Next Article in Journal
Albuminuria-Related Genetic Biomarkers: Replication and Predictive Evaluation in Individuals with and without Diabetes from the UK Biobank
Previous Article in Journal
A Cross-Talk about Radioresistance in Lung Cancer—How to Improve Radiosensitivity According to Chinese Medicine and Medicaments That Commonly Occur in Pharmacies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Conjugated Linoleic Acid-Mediated Connexin-43 Remodeling and Sudden Arrhythmic Death in Myocardial Infarction

by
Natia Qipshidze Kelm
1,2,
Jane C. Solinger
1,
Kellianne M. Piell
1 and
Marsha P. Cole
1,2,*
1
Department of Biochemistry and Molecular Genetics, Louisville, KY 40202, USA
2
Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 11208; https://doi.org/10.3390/ijms241311208
Submission received: 1 June 2023 / Revised: 15 June 2023 / Accepted: 19 June 2023 / Published: 7 July 2023
(This article belongs to the Section Molecular Biology)

Abstract

:
Connexin 43 (Cx43) is expressed in the left and right ventricles and is primarily responsible for conducting physiological responses in microvasculature. Studies have demonstrated that NADPH oxidase (NOX) enzymes are essential in cardiac redox biology and are responsible for the generation of reactive oxygen species (ROS). NOX2 is linked to left ventricular remodeling following myocardial infarction (MI). It was hypothesized that conjugated linoleic acid (cLA) treatment increases NOX-2 levels in heart tissue and disrupts connexins between the myocytes in the ventricle. Data herein demonstrate that cLA treatment significantly decreases survival in a murine model of MI. The observance of cLA-induced ventricular tachyarrhythmia’s (VT) led to the subsequent investigation of the underlying mechanism in this MI model. Mice were treated with cLA for 12 h, 24 h, 48 h, or 72 h to determine possible time-dependent changes in NOX and Cx43 signaling pathways in isolated left ventricles (LV) extracted from cardiac tissue. The results suggest that ROS generation, through the stimulation of NOX2 in the LV, triggers a decrease in Cx43 levels, causing dysfunction of the gap junctions following treatment with cLA. This cascade of events may initiate VT and subsequent death during MI. Taken together, individuals at risk of MI should use caution regarding cLA consumption.

1. Introduction

Conjugated linoleic acid (cLA) refers to a group of positional and geometrical isomers of octadecadienoic acid, with two alternating double bonds. Rumen bacteria has the unique ability to convert linoleic acid into cLA via an enzymatic isomerase reaction, and therefore at least 28 possible isomers of linoleic acid are found in meat and dairy products [1]. The prominent cLA isomers that are commercially available as dietary supplements and in food products are mixtures of cis-9, trans-11-cLA and trans-10, cis-12-cL, with cis-9, trans-11 accounting for 72–94% of total cLA in foods. There is emerging evidence that individual cLA isomers may be responsible for specific biological or biochemical changes in the body. Most studies use isomeric mixtures, as there are 28 different geometric and positional isomers of cLA [1]. However, isomers cis-9, trans-11-cLA and trans-10, cis-12-cLA are the only two isomers to date that have been linked to specific biological effects. For example, both isomers have been shown to inhibit carcinogenesis. However, individually, cis-9, trans-11 is mainly responsible for anticarcinogenic effects, whereas the trans-10, cis-12 isomer reduces body fat and is commonly referred as the most effective isomer affecting blood lipids.
Numerous studies have reported positive effects of cLA consumption, including weight loss in obese individuals [2], modulation of immune function [3,4] anti-carcinogenic activity [5], protection against atherogenesis [6,7], reversal of atherosclerosis [8], and normalization of glucose and insulin homeostasis in pre-diabetic animal models [9,10]. Additionally, the Food and Drug Administration notably reported that cLA as being “generally regarded as safe” in 2008. However, we have since shown that cLA treatment lowers physiological nitric oxide (NO) levels and impairs heart function in aged mice [11]. Following these findings, we wanted to determine if survival was also affected as a result of impaired heart function.
Connexins are gap-junction transmembrane channels that mediate the cell-to-cell movement of small molecules and/or ions to maintain intercellular homeostasis [12]. Gap junctions contain transmembrane ion-channel proteins which are cylinders constructed from 6 copies of transmembrane proteins, otherwise known as connexins [12]. In the heart, connexins mediate ion flow through the cardiac myocytes, forming intercellular pathways that allow for the organization of electrical excitation responsible for heart muscle contraction [13,14].
Approximately twenty connexins have been identified in the mouse genome and twenty-one in the human genome, where orthologous are increasingly characterized in other vertebrates [15]. There are 3 main connexins expressed in the heart, including Connexin-43 (Cx-43), Connexin-40 (Cx-40), and Connexin-45 (Cx-45) [16]. Previous studies have shown that Cx-43 is highly expressed in both ventricles, where both atria equally express Cx-43 and Cx-40 [16,17]. Myocytes of the sinoatrial and atrioventricular nodes normally have small, isolated gap junctions composed of Cx-45 [16,18]. Since it is known that connexins are of essential importance during intercellular communication, changes in connexin expression and distribution are expected to have insightful impacts on cardiac conduction and the generation of arrhythmias. Previous studies have shown Cx-43 to be an arrhythmogenic junction in the heart [16,19]. Lerner et al. have shown that Connexin-43 (Cx43)-deficient mice were susceptible for ischemia-induced VT [20].
Excess amounts of reactive oxygen species (ROS) have been implicated in the genesis of arrhythmia by altering Cx-43 [21]. NADPH oxidase (NOX) enzymes have been implicated in cardiac redox biology and are responsible for the generation of ROS [22,23]. Of the NADPH oxidase homologs, NOX2 and NOX4 are most abundantly expressed in the myocardium [24]. Under physiological conditions, nonphagocytic NADPH oxidase enzymes generate lower levels of intracellular ROS that are necessary for cell signaling, adaptation and survival, unlike phagocytic NADPH oxidase, which releases a large “respiratory burst” of superoxide (O2) for pathogen extermination [23]. The NOX family is composed of seven members (Nox1–Nox5, Duox1, and Duox2) that transfer electrons across biological membranes to generate ROS [24]. NOX2 has been linked to left ventricular remodeling after myocardial infarction [25].
In the present study, we wanted to further examine the effects and potential mechanisms of cLA treatment on MI. Our results indicate that cLA treatment significantly decreases survival in a murine model of myocardial ischemia (MI), causing life-threatening spontaneous ventricular tachycardia (VT) and sudden cardiac death (SCD). Time point treatment was used to further elucidate the possible mechanism whereby cLA induces life-threatening arrhythmias. Herein, we demonstrate that cLA significantly induces levels of NOX2 expression and ROS generation in myocytes, which results in the disruption of connexins between the myocytes in the ventricle, causing life-threatening arrhythmias and subsequent death in mice during MI.

