1
|
Morii A, Matsuo I, Suita K, Ohnuki Y, Ishikawa M, Ito A, Miyamoto G, Abe M, Mitsubayashi T, Mototani Y, Nariyama M, Matsubara R, Hayakawa Y, Amitani Y, Gomi K, Nagano T, Okumura S. Allopurinol attenuates development of Porphyromonas gingivalis LPS-induced cardiomyopathy in mice. PLoS One 2025; 20:e0318008. [PMID: 40179080 PMCID: PMC11967946 DOI: 10.1371/journal.pone.0318008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 01/08/2025] [Indexed: 04/05/2025] Open
Abstract
Oxidative stress is involved in the progression of periodontitis, independently of confounding factors such as smoking, and numerous studies suggest that periodontitis is associated with increased risk of cardiovascular disease. In this study, therefore, we examined the effects of the xanthine oxidase inhibitor allopurinol on cardiac dysfunction in mice treated with Porphyromonas gingivalis lipopolysaccharide (PG-LPS) at a dose (0.8 mg/kg/day) equivalent to the circulating level in patients with periodontal disease. Mice were divided into four groups: 1) control, 2) PG-LPS, 3) allopurinol, and 4) PG-LPS + allopurinol. After1 week, we evaluated cardiac function by echocardiography. The left ventricular ejection fraction was significantly decreased in PG-LPS-treated mice compared to the control (from 68 ± 1.3 to 60 ± 2.7%), while allopurinol ameliorated the dysfunction (67 ± 1.1%). The area of cardiac fibrosis was significantly increased (approximately 3.6-fold) and the number of apoptotic myocytes was significantly increased (approximately 7.7-fold) in the heart of the PG-LPS-treated group versus the control, and these changes were suppressed by allopurinol. The impairment of cardiac function in PG-LPS-treated mice was associated with increased production of reactive oxygen species by xanthine oxidase and NADPH oxidase 4, leading to calmodulin kinase II activation with increased ryanodine receptor 2 phosphorylation. These changes were also suppressed by allopurinol. Our results suggest that oxidative stress plays an important role in the PG-LPS-promoted development of cardiac diseases, and further indicate that allopurinol ameliorates Porphyromonas gingivalis LPS-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ichiro Matsuo
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Oral and Maxillofacial Surgery, Ibaraki Medical Center Tokyo Medical University, Ibaraki, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Go Miyamoto
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Mariko Abe
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Takao Mitsubayashi
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ren Matsubara
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasuharu Amitani
- Department of Mathematics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Takatoshi Nagano
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
2
|
Chen Y, Luo W, Wu Y. Protective effect of thymoquinone against doxorubicin-induced cardiotoxicity and the underlying mechanism. Toxicol Appl Pharmacol 2025; 495:117179. [PMID: 39645202 DOI: 10.1016/j.taap.2024.117179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/06/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Ferroptosis is a key process in doxorubicin (DOX)-induced cardiotoxicity and is a potentially important therapeutic target. Thymoquinone (TQ) is a monoterpenoid compound isolated from black cumin extract that exhibits antitumor effects and acts as a powerful mitochondrial-targeted antioxidant. In this study, we investigated the effect of TQ on DOX-induced cardiotoxicity and the potential underlying mechanisms. METHODS AND RESULTS Mice were randomly assigned to the control (CON) group, DOX (20 mg/kg) group, TQ10 (10 mg/kg/d) group, and TQ20 (20 mg/kg/d) group and intraperitoneally injected with DOX and different doses of TQ. The electrocardiogram, blood pressure, and cardiac ultrasound changes during the experiments showed that TQ exerted a protective effect against DOX-induced cardiotoxicity. The glutathione (GSH), malondialdehyde (MDA), and total antioxidant capacity (T-AOC) levels in the mouse heart tissue were significantly different from those in the CON group. Western blot analysis revealed that the expression of nuclear factor E2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), glutathione peroxidase 4 (GPX4), and ferritin heavy chain 1 (FTH1) in the DOX group was lower than that in the control group. TQ treatment decreased these changes, indicating that TQ alleviated DOX-induced cardiotoxicity and increased the antioxidant capacity of murine cardiomyocytes. The mechanism might involve activating the Nrf2/HO-1 signaling pathway and reducing iron-mediated death. Immunohistochemical staining revealed similar effects on the expression levels of NQO1, COX-2, and NOX4. Moreover, transmission electron microscopy indicated that TQ protected murine cardiomyocytes against DOX-induced mitochondrial damage. CONCLUSION The results of this study suggested that TQ can decrease oxidative stress levels and DOX-induced cardiotoxicity by activating the Nrf2/HO-1 signaling pathway to alleviate ferroptosis in murine cardiomyocytes.
Collapse
Affiliation(s)
- Yi Chen
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang of Jiangxi, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Wei Luo
- Department of Cardiology, The First People's Hospital of Nankang District, Ganzhou of Jiangxi, Xinkang East Avenue, Dongshan Street Office, Ganzhou, Jiangxi 341000, China.
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang of Jiangxi, No. 1 Minde Road, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
3
|
Piristine HC, May HI, Jiang N, Daou D, Olivares-Silva F, Elnwasany A, Szweda P, Szweda L, Kinter C, Kinter M, Sharma G, Wen X, Malloy CR, Jessen ME, Gillette TG, Hill JA. Afterload-induced Decreases in Fatty Acid Oxidation Develop Independently of Increased Glucose Utilization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613531. [PMID: 39345412 PMCID: PMC11429894 DOI: 10.1101/2024.09.17.613531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background Metabolic substrate utilization in HFpEF (heart failure with preserved ejection fraction), the leading cause of heart failure worldwide, is pivotal to syndrome pathogenesis and yet remains ill defined. Under resting conditions, oxidation of free fatty acids (FFA) is the predominant energy source of the heart, supporting its unremitting contractile activity. In the context of disease-related stress, however, a shift toward greater reliance on glucose occurs. In the setting of obesity or diabetes, major contributors to HFpEF pathophysiology, the shift in metabolic substrate use toward glucose is impaired, sometimes attributed to the lower oxygen requirement of glucose oxidation versus fat metabolism. This notion, however, has never been tested conclusively. Furthermore, whereas oxygen demand increases in the setting of increased afterload, myocardial oxygen availability remains adequate for fatty acid oxidation (FAO). Therefore, a "preference" for glucose has been proposed. Methods and Results Pyruvate dehydrogenase complex (PDC) is the rate-limiting enzyme linking glycolysis to the TCA cycle. As PDK4 (PDC kinase 4) is up-regulated in HFpEF, we over-expressed PDK4 in cardiomyocytes, ensuring that PDC is phosphorylated and thereby inhibited. This leads to diminished use of pyruvate as energy substrate, mimicking the decline in glucose oxidation in HFpEF. Importantly, distinct from HFpEF-associated obesity, this model positioned us to abrogate the load-induced shift to glucose utilization in the absence of systemic high fat conditions. As expected, PDK4 transgenic mice manifested normal cardiac performance at baseline. However, they manifested a rapid and severe decline in contractile performance when challenged with modest increases in afterload triggered either by L-NAME or surgical transverse aortic constriction (TAC). This decline in function was not accompanied by an exacerbation of the myocardial hypertrophic growth response. Surprisingly, metabolic flux analysis revealed that, after TAC, fractional FAO decreased, even when glucose/pyruvate utilization was clamped at very low levels. Additionally, proteins involved in the transport and oxidation of FFA were paradoxically downregulated after TAC regardless of genotype. Conclusions These data demonstrate that cardiomyocytes in a setting in which glucose utilization is robustly diminished and prevented from increasing do not compensate for the deficit in glucose utilization by up-regulating FFA use.
Collapse
|
4
|
Clouthier KL, Taylor AC, Xuhuai J, Liu Y, Parker S, Van Eyk J, Reddy S. A Noninvasive Circulating Signature of Combined Right Ventricular Pressure and Volume Overload in Tetralogy of Fallot/Pulmonary Atresia/Major Aortopulmonary Collateral Arteries. World J Pediatr Congenit Heart Surg 2024; 15:162-173. [PMID: 38128927 PMCID: PMC11991743 DOI: 10.1177/21501351231213626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Background: Despite surgical advances, children with tetralogy of Fallot/pulmonary atresia/major aortopulmonary collaterals (TOF/PA/MAPCAs) are subject to chronic right ventricular (RV) pressure and volume overload. Current diagnostic tools do not identify adverse myocardial remodeling and cannot predict progression to RV failure. We sought to identify a noninvasive, circulating signature of the systemic response to right heart stress to follow disease progression. Methods: Longitudinal data were collected from patients with TOF/PA/MAPCAs (N = 5) at the time of (1) early RV pressure overload and (2) late RV pressure and volume overload. Plasma protein and microRNA expression were evaluated using high-throughput data-independent mass spectroscopy and Agilent miR Microarray, respectively. Results: At the time of early RV pressure overload, median patient age was 0.34 years (0.02-9.37), with systemic RV pressures, moderate-severe hypertrophy, and preserved systolic function. Late RV pressure and volume overload occurred at a median age of 4.08 years (1.51-10.83), with moderate RV hypertrophy and dilation, and low normal RV function; 277 proteins were significantly dysregulated (log2FC ≥0.6/≤-0.6, FDR≤0.05), predicting downregulation in lipid transport (apolipoproteins), fibrinolytic system, and extracellular matrix structural proteins (talin 1, profilin 1); and upregulation in the respiratory burst. Increasing RV size and decreasing RV function correlated with decreasing structural protein expression. Similarly, miR expression predicted downregulation of extracellular matrix-receptor interactions and upregulation in collagen synthesis. Conclusion: To our knowledge, we show for the first time a noninvasive protein and miR signature reflecting the systemic response to adverse RV myocardial remodeling in TOF/PA/MAPCAs which could be used to follow disease progression.
Collapse
Affiliation(s)
- Katie L. Clouthier
- Department of Pediatrics (Cardiology), Stanford University, Palo Alto, CA, USA
| | - Anne C. Taylor
- Department of Pediatrics (Cardiology), Stanford University, Palo Alto, CA, USA
| | - Ji Xuhuai
- Human Immune Monitoring Center and Functional Genomics Facility, Stanford University, Palo Alto, CA, USA
| | - Yuhan Liu
- Department of Medicine (Quantitative Science Unit), Stanford University, Palo Alto, CA, USA
| | - Sarah Parker
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer Van Eyk
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sushma Reddy
- Department of Pediatrics (Cardiology), Stanford University, Palo Alto, CA, USA
- Cardiovascular Institute, Stanford University, Los Angeles, CA, USA
| |
Collapse
|
5
|
Labbé P, Martel C, Shi YF, Montezano A, He Y, Gillis MA, Higgins MÈ, Villeneuve L, Touyz R, Tardif JC, Thorin-Trescases N, Thorin E. Knockdown of ANGPTL2 promotes left ventricular systolic dysfunction by upregulation of NOX4 in mice. Front Physiol 2024; 15:1320065. [PMID: 38426206 PMCID: PMC10902461 DOI: 10.3389/fphys.2024.1320065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Background: Angiopoietin-like 2 (ANGPTL2) is a pro-inflammatory and pro-oxidant circulating protein that predicts and promotes chronic inflammatory diseases such as atherosclerosis in humans. Transgenic murine models demonstrated the deleterious role of ANGPTL2 in vascular diseases, while deletion of ANGPTL2 was protective. The nature of its role in cardiac tissues is, however, less clear. Indeed, in adult mice knocked down (KD) for ANGPTL2, we recently reported a mild left ventricular (LV) dysfunction originating from a congenital aortic valve stenosis, demonstrating that ANGPTL2 is essential to cardiac development and function. Hypothesis: Because we originally demonstrated that the KD of ANGPTL2 protected vascular endothelial function via an upregulation of arterial NOX4, promoting the beneficial production of dilatory H2O2, we tested the hypothesis that increased cardiac NOX4 could negatively affect cardiac redox and remodeling and contribute to LV dysfunction observed in adult Angptl2-KD mice. Methods and results: Cardiac expression and activity of NOX4 were higher in KD mice, promoting higher levels of cardiac H2O2 when compared to wild-type (WT) mice. Immunofluorescence showed that ANGPTL2 and NOX4 were co-expressed in cardiac cells from WT mice and both proteins co-immunoprecipitated in HEK293 cells, suggesting that ANGPTL2 and NOX4 physically interact. Pressure overload induced by transverse aortic constriction surgery (TAC) promoted LV systolic dysfunction in WT mice but did not further exacerbate the dysfunction in KD mice. Importantly, the severity of LV systolic dysfunction in KD mice (TAC and control SHAM) correlated with cardiac Nox4 expression. Injection of an adeno-associated virus (AAV9) delivering shRNA targeting cardiac Nox4 expression fully reversed LV systolic dysfunction in KD-SHAM mice, demonstrating the causal role of NOX4 in cardiac dysfunction in KD mice. Targeting cardiac Nox4 expression in KD mice also induced an antioxidant response characterized by increased expression of NRF2/KEAP1 and catalase. Conclusion: Together, these data reveal that the absence of ANGPTL2 induces an upregulation of cardiac NOX4 that contributes to oxidative stress and LV dysfunction. By interacting and repressing cardiac NOX4, ANGPTL2 could play a new beneficial role in the maintenance of cardiac redox homeostasis and function.
