Arribas S. Long-Term Chronic Intermittent Hypobaric Hypoxia Induces Glucose Transporter (GLUT4) Translocation Through AMP-Activated Protein Kinase (AMPK) in the Soleus Muscle in Lean Rats. Front Physiol 2018; 9:799. [PMID: 30002630 PMCID: PMC6031730 DOI: 10.3389/fphys.2018.00799] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/07/2018] [Indexed: 11/20/2022] Open
Abstract
Background: In chronic hypoxia (CH) and short-term chronic intermittent hypoxia (CIH) exposure, glycemia and insulin levels decrease and insulin sensitivity increases, which can be explained by changes in glucose transport at skeletal muscles involving GLUT1, GLUT4, Akt, and AMPK, as well as GLUT4 translocation to cell membranes. However, during long-term CIH, there is no information regarding whether these changes occur similarly or differently than in other types of hypoxia exposure. This study evaluated the levels of AMPK and Akt and the location of GLUT4 in the soleus muscles of lean rats exposed to long-term CIH, CH, and normoxia (NX) and compared the findings. Methods: Thirty male adult rats were randomly assigned to three groups: a NX (760 Torr) group (n = 10), a CIH group (2 days hypoxia/2 days NX; n = 10) and a CH group (n = 10). Rats were exposed to hypoxia for 30 days in a hypobaric chamber set at 428 Torr (4,600 m). Feeding (10 g daily) and fasting times were accurately controlled. Measurements included food intake (every 4 days), weight, hematocrit, hemoglobin, glycemia, serum insulin (by ELISA), and insulin sensitivity at days 0 and 30. GLUT1, GLUT4, AMPK levels and Akt activation in rat soleus muscles were determined by western blot. GLUT4 translocation was measured with confocal microscopy at day 30. Results: (1) Weight loss and increases in hematocrit and hemoglobin were found in both hypoxic groups (p < 0.05). (2) A moderate decrease in glycemia and plasma insulin was found. (3) Insulin sensitivity was greater in the CIH group (p < 0.05). (4) There were no changes in GLUT1, GLUT4 levels or in Akt activation. (5) The level of activated AMPK was increased only in the CIH group (p < 0.05). (6) Increased GLUT4 translocation to the plasma membrane of soleus muscle cells was observed in the CIH group (p < 0.05). Conclusion: In lean rats experiencing long-term CIH, glycemia and insulin levels decrease and insulin sensitivity increases. Interestingly, there is no increase of GLUT1 or GLUT4 levels or in Akt activation. Therefore, cellular regulation of glucose seems to primarily involve GLUT4 translocation to the cell membrane in response to hypoxia-mediated AMPK activation.
Collapse
Affiliation(s)
- Patricia Siques
- Institute of Health Studies, University Arturo Prat, Iquique, Chile
| | - Julio Brito
- Institute of Health Studies, University Arturo Prat, Iquique, Chile
| | - Karen Flores
- Institute of Health Studies, University Arturo Prat, Iquique, Chile
| | - Stefany Ordenes
- Institute of Health Studies, University Arturo Prat, Iquique, Chile
| | - Karem Arriaza
- Institute of Health Studies, University Arturo Prat, Iquique, Chile
| | - Eduardo Pena
- Institute of Health Studies, University Arturo Prat, Iquique, Chile
| | - Fabiola León-Velarde
- Department of Biological and Physiological Sciences, Facultad de Ciencias y Filosofía/IIA, Cayetano Heredia University, Lima, Peru
| | - Ángel L López de Pablo
- Department of Physiology, Faculty of Medicine, University Autonoma of Madrid, Madrid, Spain
| | - M C Gonzalez
- Department of Physiology, Faculty of Medicine, University Autonoma of Madrid, Madrid, Spain
| | - Silvia Arribas
- Department of Physiology, Faculty of Medicine, University Autonoma of Madrid, Madrid, Spain
| |
Collapse
|
20
|
Gouw AM, Efe G, Barakat R, Preecha A, Mehdizadeh M, Garan SA, Brooks GA. Roles of estrogen receptor-alpha in mediating life span: the hypothalamic deregulation hypothesis. Physiol Genomics 2016; 49:88-95. [PMID: 28011880 DOI: 10.1152/physiolgenomics.00073.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In several species caloric restriction (CR) extends life span. In this paper we integrate data from studies on CR and other sources to articulate the hypothalamic deregulation hypothesis by which estrogen receptor-alpha (ER-α) signaling in the hypothalamus and limbic system affects life span under the stress of CR in mammals. ER-α is one of two principal estrogen-binding receptors differentially expressed in the amygdala, hippocampus, and several key hypothalamic nuclei: the arcuate nucleus (ARN), preoptic area (POA), ventromedial nucleus (VMN), antero ventral periventricular nucleus (AVPV), paraventricular nucleus (PVN), supraoptic nucleus (SON), and suprachiasmatic nucleus (SCN). Estradiol signaling via ER-α is essential in basal level functioning of reproductive cycle, sexually receptive behaviors, physiological stress responses, as well as sleep cycle, and other nonsexual behaviors. When an organism is placed under long-term CR, which introduces an external stress to this ER-α signaling, the reduction of ER-α expression is attenuated over time in the hypothalamus. This review paper seeks to characterize the downstream effects of ER-α in the hypothalamus and limbic system that affect normal endocrine functioning.
