The circadian clock maintains cardiac function by regulating mitochondrial metabolism in mice

Akira Kohsaka, Partha Das, Izumi Hashimoto, Tomomi Nakao, Yoko Deguchi, Sabine S Gouraud, Hidefumi Waki, Yasuteru Muragaki, Masanobu Maeda, Akira Kohsaka, Partha Das, Izumi Hashimoto, Tomomi Nakao, Yoko Deguchi, Sabine S Gouraud, Hidefumi Waki, Yasuteru Muragaki, Masanobu Maeda

Abstract

Cardiac function is highly dependent on oxidative energy, which is produced by mitochondrial respiration. Defects in mitochondrial function are associated with both structural and functional abnormalities in the heart. Here, we show that heart-specific ablation of the circadian clock gene Bmal1 results in cardiac mitochondrial defects that include morphological changes and functional abnormalities, such as reduced enzymatic activities within the respiratory complex. Mice without cardiac Bmal1 function show a significant decrease in the expression of genes associated with the fatty acid oxidative pathway, the tricarboxylic acid cycle, and the mitochondrial respiratory chain in the heart and develop severe progressive heart failure with age. Importantly, similar changes in gene expression related to mitochondrial oxidative metabolism are also observed in C57BL/6J mice subjected to chronic reversal of the light-dark cycle; thus, they show disrupted circadian rhythmicity. These findings indicate that the circadian clock system plays an important role in regulating mitochondrial metabolism and thereby maintains cardiac function.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. H-Bmal1 −/− mice develop progressive…
Figure 1. H-Bmal1 −/− mice develop progressive congestive heart failure with age.
(A) Representative gross morphology of hearts from 12-week-old control and H-Bmal1−/− mice. (B) Ratios of heart weight to body weight (HW/BW) at 12 weeks of age (n = 6 per group). (C) Low-power views after H&E staining of transverse sections from control and Bmal1−/− hearts at 12 weeks of age. (D) LV lateral wall thickness determined in histological images used in (C) (n = 5 per group). (E) Echocardiographic analysis in 12-week-old control and H-Bmal1−/− animals (n = 8–9 per group). LV internal diameter at diastole (LVIDd) and at systole (LVIDs) and fractional shortening (FS) are shown in bar graph format. (F) Transcript expression levels of ANP and BNP in control and H-Bmal1−/− mice at 12 weeks of age (n = 6 per group). (G) Kaplan-Meier survival curves of control and H-Bmal1−/− animals (n = 31 per group). (H) Low-power views (top panels, scale bar: 1 mm) and high-magnification views (bottom panels, scale bar: 25 µm) of Masson's trichrome staining of heart sections from 33-week-old control and H-Bmal1−/− mice. (I) High-magnification views of H&E staining of lung (top panels) and liver (bottom panels) sections from the same animals used in (H). Scale bar: 50 µm. Data are the mean ± SEM. *P<0.05, **P<0.01, unpaired two-tailed Student's t-test.
Figure 2. Gene expression profiles of cardiac…
Figure 2. Gene expression profiles of cardiac energy metabolism in H-Bmal1 −/− mice.
(A) Heat-map representations of several classes of metabolic genes detected in the expression analysis of Bmal1−/− hearts from 12-week-old animals. (B-D) Relative expression levels of representative genes regulating (B) fatty acid metabolism, (C) glycolysis/TCA cycle, and (D) ETC/OXPHOS are shown (n = 6 per group). Data are the mean ± SEM. *P<0.05, **P<0.01, unpaired two-tailed Student's t-test (B-D).
Figure 3. Mitochondrial abnormalities in the hearts…
Figure 3. Mitochondrial abnormalities in the hearts of 12-week-old H-Bmal1 −/− mice.
(A) A heat-map representing the expression profiles of genes regulating mitochondrial structure and function in Bmal1−/− hearts. (B) Relative expression levels of genes associated with mitochondrial biogenesis, dynamics, and membrane potential in the heart tissue of control and H-Bmal1−/− animals (n = 6 per group). (C) Representative electron micrographs of sections taken from the left ventricular muscle from control and H-Bmal1−/− mice at two different magnifications. (D) Mitochondrial protein concentration in control and Bmal1−/− hearts (n = 8 per group). (E) Mitochondrial DNA to nuclear DNA ratio in control and Bmal1−/− hearts (n = 6 per group). (F-G) Enzymatic activities of (F) complex I and (G) complex IV in control and Bmal1−/− hearts (n = 8 per group). The activities of the mitochondrial respiratory enzymes are expressed either per milligram of tissue used for mitochondrial isolation (top panels) or per microgram of mitochondrial protein (bottom panels). (H) NAD+ and NADH concentrations in control and Bmal1−/− hearts (n = 6 per group). Data are the mean ± SEM. *P<0.05, **P<0.01, unpaired two-tailed Student's t-test.