Collapse
Affiliation(s)
- Pauline Labbé
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Cécile Martel
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Yan-Fen Shi
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
| | - Augusto Montezano
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Ying He
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | | | | | | | - Rhian Touyz
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | | | - Eric Thorin
- Montreal Heart Institute, Research Center, Montreal, QC, Canada
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
6
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
7
|
Kiyomoto K, Matsuo I, Suita K, Ohnuki Y, Ishikawa M, Ito A, Mototani Y, Tsunoda M, Morii A, Nariyama M, Hayakawa Y, Amitani Y, Gomi K, Okumura S. Oral angiotensin-converting enzyme inhibitor captopril protects the heart from Porphyromonas gingivalis LPS-induced cardiac dysfunction in mice. PLoS One 2023; 18:e0292624. [PMID: 37983238 PMCID: PMC10659197 DOI: 10.1371/journal.pone.0292624] [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: 01/30/2023] [Accepted: 09/25/2023] [Indexed: 11/22/2023] Open
Abstract
Although angiotensin converting enzyme (ACE) inhibitors are considered useful for the treatment of human heart failure, some experimental failing-heart models have shown little beneficial effect of ACE inhibitors in animals with poor oral health, particularly periodontitis. In this study, we examined the effects of the ACE inhibitor captopril (Cap; 0.1 mg/mL in drinking water) on cardiac dysfunction in mice treated with Porphyromonas gingivalis lipopolysaccharide (PG-LPS) at a dose (0.8 mg/kg/day) equivalent to the circulating level in patients with periodontal disease. Mice were divided into four groups: 1) Control, 2) PG-LPS, 3) Cap, and 4) PG-LPS + Cap. After1 week, we evaluated cardiac function by echocardiography. The left ventricular ejection fraction was significantly decreased in PG-LPS-treated mice compared to the control (from 66 ± 1.8 to 59 ± 2.5%), while Cap ameliorated the dysfunction (63 ± 1.1%). The area of cardiac fibrosis was significantly increased (approximately 2.9-fold) and the number of apoptotic myocytes was significantly increased (approximately 5.6-fold) in the heart of PG-LPS-treated group versus the control, and these changes were suppressed by Cap. The impairment of cardiac function in PG-LPS-treated mice was associated with protein kinase C δ phosphorylation (Tyr-311), leading to upregulation of NADPH oxidase 4 and xanthine oxidase, and calmodulin kinase II phosphorylation (Thr-286) with increased phospholamban phosphorylation (Thr-17). These changes were also suppressed by Cap. Our results suggest that the renin-angiotensin system might play an important role in the development of cardiac diseases induced by PG-LPS.
Collapse
Affiliation(s)
- Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ichiro Matsuo
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasuharu Amitani
- Department of Mathematics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
8
|
Ito A, Ohnuki Y, Suita K, Matsuo I, Ishikawa M, Mitsubayashi T, Mototani Y, Kiyomoto K, Tsunoda M, Morii A, Nariyama M, Hayakawa Y, Tomonari H, Okumura S. Effects of the angiotensin-converting enzyme inhibitor captopril on occlusal-disharmony-induced cardiac dysfunction in mice. Sci Rep 2023; 13:19927. [PMID: 37968296 PMCID: PMC10651878 DOI: 10.1038/s41598-023-43099-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 09/19/2023] [Indexed: 11/17/2023] Open
Abstract
Occlusal disharmony is known to affect not only the oral cavity environment, but also the autonomic nervous system in the heart. Since the renin-angiotensin system (RAS) inhibitor captopril (Cap) is one of the first-line drugs for preventing cardiac remodeling in patients with heart failure, we hypothesized that Cap might prevent cardiac dysfunction induced by occlusal disharmony. Here, to test this idea, we used our bite-opening (BO) mouse model, which was developed by cementing a suitable appliance onto the mandibular incisor. Mice were divided into four groups: (1) Control, (2) BO, (3) Cap, and (4) BO + Cap. After 2 weeks, we evaluated cardiac function by echocardiography and confirmed that cardiac function was significantly decreased in the BO group compared to the control, while Cap ameliorated the dysfunction. Cardiac fibrosis, myocyte apoptosis and oxidative stress-induced myocardial damage in the BO group were significantly increased versus the control, and these increases were suppressed by Cap. Cardiac dysfunction induced by BO was associated with dual phosphorylation on PKCδ (Tyr-311/Thr-505), leading to activation of CaMKII with increased phosphorylation of RyR2 and phospholamban. Our results suggest that the RAS might play an important role in the development of cardiac diseases induced by occlusal anomalies.
Collapse
Affiliation(s)
- Aiko Ito
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Ichiro Matsuo
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Takao Mitsubayashi
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, 236-8501, Japan
| | - Yoshio Hayakawa
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Hiroshi Tomonari
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan.
| |
Collapse
|
9
|
Tsunoda M, Matsuo I, Ohnuki Y, Suita K, Ishikawa M, Mitsubayashi T, Ito A, Mototani Y, Kiyomoto K, Morii A, Nariyama M, Hayakawa Y, Gomi K, Okumura S. Vidarabine, an anti-herpes agent, improves Porphyromonas gingivalis lipopolysaccharide-induced cardiac dysfunction in mice. J Physiol Sci 2023; 73:18. [PMID: 37558983 PMCID: PMC10717078 DOI: 10.1186/s12576-023-00873-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/02/2023] [Indexed: 08/11/2023]
Abstract
In this work, we examined the involvement of type 5 adenylyl cyclase (AC5) in cardiac dysfunction induced in mice given Porphyromonas gingivalis lipopolysaccharide (PG-LPS) at a dose equivalent to the circulating levels in periodontitis (PD) patients. Cardiac function was significantly decreased in mice given PG-LPS compared to the control, but treatment for 1 week with the AC5 inhibitor vidarabine ameliorated the dysfunction. Cardiac fibrosis and myocyte apoptosis were significantly increased in the PG-LPS group, but vidarabine blocked these changes. The PG-LPS-induced cardiac dysfunction was associated with activation of cyclic AMP/Ca2+-calmodulin-dependent protein kinase II signaling and increased phospholamban phosphorylation at threonine 17. These results suggest that pharmacological AC5 inhibition may be a promising approach to treat PD-associated cardiovascular disease.
Collapse
Affiliation(s)
- Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Ichiro Matsuo
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Takao Mitsubayashi
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Aiko Ito
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, 236-8501, Japan
| | - Yoshio Hayakawa
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan.
| |
Collapse
|
10
|
Wu M, Xing Q, Duan H, Qin G, Sang N. Suppression of NADPH oxidase 4 inhibits PM 2.5-induced cardiac fibrosis through ROS-P38 MAPK pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 837:155558. [PMID: 35504386 DOI: 10.1016/j.scitotenv.2022.155558] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/14/2022] [Accepted: 04/23/2022] [Indexed: 06/14/2023]
Abstract
Fine particulate matter (PM2.5) has been consistently linked to cardiovascular diseases, and cardiac fibrosis plays a crucial role in the occurrence and development of heart diseases. It is reported that NOX4-dependent redox signaling are responsible for TGFβ-mediated profibrotic responses. The current study was designed to explore the possible mechanisms of cardiac fibrosis by PM2.5 both in vitro and in vivo. Female C57BL/6 mice received PM2.5 (3 mg/kg b.w.) exposure with/without NOX4 inhibitor (apocynin, 25 mg/kg b.w.) or ROS scavenger (NALC, 50 mg/kg b.w.), every other day, for 4 weeks. H9C2 cells were incubated with PM2.5 (3 μg/mL) with/without 5 mM NALC, TGFβ inhibitor (SB431542, 10 μM), or siRNA-NOX4 for 24 h. The results demonstrated that PM2.5 induced evident collagen deposition and elevated expression of fibrosis biomarkers (Col1a1 & Col3a1). Significant systemic inflammatory response and cardiac oxidative stress were triggered by PM2.5. PM2.5 increased the protein expression of TGFβ1, NOX4, and P38 MAPK. Notably, the increased effects of PM2.5 could be suppressed by SB431542, siRNA-NOX4 in vitro or apocynin in vivo, and NALC. The reverse verification experiments further supported the involvement of the TGFβ/NOX4/ROS/P38 MAPK signaling pathway in the myocardial fibrosis induced by PM2.5. In summary, the current study provided evidence that PM2.5 challenge led to cardiac fibrosis through oxidative stress, systemic inflammation, and subsequent TGFβ/NOX4/ROS/P38 MAPK pathway and may offer new therapeutic targets in cardiac fibrosis.
Collapse
Affiliation(s)
- Meiqiong Wu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China; Department of Children and Adolescences Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China.
| | - Qisong Xing
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Huiling Duan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guohua Qin
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
11
|
Ji M, Liu Y, Zuo Z, Xu C, Lin L, Li Y. Downregulation of amphiregulin improves cardiac hypertrophy via attenuating oxidative stress and apoptosis. Biol Direct 2022; 17:21. [PMID: 35996142 PMCID: PMC9394079 DOI: 10.1186/s13062-022-00334-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
Amphiregulin (AREG) is a ligand of epidermal growth factor receptor and participates in the fibrosis of multiple organs. However, whether AREG can regulate hypertrophic cardiomyopathy is not well known. This research aims to explore the effect of AREG on cardiac hypertrophy, and whether the oxidative stress and apoptosis was involved in the influence of AREG on cardiac hypertrophy. Angiotensin (Ang) II induced cardiac hypertrophy in mice and neonatal rat cardiomyocytes (NRCMs) or HL-1 cells in vitro. AREG expressions raised in the heart of mice. After AREG downregulation, the increases of Ang II induced cardiac weight and cardiomyocytes area were inhibited. Down-regulation of AREG could inhibit Ang II induced the increases of atrial natriuretic peptide, brain natriuretic peptide, beta-myosin heavy chain in the heart of mice, and NRCMs and HL-1 cells. The enhancement of oxidative stress in mice heart with Ang II treatment was alleviated by AREG knockdown. The raises of Ang II induced Bax and cleaved caspase3 in mice heart were inhibited by AREG downregulation. AREG downregulation reduced myocardial hypertrophy via inhibition of oxidative and apoptosis. AREG may be a target for future cardiac hypertrophy treatment.
Collapse
Affiliation(s)
- Mingyue Ji
- Department of Cardiology, Lianshui County People's Hospital, Huaian, China
| | - Yun Liu
- Department of Intensive Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi Zuo
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Cheng Xu
- Department of Cardiology, Lianshui County People's Hospital, Huaian, China
| | - Li Lin
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, 150 JimoRoad, Shanghai, 200120, China.
| | - Yong Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
12
|
Tan R, Cong T, Xu G, Hao Z, Liao J, Xie Y, Lin Y, Yang X, Li Q, Liu Y, Xia YL. Anthracycline-Induced Atrial Structural and Electrical Remodeling Characterizes Early Cardiotoxicity and Contributes to Atrial Conductive Instability and Dysfunction. Antioxid Redox Signal 2022; 37:19-39. [PMID: 35081742 DOI: 10.1089/ars.2021.0002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aims: Cancer patients treated with anthracyclines are susceptible to atrial fibrillation (AF), while the mechanisms remain unclear. Due to sudden and unpredictable features, prediction of anthracycline-induced AF at early phase is difficult. Clinically, we tested whether anthracycline-induced early atrial remodeling in patients could be detected by echocardiography. Experimentally, we investigated the mechanisms of doxorubicin-induced atrial remodeling and AF in mice, and the protective effects of dexrazoxane and antioxidants. Methods and Results: Postsurgery breast cancer patients with an anthracycline-containing or anthracycline exclusion regimen were recruited for echocardiography before chemotherapy, and 3 and 6 months after chemotherapy. Mice were injected with doxorubicin or vehicle (5 mg/kg/week, 4 weeks), and left atrial diameter, electrical transmission, and AF inducibility were measured. Meanwhile, the level of reactive oxygen species (ROS), activity of antioxidant enzymes, cardiomyocyte size, vacuolization, inflammation, and fibrosis were also measured in mouse atria. The therapeutic effects of dexrazoxane and antioxidants on doxorubicin-induced changes in the aforementioned parameters were also determined. While ventricular parameters and functions were unchanged in cancer patients receiving anthracyclines before and after chemotherapy, left atrial reservoir and conduit function were decreased at 3 months postchemotherapy versus prechemotherapy. Doxorubicin-induced susceptibility to AF occurred in mice before onset of ventricular dysfunction. Doxorubicin-induced AF was via inducing structural remodeling (cardiomyocyte death, hypotrophy, and vacuolization) and electrical remodeling (reduction and redistribution of connexin 43) in atria, which was effectively prevented by dexrazoxane or antioxidants through inhibiting ROS generation or enhancing ROS elimination. Innovation and Conclusion: AF inducibility was induced after doxorubicin injection, which can be inhibited by repressing the ROS level. Antioxid. Redox Signal. 37, 19-39. The Clinical Trial Registration number is PJ-KS-KY-2019-73.