2. Results

2.1. Electrocardiogram (ECG) Recordings Prior to MI Surgery Demonstrate VT following Treatment with cLA

Structural and functional changes following MI are characterized by using non-invasive ECG to determine post-infarct cardiac remodeling. During the MI surgery, electrocardiogram (ECG) recordings in untreated mice detected normal sinus rhythms (Figure 1A). Following the ligation of LAD, mice without treatment developed ST-segment elevation, which is an earliest sign of MI (Figure 1B). Previous studies revealed that reduction of Cx-43 expression increases frequency of spontaneous ventricular ectopy and arrhythmogenesis compared to control mice [26]. Mice treated with cLA developed VT and died within 1–2 min following ligation of LAD (Figure 1C).

2.2. Treatment with cLA Does Not Affect Cardiac Function before MI

To establish baseline myocardial function, echocardiography was performed in 3-month-old mice (untreated). The analysis of left ventricular function, determined via percentage of fractional shortening (%FS), demonstrated that there were no significant changes in cardiac function between untreated and treated groups (Figure 2A,B). FS was normal in both control (53 ± 3%) and cLA-treated (51 ± 4%) groups (Figure 2 Table). These results indicate that cLA does not cause adverse myocardial changes in the cardiac function of normal, healthy mice. However, cLA treatment with MI results in abnormal heart rhythms and SCD.

2.3. Treatment with cLA Increases NOX2 Expression, while Decreasing CX43 Expression in Cardiomyocytes

Additionally, it is known that ROS plays a role in ventricular arrhythmogenesis by altering Cx43 between myocytes [21,27,28]. Cx43 decreased in a time-dependent manner within the left ventricle in cLA-treated mice (Figure 3A). As early as 24 h following treatment with cLA, Cx43 expression was significantly decreased compared to the control (Figure 3B). In contrast, NOX2 expression significantly increased following treatment with cLA (Figure 3A,B). Further, IHC linear staining of Cx43 was decreased 72 h following cLA treatment (Figure 4A,B). cLA-induced anti-tumorigenic effects have been linked to ROS generation and subsequent apoptosis in animals and cancer cell lines [29,30]. cLA isomers effectively inhibited the growth of cancer cells and this effect was associated with disruption of intercellular Cx43 [31]. Our results further demonstrate that cLA induces ROS within the myocardium within 72 h following treatment. Further, cLA reduces Cx43 expression, causing decreased survival in mice during MI.