Collapse
Affiliation(s)
- Arvin M Gouw
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and.,Department of Integrative Biology, University of California at Berkeley, Berkeley, California
| | - Gizem Efe
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - Rita Barakat
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - Andrew Preecha
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - Morvarid Mehdizadeh
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - Steven A Garan
- Lawrence Berkeley National Laboratories, Berkeley, California.,Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and
| | - George A Brooks
- Center for Research and Education in Aging, University of California at Berkeley, Lawrence Berkeley National Laboratories, California; and .,Department of Integrative Biology, University of California at Berkeley, Berkeley, California
| |
Collapse
|
21
|
He S, Liu S, Wu X, Xin M, Ding S, Xin D, Ouyang H, Zhang J. Protective role of downregulated MLK3 in myocardial adaptation to chronic hypoxia. J Physiol Biochem 2016; 73:371-380. [PMID: 28555332 DOI: 10.1007/s13105-017-0561-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/28/2017] [Indexed: 11/25/2022]
Abstract
A series of protective responses could be evoked to achieve compensatory adaptation once cardiomyocytes are subjected to chronic hypoxia. MLK3/JNK/c-jun signaling pathway was previously demonstrated to be involved in this process. In the present study, we aim to further examine the performance of MLK3 in hypoxic H9C2 cells and potential mechanism. Myocardial samples of patients with congenital heart disease (CHD) were collected. H9C2 cells were cultured in hypoxic conditions for various durations. MLK3 was silenced by transfection of shRNA to evaluate its role in cell viability. We found expression of MLK3 protein was lower in patients with cyanotic CHD. In hypoxic H9C2 cells, its expression was gradually decreased in a time-dependent manner. However, there was no significant difference about expression of MLK3 mRNA. According to the results of MTT, LDH, and TUNEL, faster cell growth curve, lower death rate, and less apoptotic cells could be observed in MLK-shRNA group compared with scramble-shRNA group. Silencing of MLK3 significantly reduced expression of cleaved caspase-3, cleaved PARP, Bad, and Bax, together with increased expression of Bcl-2 and ration of Bcl-2/Bax. Both ratio of phospho-JNK/total JNK and ratio of phospho-c-jun/total c-jun were significantly decreased once MLK3 was silenced. At various reoxygenation time, MLK3 shRNA could significantly promote cell survival and decrease cell death according to MTT and LDH. Our results suggested that chronic hypoxia could reduce MLK3 expression in a posttranscriptional regulatory manner. Downregulation of MLK3 protects H9C2 cells from hypoxia-induced apoptosis and H/R injury via blocking the activation of JNK and c-jun.
Collapse
Affiliation(s)
- Siyi He
- Department of Cardiovascular Surgery, Chengdu Military General Hospital, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province, 610083, China
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shunbi Liu
- Department of Cardiovascular Surgery, Chengdu Military General Hospital, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province, 610083, China
| | - Xiaochen Wu
- Department of Cardiovascular Surgery, Chengdu Military General Hospital, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province, 610083, China
| | - Mei Xin
- Department of Cardiovascular Surgery, Chengdu Military General Hospital, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province, 610083, China
| | - Sheng Ding
- Department of Cardiovascular Surgery, Chengdu Military General Hospital, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province, 610083, China
| | - Dong Xin
- Department of Cardiovascular Surgery, Chengdu Military General Hospital, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province, 610083, China
| | - Hui Ouyang
- Department of Cardiovascular Surgery, Chengdu Military General Hospital, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province, 610083, China.
| | - Jinbao Zhang
- Department of Cardiovascular Surgery, Chengdu Military General Hospital, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province, 610083, China.
| |
Collapse
|