Collapse
Affiliation(s)
- Ruopeng Tan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tao Cong
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guiwen Xu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhujing Hao
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunpeng Xie
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yajuan Lin
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaolei Yang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingsong Li
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yang Liu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yun-Long Xia
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
13
|
Panda P, Verma HK, Lakkakula S, Merchant N, Kadir F, Rahman S, Jeffree MS, Lakkakula BVKS, Rao PV. Biomarkers of Oxidative Stress Tethered to Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9154295. [PMID: 35783193 PMCID: PMC9249518 DOI: 10.1155/2022/9154295] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD) is a broad term that incorporated a group of conditions that affect the blood vessels and the heart. CVD is a foremost cause of fatalities around the world. Multiple pathophysiological mechanisms are involved in CVD; however, oxidative stress plays a vital role in generating reactive oxygen species (ROS). Oxidative stress occurs when the concentration of oxidants exceeds the potency of antioxidants within the body while producing reactive nitrogen species (RNS). ROS generated by oxidative stress disrupts cell signaling, DNA damage, lipids, and proteins, thereby resulting in inflammation and apoptosis. Mitochondria is the primary source of ROS production within cells. Increased ROS production reduces nitric oxide (NO) bioavailability, which elevates vasoconstriction within the arteries and contributes to the development of hypertension. ROS production has also been linked to the development of atherosclerotic plaque. Antioxidants can decrease oxidative stress in the body; however, various therapeutic drugs have been designed to treat oxidative stress damage due to CVD. The present review provides a detailed narrative of the oxidative stress and ROS generation with a primary focus on the oxidative stress biomarker and its association with CVD. We have also discussed the complex relationship between inflammation and endothelial dysfunction in CVD as well as oxidative stress-induced obesity in CVD. Finally, we discussed the role of antioxidants in reducing oxidative stress in CVD.
Collapse
Affiliation(s)
- Poojarani Panda
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lung Biology and Disease, Helmholtz Zentrum, 85764 Neuherberg, Munich, Germany
| | | | - Neha Merchant
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali, 304022 Rajasthan, India
| | - Fairrul Kadir
- Department of Emergency Medicine, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Shamsur Rahman
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Mohammad Saffree Jeffree
- Department of Community and Family Medicine, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, 88400 Sabah, Malaysia
| | | | - Pasupuleti Visweswara Rao
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Abdurrab University, Jalan Riau Ujung No. 73, Pekanbaru, 28292 Riau, Indonesia
- Centre for International Relations and Research Collaborations, Reva University, Rukmini Knowledge Park, Kattigenahalli, Yelahanka, Bangalore, 560064 Karnataka, India
| |
Collapse
|
14
|
Matsuo I, Kawamura N, Ohnuki Y, Suita K, Ishikawa M, Matsubara T, Mototani Y, Ito A, Hayakawa Y, Nariyama M, Morii A, Kiyomoto K, Tsunoda M, Gomi K, Okumura S. Role of TLR4 signaling on Porphyromonas gingivalis LPS-induced cardiac dysfunction in mice. PLoS One 2022; 17:e0258823. [PMID: 35648750 PMCID: PMC9159598 DOI: 10.1371/journal.pone.0258823] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 05/14/2022] [Indexed: 12/02/2022] Open
Abstract
Oral infections, particularly periodontitis, are a well-established risk factor for cardiovascular diseases, although the molecular mechanisms involved remain elusive. The aims of the present study were to investigate the effects of lipopolysaccharide derived from Porphyromonas gingivalis (PG-LPS) on cardiac function in mice, and to elucidate the underlying mechanisms. Mice (C57BL/6) were injected with PG-LPS (0.8 mg/kg/day) with or without an inhibitor of Toll-like receptor 4 (TLR4) signaling (TAK-242, 0.8 mg/kg/day) for 4 weeks. Left ventricular ejection function was significantly decreased at 1 week (from 67 ± 0.5 to 58 ± 1.2%) and remained low at 4 weeks (57 ± 1.0%). The number of apoptotic myocytes was increased (approximately 7.4-fold), the area of fibrosis was increased (approximately 3.3-fold) and the number of 8-hydroxydeoxyguanosine-positive myocytes, a sensitive indicator of oxidative DNA damage, was increased (approximately 7.6-fold) at 4 weeks in the heart of PG-LPS treated mice. However, levels of various serum pro-inflammatory cytokines in PG-LPS-treated mice were similar to those in control mice. The impairment of cardiac function in PG-LPS-treated mice appears to involve activation of TLR4-NADPH oxidase (NOX) 4 signaling, leading to abundant production of reactive oxygen species and Ca2+ leakage from sarcoplastic reticulumn induced by calmodulin kinase II (CaMKII)-mediated phosphorylation of phospholamban (at Thr-17) and ryanodine receptor 2 (at Ser-2448). Pharmacological inhibition of TLR4 with TAK-242 attenuated the changes in cardiac function in PG-LPS-treated mice. Our results indicate that TLR4-NOX4 signaling may be a new therapeutic target for treatment of cardiovascular diseases in patients with periodontitis.
Collapse
Affiliation(s)
- Ichiro Matsuo
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Naoya Kawamura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Takehiro Matsubara
- Division of BioBank, Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- * E-mail:
| |
Collapse
|
15
|
Hayakawa Y, Suita K, Ohnuki Y, Mototani Y, Ishikawa M, Ito A, Nariyama M, Morii A, Kiyomoto K, Tsunoda M, Matsuo I, Kawahara H, Okumura S. Vidarabine, an anti-herpes agent, prevents occlusal-disharmony-induced cardiac dysfunction in mice. J Physiol Sci 2022; 72:2. [PMID: 35148678 PMCID: PMC10717220 DOI: 10.1186/s12576-022-00826-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/17/2022] [Indexed: 11/10/2022]
Abstract
We recently reported a positive relationship between occlusal disharmony and cardiovascular disease via activation of β-adrenergic signaling in mice. Furthermore, inhibition of type 5 adenylyl cyclase (AC5), a major cardiac subtype in adults, protects the heart against oxidative stress. Here, we examined the role of AC5 in the development of occlusal-disharmony-induced cardiovascular disease in bite-opening (BO) mice, prepared by cementing a suitable appliance onto the mandibular incisor. We first examined the effects of BO treatment on cardiac function in mice treated or not treated for 2 weeks with vidarabine, which we previously identified as an inhibitor of cardiac AC. Cardiac function was significantly decreased in the BO group compared to the control group, but vidarabine ameliorated the dysfunction. Cardiac fibrosis, myocyte apoptosis and myocyte oxidative DNA damage were significantly increased in the BO group, but vidarabine blocked these changes. The BO-induced cardiac dysfunction was associated with increased phospholamban phosphorylation at threonine-17 and serine-16, as well as increased activation of the Ca2+-calmodulin-dependent protein kinase II/receptor-interacting protein 3 signaling pathway. These data suggest that AC5 inhibition with vidarabine might be a new therapeutic approach for the treatment of cardiovascular disease associated with occlusal disharmony.
Collapse
Affiliation(s)
- Yoshio Hayakawa
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Aiko Ito
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, 236-8501, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Ichiro Matsuo
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Hiroshi Kawahara
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, 230-8501, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, 230-8501, Japan.
| |
Collapse
|
16
|
Dhalla NS, Elimban V, Bartekova M, Adameova A. Involvement of Oxidative Stress in the Development of Subcellular Defects and Heart Disease. Biomedicines 2022; 10:biomedicines10020393. [PMID: 35203602 PMCID: PMC8962363 DOI: 10.3390/biomedicines10020393] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
It is now well known that oxidative stress promotes lipid peroxidation, protein oxidation, activation of proteases, fragmentation of DNA and alteration in gene expression for producing myocardial cell damage, whereas its actions for the induction of fibrosis, necrosis and apoptosis are considered to result in the loss of cardiomyocytes in different types of heart disease. The present article is focused on the discussion concerning the generation and implications of oxidative stress from various sources such as defective mitochondrial electron transport and enzymatic reactions mainly due to the activation of NADPH oxidase, nitric oxide synthase and monoamine oxidase in diseased myocardium. Oxidative stress has been reported to promote excessive entry of Ca2+ due to increased permeability of the sarcolemmal membrane as well as depressions of Na+-K+ ATPase and Na+-Ca2+ exchange systems, which are considered to increase the intracellular of Ca2+. In addition, marked changes in the ryanodine receptors and Ca2+-pump ATPase have been shown to cause Ca2+-release and depress Ca2+ accumulation in the sarcoplasmic reticulum as a consequence of oxidative stress. Such alterations in sarcolemma and sarcoplasmic reticulum are considered to cause Ca2+-handling abnormalities, which are associated with mitochondrial Ca2+-overload and loss of myofibrillar Ca2+-sensitivity due to oxidative stress. Information regarding the direct effects of different oxyradicals and oxidants on subcellular organelles has also been outlined to show the mechanisms by which oxidative stress may induce Ca2+-handling abnormalities. These observations support the view that oxidative stress plays an important role in the genesis of subcellular defects and cardiac dysfunction in heart disease.
Collapse
Affiliation(s)
- Naranjan S. Dhalla
- St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada;
- Correspondence: ; Tel.: +1-204-235-3417; Fax: +1-204-237-0347
| | - Vijayan Elimban
- St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada;
| | - Monika Bartekova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Dubravska cesta 9, 84104 Bratislava, Slovakia; (M.B.); (A.A.)
| | - Adriana Adameova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Dubravska cesta 9, 84104 Bratislava, Slovakia; (M.B.); (A.A.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Odbojarov 10, 83232 Bratislava, Slovakia
| |
Collapse
|
17
|
Wu Q, Gurpinar A, Roberts M, Camelliti P, Ruggieri MR, Wu C. Identification of the NADPH Oxidase (Nox) Subtype and the Source of Superoxide Production in the Micturition Centre. BIOLOGY 2022; 11:183. [PMID: 35205049 PMCID: PMC8868587 DOI: 10.3390/biology11020183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/18/2022] [Indexed: 01/25/2023]
Abstract
Oxidative inflammatory damage to specialised brain centres may lead to dysfunction of their associated peripheral organs, such as the bladder. However, the source of reactive oxygen species (ROS) in specific brain regions that regulate bladder function is poorly understood. Of all ROS-generating enzymes, the NADPH oxidase (Nox) family produces ROS as its sole function and offers an advantage over other enzymes as a drug-targetable molecule to selectively control excessive ROS. We investigated whether the Nox 2 subtype is expressed in the micturition regulatory periaqueductal gray (PAG) and Barrington's nucleus (pontine micturition centre, PMC) and examined Nox-derived ROS production in these structures. C57BL/6J mice were used; PAG, PMC, cardiac tissue, and aorta were isolated. Western blot determined Nox 2 expression. Lucigenin-enhanced chemiluminescence quantified real-time superoxide production. Western blot experiments demonstrated the presence of Nox 2 in PAG and PMC. There was significant NADPH-dependent superoxide production in both brain tissues, higher than that in cardiac tissue. Superoxide generation in these brain tissues was significantly suppressed by the Nox inhibitor diphenyleneiodonium (DPI) and also reduced by the Nox-2 specific inhibitor GSK2795039, comparable to aorta. These data provide the first evidence for the presence of Nox 2 and Nox-derived ROS production in micturition centres.
Collapse
Affiliation(s)
- Qin Wu
- School of Medicine, Jiangsu Vocational College of Medicine, Yancheng 224005, China
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK
| | - Ayse Gurpinar
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK
| | - Maxwell Roberts
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK
| | - Patrizia Camelliti
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK
| | - Michael R Ruggieri
- Department of Anatomy & Cell Biology, Temple University, Philadelphia, PA 19122, USA
| | - Changhao Wu
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
18
|
Nunes JPS, Andrieux P, Brochet P, Almeida RR, Kitano E, Honda AK, Iwai LK, Andrade-Silva D, Goudenège D, Alcântara Silva KD, Vieira RDS, Levy D, Bydlowski SP, Gallardo F, Torres M, Bocchi EA, Mano M, Santos RHB, Bacal F, Pomerantzeff P, Laurindo FRM, Teixeira PC, Nakaya HI, Kalil J, Procaccio V, Chevillard C, Cunha-Neto E. Co-Exposure of Cardiomyocytes to IFN-γ and TNF-α Induces Mitochondrial Dysfunction and Nitro-Oxidative Stress: Implications for the Pathogenesis of Chronic Chagas Disease Cardiomyopathy. Front Immunol 2021; 12:755862. [PMID: 34867992 PMCID: PMC8632642 DOI: 10.3389/fimmu.2021.755862] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
Infection by the protozoan Trypanosoma cruzi causes Chagas disease cardiomyopathy (CCC) and can lead to arrhythmia, heart failure and death. Chagas disease affects 8 million people worldwide, and chronic production of the cytokines IFN-γ and TNF-α by T cells together with mitochondrial dysfunction are important players for the poor prognosis of the disease. Mitochondria occupy 40% of the cardiomyocytes volume and produce 95% of cellular ATP that sustain the life-long cycles of heart contraction. As IFN-γ and TNF-α have been described to affect mitochondrial function, we hypothesized that IFN-γ and TNF-α are involved in the myocardial mitochondrial dysfunction observed in CCC patients. In this study, we quantified markers of mitochondrial dysfunction and nitro-oxidative stress in CCC heart tissue and in IFN-γ/TNF-α-stimulated AC-16 human cardiomyocytes. We found that CCC myocardium displayed increased levels of nitro-oxidative stress and reduced mitochondrial DNA as compared with myocardial tissue from patients with dilated cardiomyopathy (DCM). IFN-γ/TNF-α treatment of AC-16 cardiomyocytes induced increased nitro-oxidative stress and decreased the mitochondrial membrane potential (ΔΨm). We found that the STAT1/NF-κB/NOS2 axis is involved in the IFN-γ/TNF-α-induced decrease of ΔΨm in AC-16 cardiomyocytes. Furthermore, treatment with mitochondria-sparing agonists of AMPK, NRF2 and SIRT1 rescues ΔΨm in IFN-γ/TNF-α-stimulated cells. Proteomic and gene expression analyses revealed that IFN-γ/TNF-α-treated cells corroborate mitochondrial dysfunction, transmembrane potential of mitochondria, altered fatty acid metabolism and cardiac necrosis/cell death. Functional assays conducted on Seahorse respirometer showed that cytokine-stimulated cells display decreased glycolytic and mitochondrial ATP production, dependency of fatty acid oxidation as well as increased proton leak and non-mitochondrial oxygen consumption. Together, our results suggest that IFN-γ and TNF-α cause direct damage to cardiomyocytes’ mitochondria by promoting oxidative and nitrosative stress and impairing energy production pathways. We hypothesize that treatment with agonists of AMPK, NRF2 and SIRT1 might be an approach to ameliorate the progression of Chagas disease cardiomyopathy.