Figure 4. Expression profiles of genes associated…
Figure 4. Expression profiles of genes associated with pathologic ventricular remodeling in Bmal1 −/− hearts.
(A-D) Heat-map representations of genes involved in (A) the response to oxidative stress, (B) programmed cell death, (C) inflammation, and (D) fibrosis in hearts from 12-week-old Bmal1−/− mice.
Figure 5. Circadian desynchronization reduces cardiac function…
Figure 5. Circadian desynchronization reduces cardiac function in C57BL/6J mice with drug-induced cardiomyopathy.
(A) A light-dark (LD) cycle regimen used to examine the effects of a variable LD schedule on cardiac function. C57BL/6J mice were either maintained on a constant LD schedule (fixed LD cycle group) or were subjected to a 12-h phase shift in LD cycle every 3 days (disrupted LD cycle group). To compare the effects of the LD schedule between animals with healthy hearts and those with cardiomyopathy, either normal saline (NS) or phenylephrine (PE) was continuously infused via an osmotic pump in each LD cycle group. (B) Locomotor activity records of NS-infused (top panel) and PE-infused (bottom panel) mice. Two representative records from animals subjected to the fixed (left column) and disrupted (right column) LD cycle are shown. Activity counts are indicated by the vertical black marks. The records are double-plotted such that 48 h are shown for each horizontal trace. The blue shaded and unshaded areas indicate the dark and light period, respectively. The duration of NS or PE infusion is indicated by the vertical line at the right margin. (C) The ratios of heart weight to body weight (HW/BW) were increased by the PE infusion but were not influenced by the disruption in LD cycle (n = 5–8 per group). (D) The effects of PE, disrupted LD cycle, or both on ventricular function were evaluated using echocardiographic measurements (n = 5–8 per group). LVIDd, LVIDs, and FS are shown in bar graph format. Data are the mean ± SEM. *P<0.05, **P<0.01, two-way ANOVA.
Figure 6. Circadian desynchronization not only disrupts…
Figure 6. Circadian desynchronization not only disrupts rhythms but also reduces the expression levels of clock and metabolic genes in the heart of C57BL/6J mice with PE-induced cardiomyopathy.
(A-C) Relative expression levels of genes regulating (A) clock machinery as well as (B) glucose and (C) lipid metabolism in heart. All heart tissues used were from PE-infused animals subjected to either a fixed or a disrupted LD cycle as described in Figure 5A (n = 4 per group per time point). To provide a 24-h overall mean expression level, the data over a 24-h time period in each group were also averaged and are expressed using a bar graph format. Data are the mean ± SEM. *P<0.05, **P<0.01, unpaired two-tailed Student's t-test.
Figure 7. Circadian desynchronization impairs mitochondrial function…
Figure 7. Circadian desynchronization impairs mitochondrial function in the hearts of C57BL/6J mice with PE-induced cardiomyopathy.
(A-B) Relative expression levels of genes regulating (A) mitochondrial structure or (B) mitochondrial oxidative metabolism in heart. All heart tissues used are from PE-infused animals subjected to either a fixed or a disrupted LD cycle as described in Figure 5A (n = 4 per group per time point). To provide a 24-h overall mean expression level, the data over a 24-h time period in each group were also averaged and are expressed in bar graph format. (C) Enzymatic activity of complex I in PE-infused animals exposed to a fixed or disrupted LD cycle (n = 4–5 per group). The complex I activity is expressed per milligram of tissue used for mitochondrial isolation. (D) The relative number of mitochondria in the left ventricular muscle was counted using electron microscope images. Representative images are shown. Data are the mean ± SEM. *P<0.05, **P<0.01, unpaired two-tailed Student's t-test.