3. Discussion

The principal findings of this study are that treatment with cLA disrupts connexin connections between myocytes leading to life-threatening arrhythmias, resulting in VT during MI. The treatment of mice with cLA 3 days prior to MI surgery resulted in a lower survival rate during MI compared to untreated MI mice. This was caused by VT compared to untreated MI mice (Figure 5). SCD can occur without structural heart disease and is attributed in most cases to electrical disturbances leading to abnormal heart rhythms. Cx43 is the most important protein in ventricular gap junctions and reduction in Cx43 results in SCD [32,33,34]. In the heart, myocyte-to-myocyte electrical coupling is mediated via gap junctions, forming the intercellular pathway for proper conductance of electrical excitation in synchronous contraction [16,35,36]. The regulation of Cx43 is under continuous investigation as it is still unclear whether the effects of Ca2+ are direct or due to the action of intracellular mediators.
Physiological concentrations of Ca2+ are known to regulate the permeability of Cx43 in a calmodulin-dependent manner. Recently it was shown that the lipolysis/lipogenesis balance in white adipocytes is also regulated through Ca2+ flux, and that its signaling is controlled through vesicular ATP release [37]. Further, the deuterated form of linoleic polyunsaturated fatty acid (D4-Lnn), a polyunsaturated fatty acid (PUFA), induced cytoprotective effects in an oxygen-glucose deprivation (OGD) ischemic-type model through the activation of the phosphoinositide calcium system [38]. The suppression of damaging proteins and activation of protective genes was also related to an increase in ROS, resulting in an overall inhibition of neuronal apoptosis [38]. Similarly, neuronal cells treated with deuterated PUFAs, completely prevented the ROS-induced effects of oligomeric α-synuclein on lipid peroxidation [39].
NOX2 expression in heart tissue has been shown to increase ROS generation and the incidence of arrhythmias [40]. To investigate ROS generation in myocytes, heart slices were stained with MitoTracker. Mice treated with cLA showed ROS generation within 72 h, as demonstrated by a significant increase in MitoTracker staining, compared to control mice (Figure 6A,B). The use of different NOX enzyme isoforms is known to result in different ROS, which highlights the complex role of NOX-derived ROS in in cardiac pathophysiology [41]. However, it has also been reported that NOX2 is a prominent ROS generator and leads to direct myocardial damage, compared to that of other isoforms [42]. Our results demonstrate that increased ROS production at the 72 h time point may be responsible for the disruption of connexins and the development of abnormal heart rhythm following treatment with cLA in MI. Overall, further investigation is warranted as to the consumption of cLA and the potential for increased an risk of VT and SCD with MI.

4. Materials and Methods

4.1. Animals

The mice were fed standard chow and water ad libitum. All animal procedures were reviewed and approved by the independent Institutional Animal Care and Use Committee (IACUC) of the University of Louisville, School of Medicine (IACUC protocol 14079). In addition, all studies were performed in accordance with the animal care and use guidelines of the National Institutes of Health.

4.2. Mouse Model of Myocardial Infarction

Male C57BL/6 mice, when 10–12 weeks old, were anesthetized with isoflurane, intubated, and ventilated with a CWE advanced ventilator (Webster, TX, USA). Body temperature was maintained with an Indus Temperature feedback/surgical table and ECG system. An aseptic procedure was used for the preparation of the surgical site through scrubbing with a 0.8% chlorhexidine solution. A left thoracotomy was performed via the fourth intercostal space and the lungs retracted to expose the heart. After opening the pericardium, the left anterior descending (LAD) coronary artery was ligated with an 8–0 silk suture near its origin between the pulmonary outflow tract and the edge of the atrium. Ligation was deemed successful when the anterior wall of the left ventricle (LV) turned pale. The lungs were inflated by increasing positive end-expiratory pressure, and the thoracotomy side was closed in layers. The lungs were re-expanded, and the chest was closed. The animals were removed from the ventilator and allowed to recover on a heating pad. Mice were checked daily for signs of pain or distress and were given Buprenex at 0.05 mg/kg SQ before the procedure and every 12 h for 48 h. Mice were treated with cLA (10 mg/kg/d-via osmotic mini-pump) 3 days prior to the ligation of the LAD artery.
Mice were treated with cLA (10 mg/kg/d-via osmotic mini-pump) for 12, 24, 48, or 72 h. Following treatment, mice were injected with 50 mg/kg pentobarbital and euthanized. Tissue was harvested for the following experiments.

4.3. Western Blot

Heart tissue homogenate (100 μg) was electrophoresed using the SDS-PAGE method as previously described [43,44]. Affinity-purified GAPDH (1:3000) (Trevigen, Gaithersburg, MD, USA), Nox2 (1:1000) (#ab129068-Abcam, Branford, CT, USA), and Cx-43 (1:1000) (#3512S-Cell Signaling, Danvers, MA, USA) antibodies were detected using species-appropriate horseradish peroxidase-labeled secondary antibodies.

4.4. Echocardiography

Transthoracic echocardiography was performed using a VisualSonics ultrasound system (Vevo 2100; VisualSonics, Inc., Toronto, ON, Canada). Left ventricular function was analyzed via the short parasternal axis view. Echocardiograms were performed before MI surgery.