Collapse
Affiliation(s)
- João Paulo Silva Nunes
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil.,INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Pauline Andrieux
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Pauline Brochet
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Rafael Ribeiro Almeida
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Eduardo Kitano
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - André Kenji Honda
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Leo Kei Iwai
- Laboratório Especial de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Débora Andrade-Silva
- Laboratório Especial de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - David Goudenège
- Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France
| | - Karla Deysiree Alcântara Silva
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Raquel de Souza Vieira
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Débora Levy
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Paulo Bydlowski
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Frédéric Gallardo
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Magali Torres
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Edimar Alcides Bocchi
- Heart Failure Team, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | | | - Fernando Bacal
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pablo Pomerantzeff
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Priscila Camillo Teixeira
- Translational Research Sciences, Pharma Research and Early Development F. Hoffmann-La Roche, Basel, Switzerland
| | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Vincent Procaccio
- MitoLab, UMR CNRS 6015-INSERM U1083, Université d'Angers, Angers, France
| | - Christophe Chevillard
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| |
Collapse
|
19
|
Smolka C, Schlösser D, Koentges C, Tarkhnishvili A, Gorka O, Pfeifer D, Bemtgen X, Asmussen A, Groß O, Diehl P, Moser M, Bode C, Bugger H, Grundmann S, Pankratz F. Cardiomyocyte-specific miR-100 overexpression preserves heart function under pressure overload in mice and diminishes fatty acid uptake as well as ROS production by direct suppression of Nox4 and CD36. FASEB J 2021; 35:e21956. [PMID: 34605573 DOI: 10.1096/fj.202100829rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022]
Abstract
MicroRNAs are key regulators of the cardiac response to injury. MiR-100 has recently been suggested to be involved in different forms of heart failure, but functional studies are lacking. In the present study, we examined the impact of transgenic miR-100 overexpression on cardiac structure and function during physiological aging and pathological pressure-overload-induced heart failure in mice after transverse aortic constriction surgery. MiR-100 was moderately upregulated after induction of pressure overload in mice. While in our transgenic model the cardiomyocyte-specific overexpression of miR-100 did not result in an obvious cardiac phenotype in unchallenged mice, the transgenic mouse strain exhibited less left ventricular dilatation and a higher ejection fraction than wildtype animals, demonstrating an attenuation of maladaptive cardiac remodeling by miR-100. Cardiac transcriptome analysis identified a repression of several regulatory genes related to cardiac metabolism, lipid peroxidation, and production of reactive oxygen species (ROS) by miR-100 overexpression, possibly mediating the observed functional effects. While the modulation of ROS-production seemed to be indirectly affected by miR-100 via Alox5-and Nox4-downregulation, we demonstrated that miR-100 induced a direct repression of the scavenger protein CD36 in murine hearts resulting in a decreased uptake of long-chain fatty acids and an alteration of mitochondrial respiratory function with an enhanced glycolytic state. In summary, we identified miR-100 as a modulator of cardiac metabolism and ROS production without an apparent cardiac phenotype at baseline but a protective effect under conditions of pressure-overload-induced cardiac stress, providing new insight into the mechanisms of heart failure.
Collapse
Affiliation(s)
- Christian Smolka
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Delia Schlösser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Koentges
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aleksandre Tarkhnishvili
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Xavier Bemtgen
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Asmussen
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Diehl
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Moser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Sebastian Grundmann
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Franziska Pankratz
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
20
|
Zhang M, Xu Y, Chen J, Qin C, Liu J, Guo D, Wang R, Hu J, Zou Q, Yang J, Wang Z, Niu X. Beta3-Adrenergic Receptor Activation Alleviates Cardiac Dysfunction in Cardiac Hypertrophy by Regulating Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3417242. [PMID: 34646422 PMCID: PMC8505079 DOI: 10.1155/2021/3417242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/10/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Excessive myocardial oxidative stress could lead to the congestive heart failure. NADPH oxidase is involved in the pathological process of left ventricular (LV) remodeling and dysfunction. β3-Adrenergic receptor (AR) could regulate cardiac dysfunction proved by recent researches. The molecular mechanism of β3-AR regulating oxidative stress, especially NADPH oxidase, remains to be determined. METHODS Cardiac hypertrophy was constructed by the transverse aortic constriction (TAC) model. ROS and NADPH oxidase subunits expression were assessed after β3-AR agonist (BRL) or inhibitor (SR) administration in cardiac hypertrophy. Moreover, the cardiac function, fibrosis, heart size, oxidative stress, and cardiomyocytes apoptosis were also detected. RESULTS β3-AR activation significantly alleviated cardiac hypertrophy and remodeling in pressure-overloaded mice. β3-AR stimulation also improved heart function and reduced cardiomyocytes apoptosis, oxidative stress, and fibrosis. Meanwhile, β3-AR stimulation inhibited superoxide anion production and decreased NADPH oxidase activity. Furthermore, BRL treatment increased the neuronal NOS (nNOS) expression in cardiac hypertrophy. CONCLUSION β3-AR stimulation alleviated cardiac dysfunction and reduced cardiomyocytes apoptosis, oxidative stress, and fibrosis by inhibiting NADPH oxidases. In addition, the protective effect of β3-AR is largely attributed to nNOS activation in cardiac hypertrophy.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Jianghong Chen
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Chaoshi Qin
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jing Liu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Rui Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jianqiang Hu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qing Zou
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jingxiao Yang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zikuan Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaolin Niu
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
21
|
de Castro JM, Assumpção JAF, Stein DJ, Toledo RS, da Silva LS, Caumo W, Carraro CC, da Rosa Araujo AS, Torres ILS. Nicotinamide riboside reduces cardiometabolic risk factors and modulates cardiac oxidative stress in obese Wistar rats under caloric restriction. Life Sci 2020; 263:118596. [PMID: 33080243 DOI: 10.1016/j.lfs.2020.118596] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/30/2020] [Accepted: 10/08/2020] [Indexed: 12/15/2022]
Abstract
AIMS NAD-based therapeutic strategies are encouraged against obesity and heart disease. Our study, therefore, aimed to investigate the effects of nicotinamide riboside (NR), isolated or combined with caloric restriction (CR), both approaches well-known for stimulating NAD levels, on adiposity parameters, cardiometabolic factors and cardiac oxidative stress in rats submitted to cafeteria diet (CAF). MAIN METHODS After 42 days of CAF-induced obesity (hypercaloric and ultra-processed foods common to humans), we examined the effects of oral administration of NR (400 mg/kg for 28 days), combined or not with CR (-62% kcal, for 28 days), on anthropometric, metabolic, tissue, and cardiac oxidative stress parameters in obese male Wistar rats. KEY FINDINGS In obese rats, treatment with NR alone mitigated final body weight gain, reduced adiposity (visceral and subcutaneous), improved insulin resistance, and decreased TG/HDL ratio and heart size. In cardiac OS, treatment with NR increased the antioxidant capacity via glutathione peroxidase and catalase enzymes (in rats under CR) as well as reduced the pro-oxidant complex NADPH oxidase (in obese and lean rats). Hyperglycemia, hypertriglyceridemia and elevated levels of TBARS in the heart were state-dependent adverse effects, induced by treatment with NR. SIGNIFICANCE This is the first study to report effects of nicotinamide riboside on cardiac oxidative stress in an obesity model. Nicotinamide riboside, a natural dietary compound, presented antiobesity effects and cardiometabolic benefits, in addition to positively modulating oxidative stress in the heart, in a state-dependent manner.
Collapse
Affiliation(s)
- Josimar Macedo de Castro
- Programa de Pós-Graduação em Ciências Biológicas (PPG): Farmacologia e Terapêutica - Instituto de Ciências Básicas da Saúde (ICBS) - Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
| | - José Antônio Fagundes Assumpção
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas - Faculdade de Medicina - UFRGS, Porto Alegre, RS, Brazil
| | - Dirson João Stein
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas - Faculdade de Medicina - UFRGS, Porto Alegre, RS, Brazil
| | - Roberta Ströher Toledo
- Programa de Pós-Graduação em Ciências Biológicas (PPG): Farmacologia e Terapêutica - Instituto de Ciências Básicas da Saúde (ICBS) - Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil
| | - Lisiane Santos da Silva
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas - Faculdade de Medicina - UFRGS, Porto Alegre, RS, Brazil
| | - Wolnei Caumo
- Programa de Pós-Graduação em Medicina: Ciências Médicas - Faculdade de Medicina - UFRGS, Porto Alegre, RS, Brazil
| | - Cristina Campos Carraro
- Laboratório de Fisiologia Cardiovascular e Espécies Reativas de Oxigênio, Departamento de Fisiologia - ICBS - UFRGS, Porto Alegre, RS, Brazil
| | - Alex Sander da Rosa Araujo
- Laboratório de Fisiologia Cardiovascular e Espécies Reativas de Oxigênio, Departamento de Fisiologia - ICBS - UFRGS, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Programa de Pós-Graduação em Ciências Biológicas (PPG): Farmacologia e Terapêutica - Instituto de Ciências Básicas da Saúde (ICBS) - Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas - Faculdade de Medicina - UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
22
|
Waghela BN, Vaidya FU, Agrawal Y, Santra MK, Mishra V, Pathak C. Molecular insights of NADPH oxidases and its pathological consequences. Cell Biochem Funct 2020; 39:218-234. [PMID: 32975319 DOI: 10.1002/cbf.3589] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/18/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS), formed by the partial reduction of oxygen, were for a long time considered to be a byproduct of cellular metabolism. Since, increase in cellular levels of ROS results in oxidative stress leading to damage of nucleic acids, proteins, and lipids resulting in numerous pathological conditions; ROS was considered a bane for aerobic species. Hence, the discovery of NADPH oxidases (NOX), an enzyme family that specifically generates ROS as its prime product came as a surprise to redox biologists. NOX family proteins participate in various cellular functions including cell proliferation and differentiation, regulation of genes and protein expression, apoptosis, and host defence immunological response. Balanced expression and activation of NOX with subsequent production of ROS are critically important to regulate various genes and proteins to maintain homeostasis of the cell. However, dysregulation of NOX activation leading to enhanced ROS levels is associated with various pathophysiologies including diabetes, cardiovascular diseases, neurodegenerative diseases, ageing, atherosclerosis, and cancer. Although our current knowledge on NOX signifies its importance in the normal functioning of various cellular pathways; yet the choice of ROS producing enzymes which can tip the scale from homeostasis toward damage, as mediators of biological functions remain an oddity. Though the role of NOX in maintaining normal cellular functions is now deemed essential, yet its dysregulation leading to catastrophic events cannot be denied. Hence, this review focuses on the involvement of NOX enzymes in various pathological conditions imploring them as possible targets for therapies. SIGNIFICANCE OF THE STUDY: The NOXs are multi-subunit enzymes that generate ROS as a prime product. NOX generated ROS are usually regulated by various molecular factors and play a vital role in different physiological processes. The dysregulation of NOX activity is associated with pathological consequences. Recently, the dynamic proximity of NOX enzymes with different molecular signatures of pathologies has been studied extensively. It is essential to identify the precise role of NOX machinery in its niche during the progression of pathology. Although inhibition of NOX could be a promising approach for therapeutic interventions, it is critical to expand the current understanding of NOX's dynamicity and shed light on their molecular partners and regulators.