References

    1. Bray MS, Shaw CA, Moore MW, Garcia RA, Zanquetta MM, et al. (2008) Disruption of the circadian clock within the cardiomyocyte influences myocardial contractile function, metabolism, and gene expression. Am J Physiol Heart Circ Physiol 294: H1036–1047.
    1. Tsai JY, Kienesberger PC, Pulinilkunnil T, Sailors MH, Durgan DJ, et al. (2010) Direct regulation of myocardial triglyceride metabolism by the cardiomyocyte circadian clock. J Biol Chem 285: 2918–2929.
    1. Lefta M, Campbell KS, Feng HZ, Jin JP, Esser KA (2012) Development of dilated cardiomyopathy in Bmal1-deficient mice. Am J Physiol Heart Circ Physiol 303: H475–485.
    1. Arjona A, Silver AC, Walker WE, Fikrig E (2012) Immunity's fourth dimension: approaching the circadian-immune connection. Trends Immunol 33: 607–612.
    1. Mavroudis PD, Scheff JD, Calvano SE, Androulakis IP (2013) Systems biology of circadian-immune interactions. J Innate Immun 5: 153–162.
    1. Pacha J, Sumova A (2013) Circadian regulation of epithelial functions in the intestine. Acta Physiol (Oxf) 208: 11–24.
    1. Hussain MM, Pan X (2009) Clock genes, intestinal transport and plasma lipid homeostasis. Trends Endocrinol Metab 20: 177–185.
    1. Firsov D, Bonny O (2010) Circadian regulation of renal function. Kidney Int 78: 640–645.
    1. Stow LR, Gumz ML (2011) The circadian clock in the kidney. J Am Soc Nephrol 22: 598–604.
    1. Lamia KA, Storch KF, Weitz CJ (2008) Physiological significance of a peripheral tissue circadian clock. Proc Natl Acad Sci U S A 105: 15172–15177.
    1. Rudic RD, McNamara P, Curtis AM, Boston RC, Panda S, et al. (2004) BMAL1 and CLOCK, two essential components of the circadian clock, are involved in glucose homeostasis. PLoS Biol 2: e377.
    1. Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, et al. (2010) Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466: 627–631.
    1. Shimba S, Ishii N, Ohta Y, Ohno T, Watabe Y, et al. (2005) Brain and muscle Arnt-like protein-1 (BMAL1), a component of the molecular clock, regulates adipogenesis. Proc Natl Acad Sci U S A 102: 12071–12076.
    1. Grimaldi B, Bellet MM, Katada S, Astarita G, Hirayama J, et al. (2010) PER2 controls lipid metabolism by direct regulation of PPARgamma. Cell Metab 12: 509–520.
    1. Rutter J, Reick M, Wu LC, McKnight SL (2001) Regulation of clock and NPAS2 DNA binding by the redox state of NAD cofactors. Science 293: 510–514.
    1. Musiek ES, Lim MM, Yang G, Bauer AQ, Qi L, et al. (2013) Circadian clock proteins regulate neuronal redox homeostasis and neurodegeneration. J Clin Invest 123: 5389–5400.
    1. Wang TA, Yu YV, Govindaiah G, Ye X, Artinian L, et al. (2012) Circadian rhythm of redox state regulates excitability in suprachiasmatic nucleus neurons. Science 337: 839–842.
    1. Woldt E, Sebti Y, Solt LA, Duhem C, Lancel S, et al. (2013) Rev-erb-alpha modulates skeletal muscle oxidative capacity by regulating mitochondrial biogenesis and autophagy. Nat Med 19: 1039–1046.
    1. Peek CB, Affinati AH, Ramsey KM, Kuo HY, Yu W, et al. (2013) Circadian clock NAD+ cycle drives mitochondrial oxidative metabolism in mice. Science 342: 1243417.
    1. Mohawk JA, Green CB, Takahashi JS (2012) Central and peripheral circadian clocks in mammals. Annu Rev Neurosci 35: 445–462.
    1. Dibner C, Schibler U, Albrecht U (2010) The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu Rev Physiol 72: 517–549.
    1. Marin-Garcia J, Akhmedov AT, Moe GW (2013) Mitochondria in heart failure: the emerging role of mitochondrial dynamics. Heart Fail Rev 18: 439–456.
    1. Marin-Garcia J (2005) Mitochondria and the Heart. New York: Springer Science+Business Media, Inc.
    1. Leone TC, Kelly DP (2011) Transcriptional control of cardiac fuel metabolism and mitochondrial function. Cold Spring Harb Symp Quant Biol 76: 175–182.
    1. Scarpulla RC, Vega RB, Kelly DP (2012) Transcriptional integration of mitochondrial biogenesis. Trends Endocrinol Metab 23: 459–466.
    1. Scarpulla RC (2008) Transcriptional paradigms in mammalian mitochondrial biogenesis and function. Physiol Rev 88: 611–638.
    1. Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, et al. (2002) The cardiac phenotype induced by PPARalpha overexpression mimics that caused by diabetes mellitus. J Clin Invest 109: 121–130.