4.5. Histology, Immunohistochemistry and Confocal Microscopy

Hearts were collected from mice and thoroughly washed in PBS. Using Peel-A-Way disposable plastic tissue embedding molds (Polysciense Inc, Washington, PA, USA) filled with tissue freezing media (Triangle Biomedical Sciences, Durham, NC, USA), hearts were preserved and stored at −80 °C until analysis. Tissue slices (5 µm in thickness) were made using the Leica CM 1850 Cryocut microtome (Bannockburn, IL, USA). Slices were placed on Superfrost™ Plus glass slides, air-dried, and processed for immunohistochemistry (IHC) staining. Immunohistochemistry and laser-scanning confocal microscopy were used to visualize cLA-induced changes in Cx43 and mitochondrial stress (MitoTracker) expression and localization.
Immunohistochemistry was performed on frozen tissue slices using a standard IHC protocol. Primary antibodies were applied overnight (anti-Cx43). Secondary antibodies labeled with Texas Red (Invitrogen, Carlsbad, CA, USA) were applied for protein immunodetection.
Mitochondrial superoxide generation was assessed by MitoTracker (Red CMXRos) staining, a fluorogenic dye that is taken up by mitochondria. MitoTracker Red CMXRos, tetramethylrhodamine methyl ester, and 10-N-nonyl acridine orange label mitochondria in a manner dependent on the membrane potential, thus giving an indication of mitochondrial stress. The MitoTracker probe, CMXRos Red, is taken up passively by cells. In the mitochondria, the probe is oxidized by superoxide, resulting in the emission of red fluorescence. Fresh frozen 5 μm LV slices were incubated for 45 min at 37 °C with 100 nM MitoTracker (Red CMXRos).
Stained slides were analyzed for fluorescence using a laser scanning confocal microscope (Olympus FluoView-1000, objective 40X, Melville, NY, USA) set at the appropriate filter settings (Tex-red for Cx-43, Cy-3 for MitoTracker). The total fluorescence (red) intensity in 5 random fields (for each experimental sample) was measured with image analysis software off-line (Image-Pro Plus 7.0, Media Cybernetics; Bethesda, MD, USA). The fluorescence intensity values for each experimental group were averaged and presented as fluorescent intensity units (FIU).

4.6. Statistical Analysis

Statistical analyses were performed with GraphPad Prism (version 5.0) Statistical Software, and the significance level was set at p < 0.05. Values are presented as mean ± SD. Echocardiography and Western blot measurements were analyzed using one-way analysis of variance (ANOVA), followed by Bonferroni’s multiple comparison post hoc analysis. Cx-43 and MitoTracker immunofluorescence intensity were analyzed using two-tailed t test with Welch’s correction for simple two group comparisons.

5. Conclusions

Our data demonstrate that cLA causes life-threatening arrhythmias in MI. Sudden arrhythmic death is a result of an abnormal heart rhythm. A significant increase in NOX2 expression following cLA treatment in MI results in ROS generation in cardiac tissue. Further, the disruption of Cx43 between myocytes ultimately causes life-threatening arrhythmias, VT, and death during MI. While cLA is generally regarded as safe, individuals susceptible to adverse cardiovascular events such as MI should use caution regarding cLA consumption.