Collapse
Affiliation(s)
- Bhargav N Waghela
- School of Biological Sciences & Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Foram U Vaidya
- School of Biological Sciences & Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Yashika Agrawal
- Laboratory of Molecular Cancer Biology and Epigenetics, National Centre for Cell Science, Pune, Maharashtra, India
| | - Manas Kumar Santra
- Laboratory of Molecular Cancer Biology and Epigenetics, National Centre for Cell Science, Pune, Maharashtra, India
| | - Vinita Mishra
- School of Biological Sciences & Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Chandramani Pathak
- School of Biological Sciences & Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India
| |
Collapse
|
23
|
Marqués J, Cortés A, Pejenaute Á, Zalba G. Implications of NADPH oxidase 5 in vascular diseases. Int J Biochem Cell Biol 2020; 128:105851. [PMID: 32949687 DOI: 10.1016/j.biocel.2020.105851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/30/2022]
Abstract
Oxidative stress is one of the main mechanisms involved in the pathophysiology of vascular diseases. Among others, oxidative stress promotes endothelial dysfunction, and accelerated ageing and remodelling of vasculature. Lately, NADPH oxidases have been demonstrated to be involved in cardiovascular diseases. NADPH oxidase 5 has emerged as a new player in oxidative stress-mediated endothelial alterations, involved in the pathophysiology of hypertension, diabetes, atherosclerosis, myocardial infarction and stroke. This oxidase seems to mediate its detrimental effects by promoting inflammation. NADPH oxidase 5 has been studied in a lesser extent compared with the other members of the NADPH oxidase family due to its loss in the rodent genome, the main experimental research model. In addition, its potential as a therapeutic target remains unexplored given the lack of specific inhibitors. In this review the latest findings on NADPH oxidase 5 regulation, implications in vascular pathophysiology and therapeutic approaches will be updated.
Collapse
Affiliation(s)
- Javier Marqués
- Department of Biochemistry and Genetics, University of Navarra, Pamplona Spain; Navarra Institute for Health Research (IdiSNA), Pamplona Spain
| | - Adriana Cortés
- Department of Biochemistry and Genetics, University of Navarra, Pamplona Spain; Navarra Institute for Health Research (IdiSNA), Pamplona Spain
| | - Álvaro Pejenaute
- Department of Biochemistry and Genetics, University of Navarra, Pamplona Spain; Navarra Institute for Health Research (IdiSNA), Pamplona Spain
| | - Guillermo Zalba
- Department of Biochemistry and Genetics, University of Navarra, Pamplona Spain; Navarra Institute for Health Research (IdiSNA), Pamplona Spain.
| |
Collapse
|
24
|
Zhao GJ, Zhao CL, Ouyang S, Deng KQ, Zhu L, Montezano AC, Zhang C, Hu F, Zhu XY, Tian S, Liu X, Ji YX, Zhang P, Zhang XJ, She ZG, Touyz RM, Li H. Ca 2+-Dependent NOX5 (NADPH Oxidase 5) Exaggerates Cardiac Hypertrophy Through Reactive Oxygen Species Production. Hypertension 2020; 76:827-838. [PMID: 32683902 DOI: 10.1161/hypertensionaha.120.15558] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022]
Abstract
NOX5 (NADPH oxidase 5) is a homolog of the gp91phox subunit of the phagocyte NOX, which generates reactive oxygen species. NOX5 is involved in sperm motility and vascular contraction and has been implicated in diabetic nephropathy, atherosclerosis, and stroke. The function of NOX5 in the cardiac hypertrophy is unknown. Because NOX5 is a Ca2+-sensitive, procontractile NOX isoform, we questioned whether it plays a role in cardiac hypertrophy. Studies were performed in (1) cardiac tissue from patients undergoing heart transplant for cardiomyopathy and heart failure, (2) NOX5-expressing rat cardiomyocytes, and (3) mice expressing human NOX5 in a cardiomyocyte-specific manner. Cardiac hypertrophy was induced in mice by transverse aorta coarctation and Ang II (angiotensin II) infusion. NOX5 expression was increased in human failing hearts. Rat cardiomyocytes infected with adenoviral vector encoding human NOX5 cDNA exhibited elevated reactive oxygen species levels with significant enlargement and associated increased expression of ANP (atrial natriuretic peptides) and β-MHC (β-myosin heavy chain) and prohypertrophic genes (Nppa, Nppb, and Myh7) under Ang II stimulation. These effects were reduced by N-acetylcysteine and diltiazem. Pressure overload and Ang II infusion induced left ventricular hypertrophy, interstitial fibrosis, and contractile dysfunction, responses that were exaggerated in cardiac-specific NOX5 trangenic mice. These phenomena were associated with increased reactive oxygen species levels and activation of redox-sensitive MAPK (mitogen-activated protein kinase). N-acetylcysteine treatment reduced cardiac oxidative stress and attenuated cardiac hypertrophy in NOX5 trangenic. Our study defines Ca2+-regulated NOX5 as an important NOX isoform involved in oxidative stress- and MAPK-mediated cardiac hypertrophy and contractile dysfunction.
Collapse
Affiliation(s)
- Guo-Jun Zhao
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (G.-J.Z., C.-L.Z., L.-H.Z., C.Z., X.-Y.Z., S.T., X.-J.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
| | - Chang-Ling Zhao
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (G.-J.Z., C.-L.Z., L.-H.Z., C.Z., X.-Y.Z., S.T., X.-J.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
| | - Shan Ouyang
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
- Basic Medical School, Wuhan University, China (S.O., H.L.)
| | - Ke-Qiong Deng
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
- Department of Cardiology (K.-Q.D.), Zhongnan Hospital of Wuhan University, China
| | - Lihua Zhu
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (G.-J.Z., C.-L.Z., L.-H.Z., C.Z., X.-Y.Z., S.T., X.-J.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Centre, University of Glasgow, United Kingdom (A.C.M., R.M.T.)
| | - Changjiang Zhang
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (G.-J.Z., C.-L.Z., L.-H.Z., C.Z., X.-Y.Z., S.T., X.-J.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
| | - Fengjiao Hu
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
- Medical Science Research Center (F.H., X.L., Y.-X.J., P.Z., H.L.), Zhongnan Hospital of Wuhan University, China
| | - Xue-Yong Zhu
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (G.-J.Z., C.-L.Z., L.-H.Z., C.Z., X.-Y.Z., S.T., X.-J.Z., Z.-G.S., H.L.)
| | - Song Tian
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
| | - Xiaolan Liu
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
- Medical Science Research Center (F.H., X.L., Y.-X.J., P.Z., H.L.), Zhongnan Hospital of Wuhan University, China
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
- Medical Science Research Center (F.H., X.L., Y.-X.J., P.Z., H.L.), Zhongnan Hospital of Wuhan University, China
| | - Peng Zhang
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
- Medical Science Research Center (F.H., X.L., Y.-X.J., P.Z., H.L.), Zhongnan Hospital of Wuhan University, China
| | - Xiao-Jing Zhang
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
| | - Zhi-Gang She
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (G.-J.Z., C.-L.Z., L.-H.Z., C.Z., X.-Y.Z., S.T., X.-J.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Centre, University of Glasgow, United Kingdom (A.C.M., R.M.T.)
| | - Hongliang Li
- From the Department of Cardiology, Renmin Hospital of Wuhan University, China (G.-J.Z., C.-L.Z., L.-H.Z., C.Z., X.-Y.Z., S.T., X.-J.Z., Z.-G.S., H.L.)
- Institute of Model Animal of Wuhan University, China (G.-J.Z., C.-L.Z., S.O., K.-Q.D., L.-H.Z., C.Z., F.H., X.-.Z., S.T., X.L., Y.-X.J., P.Z., X.-J.Z., Z.-G.S., H.L.)
- Basic Medical School, Wuhan University, China (S.O., H.L.)
- Medical Science Research Center (F.H., X.L., Y.-X.J., P.Z., H.L.), Zhongnan Hospital of Wuhan University, China
| |
Collapse
|
25
|
Apocynin Treatment Prevents Cardiac Connexin 43 Hemichannels Hyperactivity by Reducing Nitroso-Redox Stress in Mdx Mice. Int J Mol Sci 2020; 21:ijms21155415. [PMID: 32751416 PMCID: PMC7432655 DOI: 10.3390/ijms21155415] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal disease that causes cardiomyopathy and is associated with oxidative stress. In the heart, oxidative stress interferes with the location of connexin 43 (Cx43) to the intercalated discs causing its lateralization to the plasma membrane where Cx43 forms hemichannels. We tested the hypothesis that in DMD cardiomyopathy, increased oxidative stress is associated with the formation and activation of Cx43 hemichannels. For this, we used mdx mice as a DMD model and evaluated cardiac function, nitroso-redox changes and Cx43 hemichannels permeability. Mdx hearts presented increased NADPH oxidase-derived oxidative stress and increased Cx43 S-nitrosylation compared to controls. These redox changes were associated with increased Cx43 lateralization, decreased cardiac contractility and increased arrhythmic events. Pharmacological inhibition of NADPH oxidase using apocynin (one month) reduced systemic oxidative stress and reversed the aforementioned changes towards normal, except Cx43 lateralization. Opening of Cx43 hemichannels was blocked by apocynin treatment and by acute hemichannel blockade with carbenoxolone. NADPH oxidase inhibition also prevented the occurrence of apoptosis in mdx hearts and reversed the ventricular remodeling. These results show that NADPH oxidase activity in DMD is associated with S-nitrosylation and opening of Cx43 hemichannels. These changes lead to apoptosis and cardiac dysfunction and were prevented by NADPH oxidase inhibition.
Collapse
|
26
|
Li F, Lei C. Fucoxanthin protects neonatal rat cardiomyocytes and attenuates high glucose-mediated oxidative stress via the AMPK pathway. Arch Med Sci 2020; 21:272-284. [PMID: 40190296 PMCID: PMC11969558 DOI: 10.5114/aoms.2020.94432] [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] [Received: 10/10/2019] [Accepted: 02/15/2020] [Indexed: 04/09/2025] Open
Abstract
Introduction Diabetic cardiomyopathy (DC) is associated with impaired diastolic function. Hyperglycemia-mediated oxidative stress and apoptosis are the major factors responsible for DC. Also NADPH oxidase is the main source of ROS in cardiac cells or cardiomyocytes. Here we evaluated the effect of fucoxanthin (FXN) on high glucose cultured neonatal rat cardiomyocytes. Material and methods For the study, Iry neonatal rat cardiomyocytes were cultured in a high glucose environment (30 mM/l) in the presence and absence of FXN. Apoptosis, cell viability, activity of NADPH oxidase and expression level of its subunits, levels of MDA and activity of endogenous antioxidant enzymes were studied. We also confirmed the involved pathway by studying the expression of AMPK, GSK-3β and Akt in the cardiomyocytes. Results The high glucose environment increased the production of ROS, and FXN decreased the oxidative burden by inhibiting ROS in cultured neonatal rat cardiomyocytes. FXN inhibited the activity of NADPH oxidase and Rac1 also increased the expression of its subunits. Treatment of FXN reversed the MDA, CAT, GSHpx, SOD activity and GSH contents. FXN down-regulated the levels of Bax and up-regulated the levels of Bcl-2 (anti-apoptotic protein); treatment protected the cardiomyocytes from injury. Also, FXN increased the levels of pAMPK in cardiac cells treated with high glucose. The pharmacological inhibitor of AMPK abolished the activities of FXN in high glucose induced cardiomyocytes. Conclusions FXN exerted protective action on cardiac cells subjected to high glucose-mediated apoptosis by suppressing NADPH oxidase-mediated production of ROS and maintaining the antioxidant defense in the tissues. The attenuating activity of FXN was propagated via the AMPK cascade.
Collapse
Affiliation(s)
- Fengyue Li
- Department of Pediatrics, Jiangsu Taizhou People’s Hospital, Taizhou City, Jiangsu Province, China
| | - Chunxia Lei
- Department of Neonatology, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
27
|
Resveratrol and Diabetic Cardiomyopathy: Focusing on the Protective Signaling Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7051845. [PMID: 32256959 PMCID: PMC7094200 DOI: 10.1155/2020/7051845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/01/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a common cardiovascular complication of diabetic mellitus that is characterized by diastolic disorder in the early stage and clinical heart failure in the later stage. Presently, DCM is considered one of the major causes of death in diabetic patients. Resveratrol (RSV), a naturally occurring stilbene, is widely reported as a cardioprotective substance in many heart diseases. Thus far, the specific roles of RSV in DCM prevention and treatment have attracted great attention. Here, we discuss the roles of RSV in DCM by focusing its downstream targets from both in vivo and in vitro studies. Among such targets, Sirtuins 1/3 and AMP-activated kinase have been identified as key mediators that induce cardioprotection during hyperglycemia. In addition, many other signaling molecules (e.g., forkhead box-O3a and extracellular regulated protein kinases) are also regulated in the presence of RSV and exert beneficial effects such as opposing oxidative stress, inflammation, and apoptosis in cardiomyocytes exposed to high-glucose conditions. The beneficial potential of an RSV/stem cell cotherapy is also reviewed as a promising therapeutic strategy for preventing the development of DCM.