    1. Smeets PJ, Teunissen BE, Willemsen PH, van Nieuwenhoven FA, Brouns AE, et al. (2008) Cardiac hypertrophy is enhanced in PPAR alpha-/- mice in response to chronic pressure overload. Cardiovasc Res 78: 79–89.
    1. Huss JM, Imahashi K, Dufour CR, Weinheimer CJ, Courtois M, et al. (2007) The nuclear receptor ERRalpha is required for the bioenergetic and functional adaptation to cardiac pressure overload. Cell Metab 6: 25–37.
    1. Alaynick WA, Kondo RP, Xie W, He W, Dufour CR, et al. (2007) ERRgamma directs and maintains the transition to oxidative metabolism in the postnatal heart. Cell Metab 6: 13–24.
    1. Arany Z, He H, Lin J, Hoyer K, Handschin C, et al. (2005) Transcriptional coactivator PGC-1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metab 1: 259–271.
    1. Liu C, Li S, Liu T, Borjigin J, Lin JD (2007) Transcriptional coactivator PGC-1alpha integrates the mammalian clock and energy metabolism. Nature 447: 477–481.
    1. Canaple L, Rambaud J, Dkhissi-Benyahya O, Rayet B, Tan NS, et al. (2006) Reciprocal regulation of brain and muscle Arnt-like protein 1 and peroxisome proliferator-activated receptor alpha defines a novel positive feedback loop in the rodent liver circadian clock. Mol Endocrinol 20: 1715–1727.
    1. Oishi K, Shirai H, Ishida N (2005) CLOCK is involved in the circadian transactivation of peroxisome-proliferator-activated receptor alpha (PPARalpha) in mice. Biochem J 386: 575–581.
    1. Durgan DJ, Tsai JY, Grenett MH, Pat BM, Ratcliffe WF, et al. (2011) Evidence suggesting that the cardiomyocyte circadian clock modulates responsiveness of the heart to hypertrophic stimuli in mice. Chronobiol Int 28: 187–203.
    1. Storch KF, Lipan O, Leykin I, Viswanathan N, Davis FC, et al. (2002) Extensive and divergent circadian gene expression in liver and heart. Nature 417: 78–83.
    1. Penev PD, Kolker DE, Zee PC, Turek FW (1998) Chronic circadian desynchronization decreases the survival of animals with cardiomyopathic heart disease. Am J Physiol 275: H2334–2337.
    1. Vyas MV, Garg AX, Iansavichus AV, Costella J, Donner A, et al. (2012) Shift work and vascular events: systematic review and meta-analysis. BMJ 345: e4800.
    1. Curtis AM, Cheng Y, Kapoor S, Reilly D, Price TS, et al. (2007) Circadian variation of blood pressure and the vascular response to asynchronous stress. Proc Natl Acad Sci U S A 104: 3450–3455.
    1. Jeyaraj D, Haldar SM, Wan X, McCauley MD, Ripperger JA, et al. (2012) Circadian rhythms govern cardiac repolarization and arrhythmogenesis. Nature 483: 96–99.
    1. Durgan DJ, Trexler NA, Egbejimi O, McElfresh TA, Suk HY, et al. (2006) The circadian clock within the cardiomyocyte is essential for responsiveness of the heart to fatty acids. J Biol Chem 281: 24254–24269.
    1. Asher G, Schibler U (2011) Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab 13: 125–137.
    1. Bass J, Takahashi JS (2010) Circadian integration of metabolism and energetics. Science 330: 1349–1354.
    1. Sahar S, Sassone-Corsi P (2012) Regulation of metabolism: the circadian clock dictates the time. Trends Endocrinol Metab 23: 1–8.
    1. Peek CB, Ramsey KM, Marcheva B, Bass J (2012) Nutrient sensing and the circadian clock. Trends Endocrinol Metab 23: 312–318.
    1. Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, et al. (2005) Obesity and metabolic syndrome in circadian Clock mutant mice. Science 308: 1043–1045.
    1. Madrazo JA, Kelly DP (2008) The PPAR trio: regulators of myocardial energy metabolism in health and disease. J Mol Cell Cardiol 44: 968–975.
    1. Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, et al. (2000) Peroxisome proliferator-activated receptor gamma coactivator-1 promotes cardiac mitochondrial biogenesis. J Clin Invest 106: 847–856.
    1. Asher G, Reinke H, Altmeyer M, Gutierrez-Arcelus M, Hottiger MO, et al. (2010) Poly(ADP-ribose) polymerase 1 participates in the phase entrainment of circadian clocks to feeding. Cell 142: 943–953.
    1. Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, et al. (2009) Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science 324: 651–654.
    1. Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P (2009) Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science 324: 654–657.
    1. Saeed AI, Sharov V, White J, Li J, Liang W, et al. (2003) TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34: 374–378.

Source: PubMed

3
구독하다