Author Contributions

Conceptualization and methodology, M.P.C. and N.Q.K.; formal analysis, N.Q.K., J.C.S.; writing—original draft preparation, N.Q.K.; writing—review and editing, M.P.C., N.Q.K. and K.M.P.; funding acquisition, M.P.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Institutes of Health, grant number K99/R00HL95769 and The University of Louisville to M.P.C.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Review Board (IACUC protocol 14079, original approval December 2014) for studies involving animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors wish to thank Bradley Keller for the use of the VEVO 2100 to collect heart function data.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Banni, S. Conjugated linoleic acid metabolism. Curr. Opin. Lipidol. 2002, 13, 261–266. [Google Scholar] [CrossRef] [PubMed]
  2. Shen, W.; Chuang, C.-C.; Martinez, K.; Reid, T.; Brown, J.M.; Xi, L.; Hixson, L.; Hopkins, R.; Starnes, J.; McIntosh, M. Conjugated linoleic acid reduces adiposity and increases markers of browning and inflammation in white adipose tissue of mice. J. Lipid. Res 2013, 54, 909–922. [Google Scholar] [CrossRef] [Green Version]
  3. Cook, M.E.; Miller, C.C.; Park, Y.; Pariza, M.W. Immune Modulation by Altered Nutrient Metabolism: Nutritional Control of Immune-Induced Growth Depression. Poultry. Sci. 1993, 72, 1301–1305. [Google Scholar] [CrossRef]
  4. Miller, C.C.; Park, Y.; Pariza, M.W.; Cook, M.E. Feeding Conjugated Linoleic Acid to Animals Partially Overcomes Catabolic Responses Due to Endotoxin Injection. Biochem. Biophys. Res. Commun. 1994, 198, 1107–1112. [Google Scholar] [CrossRef] [PubMed]
  5. Wahle, K.W.J.; Heys, S.D.; Rotondo, D. Conjugated linoleic acids: Are they beneficial or detrimental to health? Prog. Lipid Res. 2004, 43, 553–587. [Google Scholar] [CrossRef] [PubMed]
  6. Lee, K.N.; Kritchevsky, D.; Parizaa, M.W. Conjugated linoleic acid and atherosclerosis in rabbits. Atheroscl 1994, 108, 19–25. [Google Scholar] [CrossRef]
  7. Toomey, S.; Harhen, B.; Roche, H.M.; Fitzgerald, D.; Belton, O. Profound resolution of early atherosclerosis with conjugated linoleic acid. Atheroscler 2006, 187, 40–49. [Google Scholar] [CrossRef]
  8. Mooney, D.; McCarthy, C.; Belton, O. Effects of conjugated linoleic acid isomers on monocyte, macrophage, and foam cell phenotype in atherosclerosis. Prostaglandins Other Lipid Mediat. 2012, 98, 56–62. [Google Scholar] [CrossRef]
  9. Houseknecht, K.L.; Heuvel, J.P.V.; Moya-Camarena, S.Y.; Portocarrero, C.P.; Peck, L.W.; Nickel, K.P.; Belury, M.A. Dietary Conjugated Linoleic Acid Normalizes Impaired Glucose Tolerance in the Zucker Diabetic Fattyfa/faRat. Biochem. Biophys. Res. Commun. 1998, 244, 678–682. [Google Scholar] [CrossRef]
  10. Belury, M.A.; Mahon, A.; Banni, S. The Conjugated Linoleic Acid (CLA) Isomer, t10c12-CLA, Is Inversely Associated with Changes in Body Weight and Serum Leptin in Subjects with Type 2 Diabetes Mellitus. J. Nutr. 2003, 133, 257S–260S. [Google Scholar] [CrossRef] [Green Version]
  11. Piell, K.M.; Qipshidze Kelm, N.; Caroway, M.P.; Aman, M.; Cole, M.P. Nitrite treatment rescues cardiac dysfunction in aged mice treated with conjugated linoleic acid. Free Rad. Biol. Med. 2014, 72, 66–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Zhou, J.Z.; Jiang, J.X. Gap junction and hemichannel-independent actions of connexins on cell and tissue functions—An update. FEBS Lett. 2014, 588, 1186–1192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Grippo, A.J.; Moffitt, J.A.; Henry, M.K.; Firkins, R.; Senkler, J.; McNeal, N.; Wardwell, J.; Scotti, M.-A.L.; Dotson, A.; Schultz, R. Altered Connexin 43 and Connexin 45 Protein Expression in the Heart as a Function of Social and Environmental Stress in the Prairie Vole. Stress 2015, 18, 107–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Tulenko, T.N. Regulating Cross-Talk Between Vascular Smooth Muscle Cells. Arterioscler Thromb. Vasc. Biol. 2003, 23, 1707–1709. [Google Scholar] [CrossRef] [Green Version]
  15. Eiberger, J.; Degen, J.; Romualdi, A.; Deutsch, U.; Willecke, K.; Sohl, G. Connexin genes in the mouse and human genome. Cell Commun Adhes 2001, 8, 163–165. [Google Scholar] [CrossRef]