Collapse
|
28
|
Bkaily G, Najibeddine W, Jacques D. Increase of NADPH oxidase 3 in heart failure of hereditary cardiomyopathy. Can J Physiol Pharmacol 2019; 97:902-908. [DOI: 10.1139/cjpp-2019-0055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the development of heart failure in humans and animal models, an increase in reactive oxygen species (ROS) levels was observed. However, there is no information whether this increase of ROS is associated with an increase in the density of specific isoforms of NADPH oxidases (NOXs) 1–5. The objective of this study was to verify whether the densities of NOXs 1–5 change during the development of heart failure. Using the well-known model of cardiomyopathic hamsters, the UM-X 7.1 line, a model that strongly resembles the pathology observed in humans from a morphological and functional point of view, our studies showed that, as in humans, NOXs 1–5 are present in both normal and UM-X7.1 hamster hearts. Even though the densities of NOXs 2 and 5 were unchanged, the levels of both NOXs 1 and 4 significantly decreased in UM-X7.1 hamster hearts during heart failure. These changes were accompanied with a significant increase in NOX3 level. These results suggest that, during heart failure, NOX3 plays a vital role in compensating the decrease of NOXs 1 and 4. This increase in NOX3 may also be responsible, at least in part, for the reported increase in ROS levels in heart failure.
Collapse
Affiliation(s)
- Ghassan Bkaily
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Wassim Najibeddine
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Danielle Jacques
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
29
|
MicroRNA-21 abrogates palmitate-induced cardiomyocyte apoptosis through caspase-3/NF-κB signal pathways. Anatol J Cardiol 2019; 20:336-346. [PMID: 30504734 PMCID: PMC6287441 DOI: 10.14744/anatoljcardiol.2018.03604] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Objective: The aim of the study was to investigate the role of microRNA-21 (miR-21) in cardiomyocyte apoptosis and to determine a possible mechanism. Methods: H9c2 embryonic rat heart-derived cells were used in the study. Cell viability was determined using the 3-(4.5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, and flow cytometry was used to evaluate cell apoptosis. Reverse transcription-polymerase chain reaction and western blot assays were used to detect mRNA and protein expression of the apoptosis-related proteins and miR-21. ELISA was used to detect reactive oxygen species (ROS). Results: Palmitate exposure greatly reduced miR-21 expression in cardiomyocytes. Apoptosis increased when miR-21 was inhibited with or without palmitate exposure. Consistently, reduced apoptosis was observed when miR-21 was overexpressed in cardiomyocytes. Caspase-3 activity was reduced after palmitate exposure. Bcl-2 protein expression was increased in H9c2 cells when transfected with the miR-21 mimic. MiR-21 overexpression alone did not induce ROS or DNA fragmentation; however, in conjunction with palmitate exposure, miR-21 mimic reduced ROS and DNA fragmentation. Moreover, palmitate administration overcame the antioxidant effect of 3 mM N-acetylcysteine to significantly inhibit apoptosis, DNA fragmentation, and caspase-3 activity. The exposure to palmitate greatly reduced p65 and p-p38 expression in the nucleus. A p38 inhibitor had no effect on the expression of Bcl-2 and cleaved caspase-3 in H9c2 cells alone; however, when combined with exposure to palmitate the p38 inhibitor induced Bcl-2 expression and inhibited caspase-3 activity. The p38 inhibitor by itself did not induce apoptosis, ROS production, or DNA fragmentation in H9c2 cells, but when palmitate was included with the p38 inhibitor, apoptosis, ROS production, and DNA fragmentation were reduced. Conclusion: miR-21 protects cardiomyocytes from apoptosis that is induced by palmitate through the caspase-3/NF-κB signal pathways.
Collapse
|
30
|
H 2O 2 Metabolism in Normal Thyroid Cells and in Thyroid Tumorigenesis: Focus on NADPH Oxidases. Antioxidants (Basel) 2019; 8:antiox8050126. [PMID: 31083324 PMCID: PMC6563055 DOI: 10.3390/antiox8050126] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 12/23/2022] Open
Abstract
Thyroid hormone synthesis requires adequate hydrogen peroxide (H2O2) production that is utilized as an oxidative agent during the synthesis of thyroxin (T4) and triiodothyronine (T3). Thyroid H2O2 is generated by a member of the family of NADPH oxidase enzymes (NOX-es), termed dual oxidase 2 (DUOX2). NOX/DUOX enzymes produce reactive oxygen species (ROS) as their unique enzymatic activity in a timely and spatially regulated manner and therefore, are important regulators of diverse physiological processes. By contrast, dysfunctional NOX/DUOX-derived ROS production is associated with pathological conditions. Inappropriate DUOX2-generated H2O2 production results in thyroid hypofunction in rodent models. Recent studies also indicate that ROS improperly released by NOX4, another member of the NOX family, are involved in thyroid carcinogenesis. This review focuses on the current knowledge concerning the redox regulation of thyroid hormonogenesis and cancer development with a specific emphasis on the NOX and DUOX enzymes in these processes.
Collapse
|
31
|
Ding M, Guan L, Zhang C, Yang P, Yang H. (3R)‑5,6,7‑trihydroxy‑3‑isopropyl‑3‑methylisochroman‑1‑one attenuates cardiac dysfunction via the apelin/APJ signaling pathway. Mol Med Rep 2019; 19:5007-5014. [PMID: 30942406 DOI: 10.3892/mmr.2019.10122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 03/26/2019] [Indexed: 11/05/2022] Open
Abstract
Myocardial infarction (MI) is associated with a high risk of mortality and is a major global health concern. The present study aimed to investigate the protective effects of (3R)‑5,6,7‑trihydroxy‑3‑isopropyl‑3‑methylisochroman‑1‑one (TIM) against MI induced by isoproterenol (ISO) in a rat model and the underlying mechanisms. Wistar rats were assigned to 4 groups (n=10): The control group received saline treatment; the ISO group received an intraperitoneal injection of ISO (100 mg/kg); and the TIM (low) and TIM (high) groups received an intraperitoneal injection of ISO, plus a 1 and 2 mg/kg dose of TIM orally, respectively. TIM rats were treated with TIM daily for 12 days and received ISO injections on the final 2 days to induce MI. Cardiac function, apoptosis index and protein expression were subsequently determined. The levels of oxidative stress markers were determined by ELISAs, whereas DNA damage was detected using a Cell Death Detection ELISA kit. Gene and protein expression were determined via reverse transcription‑quantitative polymerase chain reaction and western blot analyses, respectively. Following treatment with ISO, the maximum left ventricular contraction/relaxation velocity and left ventricular systolic pressure were significantly decreased, whereas the left ventricular end‑diastolic pressure was increased; however, treatment with TIM significantly ameliorated ISO‑induced cardiac dysfunction. Additionally, TIM treatment significantly decreased oxidative stress and inhibited the apoptosis of cardiomyocytes, as determined by a decrease in caspase activities, increased expression of B‑cell lymphoma 2 (Bcl‑2) and reduced expression of cleaved caspase‑3, cleaved caspase‑9 and Bcl‑2‑associated X. Furthermore, treatment with TIM upregulated the levels of apelin in the plasma and myocardium of ISO‑treated rats. The results indicated that TIM protected cardiomyocytes against ISO‑induced MI, potentially via the apelin/apelin receptor signaling pathway. The results of the present study suggested that TIM may be a potential novel therapy for the treatment of MI.
Collapse
Affiliation(s)
- Mei Ding
- Department of Cardiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Lianyue Guan
- Department of Hepatobiliary‑Pancreatic Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Chunmei Zhang
- Department of Emergency, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ping Yang
- Department of Cardiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hailing Yang
- Department of Emergency, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
32
|
Neidhardt S, Garbade J, Emrich F, Klaeske K, Borger MA, Lehmann S, Jawad K, Dieterlen MT. Ischemic Cardiomyopathy Affects the Thioredoxin System in the Human Myocardium. J Card Fail 2019; 25:204-212. [PMID: 30721734 DOI: 10.1016/j.cardfail.2019.01.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/03/2018] [Accepted: 01/23/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Oxidative stress due to reactive oxygen species (ROS) production is a key factor in the development of heart failure (HF). This study investigated the thioredoxin (Trx) system, which plays a major role in antioxidant defense, in patients suffering from ischemic (ICM) or dilated (DCM) cardiomyopathy. METHODS AND RESULTS Myocardial tissue from ICM (n = 13) and DCM (n = 13) patients, as well as septal tissue of patients with aortic stenosis but without diagnosed hypertrophic cardiomyopathy or subaortic stenosis (control; n = 12), was analyzed for Trx1, Trx-interacting protein (TXNIP) and E3 ligase ITCH (E3 ubiquitin-protein ligase Itchy homolog) expression. Trx-reductase 1 (TXNRD1) amount and activity, cytosolic cytochrome C content, and apoptosis markers were quantified by means of enzyme-linked immunosorbent assay and multiplexing. Compared with control samples, ITCH and Trx1 expression, TXNRD1 amount and activity were reduced and TXNIP expression was increased in ICM (ITCH: P = .013; Trx1: P = .028; TXNRD1 amount: P = .035; TXNRD1 activity: P = .005; TXNIP: P = .014) but not in DCM samples. A higher level of the downstream apoptosis marker caspase-9 (ICM: 582 ± 262 MFI [P = .995]; DCM: 1251 ± 548 MFI [P = .002], control: 561 ± 214 MFI) was detected in DCM tissue. A higher expression of Bcl-2 was found in DCM (P = .011). CONCLUSION The Trx system was impaired in ICM but not in DCM. ITCH appeared to be responsible for the down-regulation of the Trx system. ROS-induced mitochondrial instability appeared to play a role in DCM.
Collapse
Affiliation(s)
- Stephan Neidhardt
- Department of Cardiac Surgery, University of Leipzig, Heart Center Leipzig, Helios Clinic, Leipzig, Germany
| | - Jens Garbade
- Department of Cardiac Surgery, University of Leipzig, Heart Center Leipzig, Helios Clinic, Leipzig, Germany
| | - Fabian Emrich
- Department of Cardiac Surgery, University of Leipzig, Heart Center Leipzig, Helios Clinic, Leipzig, Germany
| | - Kristin Klaeske
- Department of Cardiac Surgery, University of Leipzig, Heart Center Leipzig, Helios Clinic, Leipzig, Germany
| | - Michael A Borger
- Department of Cardiac Surgery, University of Leipzig, Heart Center Leipzig, Helios Clinic, Leipzig, Germany
| | - Sven Lehmann
- Department of Cardiac Surgery, University of Leipzig, Heart Center Leipzig, Helios Clinic, Leipzig, Germany
| | - Khalil Jawad
- Department of Cardiac Surgery, University of Leipzig, Heart Center Leipzig, Helios Clinic, Leipzig, Germany
| | - Maja-Theresa Dieterlen
- Department of Cardiac Surgery, University of Leipzig, Heart Center Leipzig, Helios Clinic, Leipzig, Germany.
| |
Collapse
|
33
|
Zeglinski MR, Moghadam AR, Ande SR, Sheikholeslami K, Mokarram P, Sepehri Z, Rokni H, Mohtaram NK, Poorebrahim M, Masoom A, Toback M, Sareen N, Saravanan S, Jassal DS, Hashemi M, Marzban H, Schaafsma D, Singal P, Wigle JT, Czubryt MP, Akbari M, Dixon IM, Ghavami S, Gordon JW, Dhingra S. Myocardial Cell Signaling During the Transition to Heart Failure. Compr Physiol 2018; 9:75-125. [DOI: 10.1002/cphy.c170053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
34
|
Casas AI, Geuss E, Kleikers PWM, Mencl S, Herrmann AM, Buendia I, Egea J, Meuth SG, Lopez MG, Kleinschnitz C, Schmidt HHHW. NOX4-dependent neuronal autotoxicity and BBB breakdown explain the superior sensitivity of the brain to ischemic damage. Proc Natl Acad Sci U S A 2017. [PMID: 29087944 DOI: 10.1073/pnas.1705034114/-/dcsupplemental] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Ischemic injury represents the most frequent cause of death and disability, and it remains unclear why, of all body organs, the brain is most sensitive to hypoxia. In many tissues, type 4 NADPH oxidase is induced upon ischemia or hypoxia, converting oxygen to reactive oxygen species. Here, we show in mouse models of ischemia in the heart, brain, and hindlimb that only in the brain does NADPH oxidase 4 (NOX4) lead to ischemic damage. We explain this distinct cellular distribution pattern through cell-specific knockouts. Endothelial NOX4 breaks down the BBB, while neuronal NOX4 leads to neuronal autotoxicity. Vascular smooth muscle NOX4, the common denominator of ischemia within all ischemic organs, played no apparent role. The direct neuroprotective potential of pharmacological NOX4 inhibition was confirmed in an ex vivo model, free of vascular and BBB components. Our results demonstrate that the heightened sensitivity of the brain to ischemic damage is due to an organ-specific role of NOX4 in blood-brain-barrier endothelial cells and neurons. This mechanism is conserved in at least two rodents and humans, making NOX4 a prime target for a first-in-class mechanism-based, cytoprotective therapy in the unmet high medical need indication of ischemic stroke.