  16. Severs, N.J.; Bruce, A.F.; Dupont, E.; Rothery, S. Remodeling of gap junctions and connexin expression in diseased myocardium. Cardiovasc. Res. 2008, 80, 9–19. [Google Scholar] [CrossRef] [Green Version]
  17. Lin, X.; Gemel, J.; Glass, A.; Zemlin, C.W.; Beyer, E.C.; Veenstra, R.D. Connexin40 and connexin43 determine gating properties of atrial gap junction channels. J. Mol. Cell Cardiol. 2010, 48, 238–245. [Google Scholar] [CrossRef] [Green Version]
  18. Bao, M.; Kanter, E.M.; Huang, R.Y.C.; Maxeiner, S.; Frank, M.; Zhang, Y.; Schuessler, R.B.; Smith, T.W.; Townsend, R.R.; Rohrs, H.W.; et al. Residual Cx45 and its relationship to Cx43 in murine ventricular myocardium. Channels 2011, 5, 489–499. [Google Scholar] [CrossRef] [Green Version]
  19. Severino, A.; Narducci, M.L.; Pedicino, D.; Pazzano, V.; Giglio, A.F.; Biasucci, L.M.; Liuzzo, G.; Casella, M.; Bartoletti, S.; Dello Russo, A.; et al. Reversible atrial gap junction remodeling during hypoxia/reoxygenation and ischemia: A possible arrhythmogenic substrate for atrial fibrillation. Gen. Physiol. Biophys. 2012, 31, 439–448. [Google Scholar] [CrossRef] [Green Version]
  20. Lerner, D.L.; Yamada, K.A.; Schuessler, R.B.; Saffitz, J.E. Accelerated Onset and Increased Incidence of Ventricular Arrhythmias Induced by Ischemia in Cx43-Deficient Mice. Circulation 2011, 101, 547–552. [Google Scholar] [CrossRef] [Green Version]
  21. Sovari, A.A.; Rutledge, C.A.; Jeong, E.-M.; Dolmatova, E.; Arasu, D.; Liu, H.; Vahdani, N.; Gu, L.; Zandieh, S.; Xiao, L.; et al. Mitochondria Oxidative Stress, Connexin43 Remodeling, and Sudden Arrhythmic Death. Circ. Arrhythm. Electrophysiol 2013, 6, 623–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Maraldi, T. Natural Compounds as Modulators of NADPH Oxidases. Oxid. Med. Cell. Longev. 2013, 2013, 271602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zhang, M.; Shah, A.M. ROS signaling between endothelial cells and cardiac cells. Cardiovasc. Res. 2014, 102, 249–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Bedard, K.; Krause, K.-H. The NOX Family of ROS-Generating NADPH Oxidases: Physiology and Pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef]
  25. Looi, Y.H.; Grieve, D.J.; Siva, A.; Walker, S.J.; Anilkumar, N.; Cave, A.C.; Marber, M.; Monaghan, M.J.; Shah, A.M. Involvement of Nox2 NADPH Oxidase in Adverse Cardiac Remodeling After Myocardial Infarction. Hypertension 2008, 51, 319–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Betsuyaku, T.; Kanno, S.; Lerner, D.L.; Schuessler, R.B.; Saffitz, J.E.; Yamada, K.A. Spontaneous and inducible ventricular arrhythmias after myocardial infarction in mice. Cardiovasc. Pathol. 2004, 13, 156–164. [Google Scholar] [CrossRef]
  27. Aikawa, R.; Komuro, I.; Yamazaki, T.; Zou, Y.; Kudoh, S.; Tanaka, M.; Shiojima, I.; Hiroi, Y.; Yazaki, Y. Oxidative stress activates extracellular signal-regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats. J. Clin. Invest. 1997, 100, 1813–1821. [Google Scholar] [CrossRef] [Green Version]
  28. Haendeler, J.; Hoffmann, J.; Brandes, R.P.; Zeiher, A.M.; Dimmeler, S. Hydrogen peroxide triggers nuclear export of telomerase reverse transcriptase via Src kinase family-dependent phosphorylation of tyrosine 707. Mol. Cell. Biol. 2003, 23, 4598–4610. [Google Scholar] [CrossRef] [Green Version]
  29. Islam, M.A.; Kim, Y.S.; Jang, W.J.; Lee, S.M.; Kim, H.G.; Kim, S.Y.; Kim, J.O.; Ha, Y.L. A mixture of trans, trans conjugated linoleic acid induces apoptosis in MCF-7 human breast cancer cells with reciprocal expression of Bax and Bcl-2. J. Agric. Food Chem. 2008, 56, 5970–5976. [Google Scholar] [CrossRef]
  30. Islam, M.A.; Kim, Y.S.; Oh, T.W.; Kim, G.S.; Won, C.K.; Kim, H.G.; Choi, M.S.; Kim, J.O.; Ha, Y.L. Superior anticarcinogenic activity of trans, trans-conjugated linoleic acid in N-methyl-N-nitrosourea-induced rat mammary tumorigenesis. J. Agric. Food. Chem. 2010, 58, 5670–5678. [Google Scholar] [CrossRef]
  31. Rakib, M.A.; Lee, W.S.; Kim, G.S.; Han, J.H.; Kim, J.O.; Ha, Y.L. Antiproliferative Action of Conjugated Linoleic Acid on Human MCF-7 Breast Cancer Cells Mediated by Enhancement of Gap Junctional Intercellular Communication through Inactivation of NF- kappa B. Evid. Based Complement Altern. Med. 2013, 2013, 429393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Gutstein, D.E.; Morley, G.E.; Tamaddon, H.; Vaidya, D.; Schneider, M.D.; Chen, J.; Chien, K.R.; Stuhlmann, H.; Fishman, G.I. Conduction Slowing and Sudden Arrhythmic Death in Mice with Cardiac-Restricted Inactivation of Connexin43. Circ. Res. 2001, 88, 333–339. [Google Scholar] [CrossRef]