Collapse
Affiliation(s)
- Ana I Casas
- Department of Pharmacology and Personalized Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Eva Geuss
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Pamela W M Kleikers
- Department of Pharmacology and Personalized Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Stine Mencl
- Department of Neurology, University Clinics Essen, D-45147 Essen, Germany
| | - Alexander M Herrmann
- Department of Neurology, Westfälische Wilhelms-Universität Münster, 48149 Münster, Germany
| | - Izaskun Buendia
- Instituto Teofilo Hernando, Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Javier Egea
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Sven G Meuth
- Department of Neurology, Westfälische Wilhelms-Universität Münster, 48149 Münster, Germany
| | - Manuela G Lopez
- Instituto Teofilo Hernando, Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany;
- Department of Neurology, University Clinics Essen, D-45147 Essen, Germany
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalized Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
35
|
NOX4-dependent neuronal autotoxicity and BBB breakdown explain the superior sensitivity of the brain to ischemic damage. Proc Natl Acad Sci U S A 2017; 114:12315-12320. [PMID: 29087944 PMCID: PMC5699031 DOI: 10.1073/pnas.1705034114] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Ischemic injury represents the most frequent cause of death and disability, and it remains unclear why, of all body organs, the brain is most sensitive to hypoxia. In many tissues, type 4 NADPH oxidase is induced upon ischemia or hypoxia, converting oxygen to reactive oxygen species. Here, we show in mouse models of ischemia in the heart, brain, and hindlimb that only in the brain does NADPH oxidase 4 (NOX4) lead to ischemic damage. We explain this distinct cellular distribution pattern through cell-specific knockouts. Endothelial NOX4 breaks down the BBB, while neuronal NOX4 leads to neuronal autotoxicity. Vascular smooth muscle NOX4, the common denominator of ischemia within all ischemic organs, played no apparent role. The direct neuroprotective potential of pharmacological NOX4 inhibition was confirmed in an ex vivo model, free of vascular and BBB components. Our results demonstrate that the heightened sensitivity of the brain to ischemic damage is due to an organ-specific role of NOX4 in blood-brain-barrier endothelial cells and neurons. This mechanism is conserved in at least two rodents and humans, making NOX4 a prime target for a first-in-class mechanism-based, cytoprotective therapy in the unmet high medical need indication of ischemic stroke.
Collapse
|
36
|
Wang Y, Zhong L, Liu X, Zhu YZ. ZYZ-772 Prevents Cardiomyocyte Injury by Suppressing Nox4-Derived ROS Production and Apoptosis. Molecules 2017; 22:molecules22020331. [PMID: 28230797 PMCID: PMC6155929 DOI: 10.3390/molecules22020331] [Citation(s) in RCA: 12] [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: 01/16/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 01/17/2023] Open
Abstract
Nox-dependent signaling plays critical roles in the development of heart failure, cardiac hypertrophy, and myocardial infarction. NADPH oxidase 4 (Nox4) as a major source of oxidative stress in the heart offers a new therapeutic target in cardiovascular disease. In the present work, a novel flavonoid was isolated from Zanthoxylum bungeanum. Its structure was elucidated as Quercetin-3-O-(6′′-O-α-l-rhamnopyransoyl)-β-d-glucopyranoside-7-O-β-d-glucopyranoside (ZYZ-772) for the first time. ZYZ-772 exhibited significant cardio-protective property against CoCl2 induced H9c2 cardiomyocyte cells injury. In CoCl2 stimulated cardiomyocyte injury, ZYZ-772 inhibited expression of Nox4, and alleviated ROS overproduction. Importantly, ROS triggered MAPKs phosphorylation and P53 signaling mediated apoptosis were restored by ZYZ-772. Our findings present the first piece of evidence for the therapeutic properties of ZYZ-772 in preventing cardiomyocyte injury, which could be attributed to the suppression of Nox4/MAPKs/P53 axis. This will offer a novel therapeutic strategy for the treatment of cardiac ischemia disease.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Liangjie Zhong
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Xinhua Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Yi Zhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
- School of Pharmacy, Macao University of Science and Technology, Macao.
| |
Collapse
|
37
|
Cardamonin Alleviates Pressure Overload-induced Cardiac Remodeling and Dysfunction Through Inhibition of Oxidative Stress. J Cardiovasc Pharmacol 2016; 68:441-451. [DOI: 10.1097/fjc.0000000000000430] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
38
|
Yu QG, Zhang Y. Transforming growth factor-β1 mediates NADPH oxidase 4: A significant contributor to the pathogenesis of myocardial fibrosis. Int J Cardiol 2016; 227:53-54. [PMID: 27846463 DOI: 10.1016/j.ijcard.2016.10.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/30/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Qi-Gui Yu
- The Binhu Hospital of Hefei, Hefei, Anhui, China; The First People of Hefei, Hefei, Anhui, China
| | - Ying Zhang
- The Binhu Hospital of Hefei, Hefei, Anhui, China; The First People of Hefei, Hefei, Anhui, China.
| |
Collapse
|
39
|
Statins and oxidative stress in chronic heart failure. Rev Port Cardiol 2016; 35:41-57. [PMID: 26763895 DOI: 10.1016/j.repc.2015.09.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/13/2015] [Indexed: 11/23/2022] Open
Abstract
Statins are the most commonly prescribed drugs for the treatment of dyslipidemia. They are also recommended in primary and secondary prevention of cardiovascular disease. In addition to decreasing cholesterol synthesis, statins interfere with the synthesis of isoprenoid intermediates, which may explain many of their pleiotropic properties, including their antioxidant effects. Oxidative stress is defined as an imbalance between the synthesis of reactive oxygen species and their elimination by antioxidant defense systems, with a prevailing pro-oxidant status that results in macromolecular damage and disruption of cellular redox signaling. Reactive oxygen species interfere with various processes that affect cardiac structure and function, contributing to the contractile dysfunction, myocardial hypertrophy and fibrosis observed in the pathophysiology of heart failure. By regulating several molecular pathways that control nicotinamide adenine dinucleotide phosphate oxidase and endothelial nitric oxide synthase activity, statins help restore redox homeostasis. These drugs also contribute to the control of inflammation and appear to have a protective role in various diseases. The results of observational studies and clinical trials with statins in heart failure have not been consensual. This review aims to analyze the role of oxidative stress in heart failure and the molecular mechanisms underlying statins' antioxidant properties. It also examines current scientific evidence on the use of these drugs as a specific treatment for heart failure.
Collapse
|
40
|
Statins and oxidative stress in chronic heart failure. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2016. [DOI: 10.1016/j.repce.2015.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
41
|
Lee KP, Kim JE, Park WH. Cytoprotective effect of rhamnetin on miconazole-induced H9c2 cell damage. Nutr Res Pract 2015; 9:586-91. [PMID: 26634046 PMCID: PMC4667198 DOI: 10.4162/nrp.2015.9.6.586] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 05/28/2015] [Accepted: 06/29/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/OBJECTIVES Reactive oxygen species (ROS) formation is closely related to miconazole-induced heart dysfunction. Although rhamnetin has antioxidant effects, it remained unknown whether it can protect against miconazole-induced cardiomyocyte apoptosis. Thus, we investigated the effects of rhamnetin on miconazole-stimulated H9c2 cell apoptosis. MATERIALS/METHODS Cell morphology was observed by inverted microscope and cell viability was determined using a WelCount™ cell proliferation assay kit. Miconazole-induced ROS production was evaluated by fluorescence-activated cell sorting with 6-carboxy-2',7'-dichlorofluoroscein diacetate (H2DCF-DA) stain. Immunoblot analysis was used to determine apurinic/apyrimidinic endonuclease 1 (APE/Ref-1) and cleaved cysteine-aspartic protease (caspase) 3 expression. NADPH oxidase levels were measured using real-time polymerase chain reaction. RESULTS Miconazole (3 and 10 µM) induced abnormal morphological changes and cell death in H9c2 cells. Rhamnetin enhanced the viability of miconazole (3 µM)-treated cells in a dose-dependent manner. Rhamnetin (1 and 3 µM) treatment downregulated cleaved caspase 3 and upregulated APE/Ref-1 expression in miconazole-stimulated cells. Additionally, rhamnetin significantly reduced ROS generation. CONCLUSIONS Our data suggest that rhamnetin may have cytoprotective effects in miconazole-stimulated H9c2 cardiomyocytes via ROS inhibition. This effect most likely occurs through the upregulation of APE/Ref-1 and attenuation of hydrogen peroxide levels.
Collapse
Affiliation(s)
- Kang Pa Lee
- Department of Medical Science, School of Medicine, Konkuk University, Seoul 143-701, Korea
| | - Jai-Eun Kim
- Department of Pathology, College of Korean Medicine, Dongguk University, Gyeonggi-Do 410-820, Korea
| | - Won-Hwan Park
- Department of Diagnosis, College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-Do 410-820, Korea
| |
Collapse
|
42
|
Guo S, Yao Q, Ke Z, Chen H, Wu J, Liu C. Resveratrol attenuates high glucose-induced oxidative stress and cardiomyocyte apoptosis through AMPK. Mol Cell Endocrinol 2015; 412:85-94. [PMID: 26054749 DOI: 10.1016/j.mce.2015.05.034] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 05/27/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) suggests a direct cellular insult to myocardium. Hyperglycemia-induced oxidative stress and apoptosis have been implicated in the pathogenesis of DCM. NADPH oxidase is a major source of reactive oxygen species (ROS) generation in cardiomyocytes. Resveratrol, a naturally occurring polyphenol, has shown beneficial effects on some cardiovascular complications associated with diabetes. OBJECTIVES We aimed to examine the role of resveratrol on high glucose-induced NADPH oxidase-derived ROS production and cardiac apoptosis, together with modulation of protein signaling pathways in cardiomyocytes. METHODS Primary cultures of neonatal rat cardiomyocytes were exposed to 30 mmol/L high glucose with or without resveratrol. Cell viability, apoptosis, superoxide formation, NADPH oxidase activity and its subunits expression, antioxidant enzymes activities, as well as the potential regulatory molecules AMPK, Akt and GSK-3β were assessed in cardiac cells. RESULTS Elevated ROS production induced by 30 mmol/L high glucose was inhibited with the addition of resveratrol in primary cultured neonatal rat cardiomyocytes. Consistently, resveratrol markedly suppressed the increased activity of NADPH oxidase and Rac1, as well as the enhanced expression of p67(phox), p47(phox), and gp91(phox) induced by high glucose. Lipid peroxidation, SOD, catalase, GSH-px activity and GSH content was reversed in the presence of resveratrol. Moreover, the expression of pro-apoptotic protein Bax was down regulated while anti-apoptotic protein Bcl-2 was up regulated. And cardiac cell injury and apoptosis were markedly rescued by resveratrol. In addition, resveratrol significantly increased phosphorylation of AMP-activated protein kinase (AMPK) at Thr172 in cardiomyocytes exposed to high glucose. Compound C, the pharmacologic inhibitor of AMPK, could mostly abrogate roles of resveratrol on cardiomyocytes in high glucose. In contrast, Akt and GSK-3β were little influenced by resveratrol. CONCLUSIONS Our data demonstrated that resveratrol protected cardiomyocytes against high glucose-induced apoptosis through suppression NADPH oxidase-derived ROS generation and maintenance endogenous antioxidant defenses. And the protective effects of resveratrol are mostly mediated by AMPK related pathway.
Collapse
Affiliation(s)
- Shuang Guo
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Qing Yao
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Zhiqiang Ke
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Hongguang Chen
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Jiliang Wu
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China.
| | - Chao Liu
- Hubei Province Key Laboratory on Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China.
| |
Collapse
|
43
|
Liu Y, Jiao R, Ma ZG, Liu W, Wu QQ, Yang Z, Li FF, Yuan Y, Bian ZY, Tang QZ. Sanguinarine inhibits angiotensin II-induced apoptosis in H9c2 cardiac cells via restoring reactive oxygen species-mediated decreases in the mitochondrial membrane potential. Mol Med Rep 2015; 12:3400-3408. [PMID: 26017473 PMCID: PMC4526052 DOI: 10.3892/mmr.2015.3841] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 04/15/2015] [Indexed: 12/25/2022] Open
Abstract
Cell apoptosis induced by Angiotensin II (Ang II) has a critical role in the development of cardiovascular diseases. The aim of the present study was to investigate whether sanguinarine (SAN), a drug which was proved to have anti-oxidant, anti-proliferative and immune enhancing effects, can abolish cell apoptosis induced by Ang II. In the present study, H9c2 cardiac cells were stimulated with 10 µM Ang II with or without SAN. The level of intracellular reactive oxygen species (ROS) generation was assessed using dichlorodihydrofluorescein diacetate, and changes of the mitochondrial membrane potential (MMP) were assessed using JC-1 staining. Furthermore, mRNA expression of NOX2 was determined by reverse transcription quantitative polymerase chain reaction, and apoptosis was detected by Annexin V/propidium iodide staining and flow cytometry. The expression of B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax) as well as cleaved (c)-caspase 3 and -9 were detected by western blot analysis, and the activity of caspase 3 and -9 was detected using an ELISA. The results of the present study showed that NOX2 expression and ROS generation induced by Ang II were inhibited by SAN, and the Ang 2-induced MMP loss was also ameliorated. Furthermore, Ang II-induced H9c2 cardiac cell apoptosis as well as c-caspase 3 and -9 levels were significantly reduced by SAN. Investigation of the possible pathway involved in the anti-apoptotic effect of SAN showed that the expression of Bcl-2 was decreased, while that of Bax was increased following stimulation with Ang II, which was reversed following treatment with SAN. In addition, Ang II enhanced the activity of caspase 9 and cleaved downstream caspases such as caspase-3, initiating the caspase cascade, while pre-treatment of H9c2 cardiac cells with SAN blocked these effects. In conclusion, the findings of the present study indicated that SAN inhibits the apoptosis of H9c2 cardiac cells induced by Ang II, most likely via restoring ROS-mediated decreases of the MMP.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Rong Jiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing-Qing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fang-Fang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhou-Yan Bian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
44
|
Li B, Tian J, Sun Y, Xu TR, Chi RF, Zhang XL, Hu XL, Zhang YA, Qin FZ, Zhang WF. Activation of NADPH oxidase mediates increased endoplasmic reticulum stress and left ventricular remodeling after myocardial infarction in rabbits. Biochim Biophys Acta Mol Basis Dis 2015; 1852:805-15. [DOI: 10.1016/j.bbadis.2015.01.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/12/2015] [Accepted: 01/16/2015] [Indexed: 12/21/2022]
|
45
|
Such GK, Yan Y, Johnston APR, Gunawan ST, Caruso F. Interfacing materials science and biology for drug carrier design. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:2278-2297. [PMID: 25728711 DOI: 10.1002/adma.201405084] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/11/2014] [Indexed: 06/04/2023]
Abstract
Over the last ten years, there has been considerable research interest in the development of polymeric carriers for biomedicine. Such delivery systems have the potential to significantly reduce side effects and increase the bioavailability of poorly soluble therapeutics. The design of carriers has relied on harnessing specific variations in biological conditions, such as pH or redox potential, and more recently, by incorporating specific peptide cleavage sites for enzymatic hydrolysis. Although much progress has been made in this field, the specificity of polymeric carriers is still limited when compared with their biological counterparts. To synthesize the next generation of carriers, it is important to consider the biological rationale for materials design. This requires a detailed understanding of the cellular microenvironments and how these can be harnessed for specific applications. In this review, several important physiological cues in the cellular microenvironments are outlined, with a focus on changes in pH, redox potential, and the types of enzymes present in specific regions. Furthermore, recent studies that use such biologically inspired triggers to design polymeric carriers are highlighted, focusing on applications in the field of therapeutic delivery.
Collapse
Affiliation(s)
- Georgina K Such
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | | | | | | |
Collapse
|
46
|
Abstract
SIGNIFICANCE Despite recent medical advances, cardiovascular disease and heart failure (HF) continue to be major health concerns, and related mortality remains high. As a result, investigation of the mechanisms involved in the development of HF continues to be an active field of study. RECENT ADVANCES The renin-angiotensin system (RAS) and its effector molecule, angiotensin (Ang) II, affect cardiac function through both systemic and local actions, and have been shown to play a major role in cardiac remodeling and dysfunction in the failing heart. Many of the downstream effects of AngII signaling are mediated by elevated levels of reactive oxygen species (ROS) and oxidative stress, which have also been implicated in the pathology of HF. CRITICAL ISSUES Inhibitors of the RAS have proven beneficial in the treatment of patients at risk for and suffering from HF, but remain only partially effective. ROS can be generated from several different sources, and the oxidative state is normally tightly regulated in the heart. How AngII increases ROS levels and causes dysregulation of the cardiac oxidative state has been the subject of considerable interest in recent years. FUTURE DIRECTIONS A better understanding of this process and the mechanisms involved should lead to the development of more effective HF therapies and improved outcomes.
Collapse
Affiliation(s)
- Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, University of Medicine and Dentistry of New Jersey , Newark, New Jersey
| | | |
Collapse
|
47
|
Koka S, Das A, Salloum FN, Kukreja RC. Phosphodiesterase-5 inhibitor tadalafil attenuates oxidative stress and protects against myocardial ischemia/reperfusion injury in type 2 diabetic mice. Free Radic Biol Med 2013; 60:80-8. [PMID: 23385031 DOI: 10.1016/j.freeradbiomed.2013.01.031] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 01/04/2013] [Accepted: 01/29/2013] [Indexed: 12/21/2022]
Abstract
Diabetic patients exhibit increased risk for the development of cardiovascular diseases primarily because of impaired nitric oxide (NO) bioavailability. The phosphodiesterase-5 (PDE-5) inhibitor sildenafil restores NO signaling and protects against ischemia/reperfusion (I/R) injury. In this study, we determined the effect of the long-acting PDE-5 inhibitor tadalafil on diabetes-associated complications and its role in attenuating oxidative stress after I/R injury in type 2 diabetic db/db mice. Adult male db/db mice (n=40/group) were randomized to receive dimethyl sulfoxide (10% DMSO, 0.2ml, ip) or tadalafil (1mg/kg in 10% DMSO, ip) for 28 days. After 28 days treatment, the hearts were isolated and subjected to 30min global ischemia followed by 60min reperfusion in the Langendorff mode. Infarct size was measured using computer morphometry of tetrazolium-stained sections. Cardiomyocytes were isolated from a subset of hearts and subjected to 40min simulated ischemia followed by 1h of reoxygenation (SI/RO). Dichlorodihydrofluorescein diacetate and JC-1 staining was used to measure reactive oxygen species (ROS) generation and mitochondrial membrane potential (Δψm), respectively. Another subset of hearts was used for the estimation of lipid peroxidation, glutathione, and the expression of myocardial pRac1, Rac1, gp91(phox), p47(phox), and p67(phox) by Western blot. Tadalafil treatment improved the metabolic status and reduced infarct size compared to the untreated db/db mice (21.2±1.8% vs 45.8±2.8%; p<0.01). The db/db mice showed enhanced oxidative stress in cardiomyocytes as indicated by a significant increase in ROS production. Cardiac NAD(P)H oxidase activity, lipid peroxidation, and oxidized glutathione were also increased in db/db mice compared to nondiabetic control animals. Tadalafil treatment in db/db mice suppressed oxidative stress, attenuated myocardial expression of pRac1 and gp91(phox), and also preserved the loss of Δψm in cardiomyocytes after SI/RO. In conclusion, these results demonstrate that chronic treatment with tadalafil attenuates oxidative stress and improves mitochondrial integrity while providing powerful cardioprotective effects in type 2 diabetes.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Carbolines/administration & dosage
- Cardiotonic Agents/administration & dosage
- Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Humans
- Mice
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/pathology
- Myocardial Reperfusion Injury/drug therapy
- Myocardial Reperfusion Injury/pathology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Nitric Oxide/metabolism
- Oxidative Stress/drug effects
- Oxidative Stress/genetics
- Phosphodiesterase 5 Inhibitors/administration & dosage
- Reactive Oxygen Species/metabolism
- Tadalafil
Collapse
Affiliation(s)
- Saisudha Koka
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298-0204, USA
| | | | | | | |
Collapse
|
48
|
QSYQ Attenuates Oxidative Stress and Apoptosis Induced Heart Remodeling Rats through Different Subtypes of NADPH-Oxidase. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:824960. [PMID: 23861715 PMCID: PMC3686095 DOI: 10.1155/2013/824960] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/08/2013] [Accepted: 05/11/2013] [Indexed: 11/17/2022]
Abstract
We aim to investigate the therapeutic effects of QSYQ, a drug of heart failure (HF) in clinical practice in China, on a rat heart failure (HF) model. 3 groups were divided: HF model group (LAD ligation), QSYQ group (LAD ligation and treated with QSYQ), and sham-operated group. After 4 weeks, rats were sacrificed for cardiac injury measurements. Rats with HF showed obvious histological changes including necrosis and inflammation foci, elevated ventricular remodeling markers levels(matrix metalloproteinases-2, MMP-2), deregulated ejection fraction (EF) value, increased formation of oxidative stress (Malondialdehyde, MDA), and up-regulated levels of apoptotic cells (caspase-3, p53 and tunnel) in myocardial tissue. Treatment of QSYQ improved cardiac remodeling through counter-acting those events. The improvement of QSYQ was accompanied with a restoration of NADPH oxidase 4 (NOX4) and NADPH oxidase 2 (NOX2) pathways in different patterns. Administration of QSYQ could attenuate LAD-induced HF, and AngII-NOX2-ROS-MMPs pathway seemed to be the critical potential targets for QSYQ to reduce the remodeling. Moreover, NOX4 was another key targets to inhibit the p53 and Caspase3, thus to reduce the hypertrophy and apoptosis, and eventually provide a synergetic cardiac protective effect.
Collapse
|
49
|
Muller O, Ntalianis A, Wijns W, Delrue L, Dierickx K, Auer R, Rodondi N, Mangiacapra F, Trana C, Hamilos M, Valentin E, De Bruyne B, Barbato E, Bartunek J. Association of biomarkers of lipid modification with functional and morphological indices of coronary stenosis severity in stable coronary artery disease. J Cardiovasc Transl Res 2013; 6:536-44. [PMID: 23670230 DOI: 10.1007/s12265-013-9468-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/25/2013] [Indexed: 12/12/2022]
Abstract
Biomarkers of blood lipid modification and oxidative stress have been associated with increased cardiovascular morbidity. We sought to determine whether these biomarkers were related to functional indices of stenosis severity among patients with stable coronary artery disease. We studied 197 consecutive patients with stable coronary artery disease due to single vessel disease. Fractional flow reserve (FFR) ≤ 0.80 was assessed as index of a functionally significant lesion. Serum levels of secretory phospholipase A2 (sPLA2) activity, secretory phospholipase A2 type IIA (sPLA2-IIA), myeloperoxydase (MPO), lipoprotein-associated phospholipase A2 (Lp-PLA2), and oxidized low-density lipoprotein (OxLDL) were assessed using commercially available assays. Patients with FFR > 0.8 had higher sPLA2 activity, sPLA2 IIA, and OxLDL levels than patients with FFR ≤ 0.8 (21.25 [16.03-27.28] vs 25.85 [20.58-34.63] U/mL, p < 0.001, 2.0 [1.5-3.4] vs 2.6 [2.0-3.4] ng/mL, p < 0.01; and 53.0 [36.0-71.0] vs 64.5 [50-89.25], p < 0.001 respectively). Patients with FFR > 0.80 had similar Lp-PLA2 and MPO levels versus those with FFR ≤ 0.8. sPLA2 activity, sPLA2 IIA significantly increased area under the curve over baseline characteristics to predict FFR ≤ 0.8 (0.67 to 0.77 (95 % confidence interval [CI]: 0.69-0.85) p < 0.01 and 0.67 to 0.77 (95 % CI: 0.69-0.84) p < 0.01, respectively). Serum sPLA2 activity as well as sPLA2-IIA level is related to functional characteristics of coronary stenoses in patients with stable coronary artery disease.
Collapse
Affiliation(s)
- Olivier Muller
- Cardiovascular Center and Translational Cardiology Unit, Aalst, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hahn NE, Meischl C, Kawahara T, Musters RJP, Verhoef VMJ, van der Velden J, Vonk ABA, Paulus WJ, van Rossum AC, Niessen HWM, Krijnen PAJ. NOX5 expression is increased in intramyocardial blood vessels and cardiomyocytes after acute myocardial infarction in humans. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2222-9. [PMID: 22503554 DOI: 10.1016/j.ajpath.2012.02.018] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 01/23/2012] [Accepted: 02/21/2012] [Indexed: 12/28/2022]
Abstract
Reactive oxygen species producing NADPH oxidases play important roles under different (patho)physiological conditions. NOX1, NOX2, and NOX4 are important sources of reactive oxygen species in the heart, but knowledge of the calcium-dependent NOX5 in the heart is lacking. The presence of NOX5 was studied via RT-PCR in heart tissue from patients with end-stage heart failure; the tissue was obtained during cardiac transplantation surgery. NOX5 positivity and cellular localization were studied via IHC and digital-imaging microscopy in heart tissues of patients who did not have heart disease and in infarction areas of patients who died of myocardial infarctions of different durations. Furthermore, NOX5 expression was analyzed in vitro by using Western blot analysis. NOX5 RNA was found in the hearts of controls and patients with ischemic cardiomyopathy. In controls, NOX5 localized to the endothelium of a limited number of intramyocardial blood vessels and to a limited number of scattered cardiomyocytes. In infarcted hearts, NOX5 expression increased, especially in infarctions >12 hours, which manifested as an increase in NOX5-positive intramyocardial blood vessels, as well as in endothelium, smooth muscle, and cardiomyocytes. NOX5 was found in cardiomyocyte cytoplasm, plasma membrane, intercalated disks, and cross striations. Western blot analysis confirmed NOX5 expression in isolated human cardiomyocytes. For the first time to our knowledge, we demonstrate NOX5 expression in human intramyocardial blood vessels and cardiomyocytes, with significant increases in the affected myocardium after acute myocardial infarction.
Collapse
Affiliation(s)
- Nynke E Hahn
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|