  33. Gutstein, D.E.; Morley, G.E.; Vaidya, D.; Liu, F.; Chen, F.L.; Stuhlmann, H.; Fishman, G.I. Heterogeneous expression of Gap junction channels in the heart leads to conduction defects and ventricular dysfunction. Circulation 2001, 104, 1194–1199. [Google Scholar] [CrossRef] [Green Version]
  34. Van Norstrand, D.W.; Asimaki, A.; Rubinos, C.; Dolmatova, E.; Srinivas, M.; Tester, D.J.; Saffitz, J.E.; Duffy, H.S.; Ackerman, M.J. Connexin43 mutation causes heterogeneous gap junction loss and sudden infant death. Circulation 2012, 125, 474–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Söhl, G.; Willecke, K. Gap junctions and the connexin protein family. Cardiovasc. Res. 2004, 62, 228–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Severs, N.J.; Dupont, E.; Thomas, N.; Kaba, R.; Rothery, S.; Jain, R.; Sharpey, K.; Fry, C.H. Alterations in cardiac connexin expression in cardiomyopathies. Adv. Cardiol. 2006, 42, 228–242. [Google Scholar]
  37. Turovsky, E.A.; Varlamova, E.G.; Turovskaya, M.V. Activation of Cx43 hemichannels induces the generation of Ca2+ oscillations in white adipocytes and stimulates lipolysis. Int. J. Mol. Sci. 2021, 22, 8095–8123. [Google Scholar] [CrossRef]
  38. Turovsky, E.A.; Varlamova, E.G.; Gudkov, S.V.; Plotnikov, E.Y. The protective mechanism of deuterated linoleic acid involves the activation of the Ca2+ signaling system of astrocytes in ischemia in vitro. Int. J. Mol. Sci 2021, 22, 13216–13235. [Google Scholar] [CrossRef]
  39. Angelova, P.R.; Horrocks, M.H.; Klenerman, D.; Ganhi, S.; Abramov, A.Y.; Shchepinov, M.S. Lipid peroxidation is essential for α-synuclein-induced cell death. J. Neuro. Chem. 2015, 133, 582–589. [Google Scholar] [CrossRef] [Green Version]
  40. Kim, Y.M.; Guzik, T.J.; Zhang, Y.H.; Zhang, M.H.; Kattach, H.; Ratnatunga, C.; Pillai, R.; Channon, K.M.; Casadei, B. A myocardial Nox2 containing NAD(P)H oxidase contributes to oxidative stress in human atrial fibrillation. Circ. Res. 2005, 97, 629–636. [Google Scholar] [CrossRef]
  41. Braunersreuther, V.; Montecucco, F.; Asrih, M.; Pelli, G.; Galan, K.; Frias, M.; Burger, F.; Quindere, A.L.; Montessuit, C.; Krause, K.H.; et al. Role of NADPH oxidase isoforms NOX1, NOX2 and NOX4 in myocardial ischemia/reperfusion injury. J. Mol. Cell. Cardiol. 2013, 64, 99–107. [Google Scholar] [CrossRef] [PubMed]
  42. Maghzal, G.J.; Krause, K.H.; Stocker, R.; Jaquet, V. Detection of reactive oxygen species derived from the family of NOX NADPH oxidases. Free Radic. Biol. Med. 2012, 53, 1903–1918. [Google Scholar] [CrossRef] [PubMed]
  43. Cole, M.P.; Chaiswing, L.; Oberley, T.D.; Edelmann, S.E.; Piascik, M.T.; Lin, S.-M.; Kiningham, K.K.; St. Clair, D.K. The protective roles of nitric oxide and superoxide dismutase in adriamycin-induced cardiotoxicity. Cardiovasc. Res. 2006, 69, 186–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Combadière, C.; Raoul, W.; Guillonneau, X.; Sennlaub, F. Comment on “Ccl2, Cx3cr1 and Ccl2/Cx3cr1 chemokine deficiencies are not sufficient to cause age-related retinal degeneration” by Luhmann et al. (Exp. Eye Res. 2013; 107, 80. https://doi.org/10.1016). Exp. Eye. Res. 2013, 111, 134–135. [Google Scholar] [CrossRef] [Green Version]
Figure 1. cLA treatment induces ventricular fibrillation following MI. ECG data from control and cLA-treated mice (A) before MI, (B) 5 min after MI in control mice, and (C) 5 min after MI in cLA-treated mice demonstrate changes in ST segment elevation following LAD. MI surgery in untreated mice (n = 15); MI surgery in cLA-treated mice (n = 35).
Figure 1. cLA treatment induces ventricular fibrillation following MI. ECG data from control and cLA-treated mice (A) before MI, (B) 5 min after MI in control mice, and (C) 5 min after MI in cLA-treated mice demonstrate changes in ST segment elevation following LAD. MI surgery in untreated mice (n = 15); MI surgery in cLA-treated mice (n = 35).
Ijms 24 11208 g001
Figure 2. cLA treatment does not affect cardiac dysfunction after 72 h of treatment. Treatment with cLA does not affect cardiac function, FS in (A) control and (B) cLA-treated mice were 53 ± 3 and 51 ± 4, respectively. MI surgery in untreated mice (n = 15); MI surgery in cLA-treated mice (n = 35).
Figure 2. cLA treatment does not affect cardiac dysfunction after 72 h of treatment. Treatment with cLA does not affect cardiac function, FS in (A) control and (B) cLA-treated mice were 53 ± 3 and 51 ± 4, respectively. MI surgery in untreated mice (n = 15); MI surgery in cLA-treated mice (n = 35).
Ijms 24 11208 g002
Figure 3. cLA treatment alters Cx-43 level and increases NOX2 level in heart tissue. cLA decreases levels of Cx-43expression in time point treated mice (A). Conversely, it increases levels of NOX-expression (B). MI surgery in untreated mice (n = 15); MI surgery in cLA-treated mice (n = 35). * p < 0.05 vs. control, # p < 0.05 vs. cLA 12 h.
Figure 3. cLA treatment alters Cx-43 level and increases NOX2 level in heart tissue. cLA decreases levels of Cx-43expression in time point treated mice (A). Conversely, it increases levels of NOX-expression (B). MI surgery in untreated mice (n = 15); MI surgery in cLA-treated mice (n = 35). * p < 0.05 vs. control, # p < 0.05 vs. cLA 12 h.
Ijms 24 11208 g003
Figure 4. cLA exacerbates connexin-43 disruption in mice after 72 h of treatment. Cx-43 is stained in red, in control (A) Cx-43 is represents linear staining, which is disrupted in cLA-treated mice after 72 h of treatment (A). Quantitated protein expression reveals that cLA lowers CX-43 expression in mice treated with cLA after 72 h of treatment (B). MI surgery in untreated mice (n = 15); MI surgery in cLA-treated mice (n = 35).
Figure 4. cLA exacerbates connexin-43 disruption in mice after 72 h of treatment. Cx-43 is stained in red, in control (A) Cx-43 is represents linear staining, which is disrupted in cLA-treated mice after 72 h of treatment (A). Quantitated protein expression reveals that cLA lowers CX-43 expression in mice treated with cLA after 72 h of treatment (B). MI surgery in untreated mice (n = 15); MI surgery in cLA-treated mice (n = 35).
Ijms 24 11208 g004
Figure 5. cLA alters survival in mice during myocardial infarction (MI). cLA decreases survival during MI. * p < 0.05 vs. control, # p < 0.05 vs. MI. MI surgery in untreated mice results in a cumulative survival of 80%, however mice treated with cLA had a cumulative survival of 20% during MI surgery. Numbers on graph represent the number of animals analyzed in each group.
Figure 5. cLA alters survival in mice during myocardial infarction (MI). cLA decreases survival during MI. * p < 0.05 vs. control, # p < 0.05 vs. MI. MI surgery in untreated mice results in a cumulative survival of 80%, however mice treated with cLA had a cumulative survival of 20% during MI surgery. Numbers on graph represent the number of animals analyzed in each group.
Ijms 24 11208 g005
Figure 6. cLA treatment increases ROS generation after 72 h of treatment. Immunohistochemistry images demonstrate MitoTracker staining, which measures ROS generation, which was increased in treated mice with cLA after 72 h of treatment (A). Quantitated expression of MitoTracker (B).
Figure 6. cLA treatment increases ROS generation after 72 h of treatment. Immunohistochemistry images demonstrate MitoTracker staining, which measures ROS generation, which was increased in treated mice with cLA after 72 h of treatment (A). Quantitated expression of MitoTracker (B).
Ijms 24 11208 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kelm, N.Q.; Solinger, J.C.; Piell, K.M.; Cole, M.P. Conjugated Linoleic Acid-Mediated Connexin-43 Remodeling and Sudden Arrhythmic Death in Myocardial Infarction. Int. J. Mol. Sci. 2023, 24, 11208. https://doi.org/10.3390/ijms241311208

AMA Style

Kelm NQ, Solinger JC, Piell KM, Cole MP. Conjugated Linoleic Acid-Mediated Connexin-43 Remodeling and Sudden Arrhythmic Death in Myocardial Infarction. International Journal of Molecular Sciences. 2023; 24(13):11208. https://doi.org/10.3390/ijms241311208

Chicago/Turabian Style

Kelm, Natia Qipshidze, Jane C. Solinger, Kellianne M. Piell, and Marsha P. Cole. 2023. "Conjugated Linoleic Acid-Mediated Connexin-43 Remodeling and Sudden Arrhythmic Death in Myocardial Infarction" International Journal of Molecular Sciences 24, no. 13: 11208. https://doi.org/10.3390/ijms241311208

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop