NDUFAB1 confers cardio-protection by enhancing mitochondrial bioenergetics through coordination of respiratory complex and supercomplex assembly

Tingting Hou, Rufeng Zhang, Chongshu Jian, Wanqiu Ding, Yanru Wang, Shukuan Ling, Qi Ma, Xinli Hu, Heping Cheng, Xianhua Wang, Tingting Hou, Rufeng Zhang, Chongshu Jian, Wanqiu Ding, Yanru Wang, Shukuan Ling, Qi Ma, Xinli Hu, Heping Cheng, Xianhua Wang

Abstract

The impairment of mitochondrial bioenergetics, often coupled with exaggerated reactive oxygen species (ROS) production, is a fundamental disease mechanism in organs with a high demand for energy, including the heart. Building a more robust and safer cellular powerhouse holds the promise for protecting these organs in stressful conditions. Here, we demonstrate that NADH:ubiquinone oxidoreductase subunit AB1 (NDUFAB1), also known as mitochondrial acyl carrier protein, acts as a powerful cardio-protector by conferring greater capacity and efficiency of mitochondrial energy metabolism. In particular, NDUFAB1 not only serves as a complex I subunit, but also coordinates the assembly of respiratory complexes I, II, and III, and supercomplexes, through regulating iron-sulfur biosynthesis and complex I subunit stability. Cardiac-specific deletion of Ndufab1 in mice caused defective bioenergetics and elevated ROS levels, leading to progressive dilated cardiomyopathy and eventual heart failure and sudden death. Overexpression of Ndufab1 effectively enhanced mitochondrial bioenergetics while limiting ROS production and protected the heart against ischemia-reperfusion injury. Together, our findings identify that NDUFAB1 is a crucial regulator of mitochondrial energy and ROS metabolism through coordinating the assembly of respiratory complexes and supercomplexes, and thus provide a potential therapeutic target for the prevention and treatment of heart failure.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Cardiac-specific knockout of Ndufab1 (cKO) causes dilated cardiomyopathy leading to heart failure in mice. a Western blots of NDUFAB1 in wild-type (WT) and cKO cardiomyocytes. ATPB served as the loading control. b Left, photograph of WT and cKO mice at 14 weeks of age. Right, effect of cardiac NDUFAB1 ablation on mouse body weight (mean ± s.e.m.; n = 3–16 male mice per group per time point; **p < 0.01 versus WT at the same time point). c Kaplan–Meier survival curves for WT and cKO mice (n = 11 male mice per group; **p < 0.01 versus WT). d Representative longitudinal sections of hearts from male mice at 6–16 weeks (w) old. Scale bar, 5 mm. e Ratios of heart weight (HW) to body weight (BW) or tibia length (TL) (mean ± s.e.m.; n = 3–14 male mice per group; *p < 0.05, **p < 0.01 versus WT at the same age). f Progressive hypertrophy of cardiomyocytes from cKO mice. Left: representative confocal micrographs of cardiomyocytes stained with DCF (scale bar, 20 μm). Right: statistical analysis (mean ± s.e.m.; n = 97–611 cells from 4 male mice per group; ** p < 0.01 versus WT at the same age). g Fibrosis of left ventricular tissue (Masson’s trichrome staining; scale bar, 100 μm). h Echocardiographic analysis of cardiac function. EF, ejection fraction; FS, fractional shortening (mean ± s.e.m.; n = 3–9 male mice per group; **p < 0.01 versus WT at the same age)
Fig. 2
Fig. 2
Defective mitochondrial bioenergetics and ROS metabolism in Ndufab1 cKO mice. a Electron micrographs of mitochondria in left ventricular tissue from 6- and 16-week-old male mice (scale bars, 1 μm). b Decline of ΔΨm in cKO myocardium assessed by TMRM staining and confocal imaging of isolated cardiomyocytes. The intensity of FCCP-sensitive TMRM fluorescence was calculated and the intensity in cKO group was normalized to that of WT at the same age (n = 13–26 cells from three male mice per group; *p < 0.05, **p < 0.01 versus WT). c Progressive elevation of mitochondrial ROS measured with mitoSOX in isolated cardiomyocytes. Antimycin A (AA, 5 μg/mL) was used to induce massive ROS production. The intensities of mitoSOX fluorescence in the groups of cKO, WT + AA and cKO + AA were normalized to that of WT at the same age (n= 31–179 cells from 4–5 male mice per group; ** p < 0.01 versus WT). d ATP levels of isolated cardiomyocytes measured with luciferin assay (n = 3–4 male mice per group; ** p < 0.01 versus WT). e Top 5 enriched Gene Ontology terms of downregulated and upregulated genes. RNA-seq analysis was performed on cKO and WT hearts at 8–16 weeks (n = 4 male mice per group) and the genes upregulated or downregulated were analyzed
Fig. 3
Fig. 3
Defective respiration and impaired assembly of ETC complexes I-III and SCs in cKO hearts. a Effect of NDUFAB1 ablation on state III oxygen consumption rate (OCR) in isolated mitochondria at the age of 6 or 10 weeks. For each measurement, 100 μg mitochondria were used. Different respiratory substrates were used: malate/glutamate (Mala/glu, 5 mM) for complex I, succinate (Succ, 5 mM) for complex II, glycerol-3-phosphate (G3P, 5 mM) for complex III, and ascorbate (Asc, 2.5 mM)/N,N,N’,N’-tetramethyl-p-phenylenediamine (TMPD, 0.5 mM) for complex IV, and ADP (100 μM) (mean ± s.e.m.; n = 3–14 male mice of per group; *p < 0.05, **p < 0.01 versus WT at the same age). b In-gel activity of SCs and complex I at the age of 6 or 16 weeks. c BN-PAGE immunoblots of individual ETC complexes and SCs at the age of 6 or 16 weeks. SCs were visualized by antibodies against subunits of complex I (CI, NDUFB8), complex III (CIII, UQCRFS1 or UQCRC1), and complex IV (CIV, COX IV). Complex II was visualized by antibody against SDHA, and complex V with antibody against ATPB. d–f Western blots of subunits of complex I (d), complex II (e), and complex III (f) at the age of 6 or 16 weeks. Dagger represents FeS-containing subunits. ATP5A served as the loading control
Fig. 4
Fig. 4
Enhanced mitochondrial bioenergetics and SC assembly in the heart of Ndufab1 transgenic mice (TG). TG and WT littermates of 2–4 months old were used. a Western blots for NDUFAB1 expression in WT and TG hearts. ATPB served as the loading control. b Decreased mitochondrial ROS level in TG cardiomyocytes. The intensity of mitoSOX fluorescence of TG was normalized to that of WT (n= 34–41 cells from 3 male mice; **p < 0.01 versus WT). c Increased ΔΨm in TG cardiomyocytes. The intensity of FCCP-sensitive TMRM fluorescence of TG was normalized to that of WT (n= 12–15 cells from three male mice per group; *p < 0.05 versus WT). d, e Whole-cell OCR measured with Seahorse using glucose/pyruvate (d) or palmitate (e) as the substrate. Arrows indicate the time of adding palmitate (PAL, 200 µM), oligomycin (Oligo, 1 µM), FCCP (0.5 µM), and rotenone/antimycin A (Rot/AA, 1 µM for each). f, g Statistical analysis of maximal OCR in d and e (mean ± s.e.m.; n= 10–13 wells from 3 male mice per group; *p < 0.05 versus WT). h Changes of respiratory control ratio (RCR) with different substrates in isolated mitochondria (mean ± s.e.m.; n= 3–8 male mice per group; *p < 0.05, **p < 0.01 versus WT). i In-gel activity of SCs and complex I. j Statistical analysis of i. The activity of TG was normalized to WT (mean ± s.e.m.; n = 5 male mice per group; *p < 0.05, **p < 0.01 versus WT). k BN-PAGE immunoblots of SCs and individual ETC complexes as in Fig. 3c. l Statistical analysis of k. The expression level of TG complexes and supercomplexes was normalized to that of WT (mean ± s.e.m.; n= 4–8 male mice per group; *p < 0.05 versus WT)
Fig. 5
Fig. 5
NDUFAB1 overexpression protects the heart against IR injury. TG and WT male littermates of 2–4 months old were used. a Alleviated myocardial ROS production in TG hearts. Confocal images of DCF-stained myocardium after Langendorff perfusion and IR injury. Ischemia (IS) was mimicked by stopping perfusion. See Supplementary information, Fig. S19a for experimental protocol. Scale bars, 20 μm. b Statistical analysis of a (mean ± s.e.m.; n= 32–51 image frames from 4–7 male mice per group; **p < 0.01 versus WT, #p < 0.01 after IR versus basal). c Rotenone (Rot)-sensitive H2O2 production at complex I in WT and TG cardiac mitochondria (mean ± s.e.m.; n= 6 male mice per group; **p < 0.01 versus WT). Succinate (Succ, 5 mM) was used as the substrate of complex II and H2O2 was measured with Amplex red. d Images for simultaneous measurement of ∆Ψm (TMRM, pseudocolor: red) and cardiomyocyte death (EBD uptake, pseudocolor: green) in Langendorff-perfused hearts subjected to IR injury. Note that all cells with intact ∆Ψm are EBD-negative, and all EBD-positive cells display loss of ∆Ψm, but not vice versa. Scale bars, 50 μm. e, f Percentages of cells with loss of ∆Ψm (e) and EBD-positive cells (f) (mean ± s.e.m.; n = 9–10 male mice per group; *p < 0.05 versus WT, #p < 0.01 after IR versus basal). g Representative cross-sections of WT and TG hearts after IR injury. See Supplementary information, Fig. S19b for the experimental protocol. White, infarcted area (IF); red, rest of the area at risk (AAR); blue, tissue not at risk. The AAR was demarcated by dotted line. Scale bar, 1 mm. h, i Statistical analysis of AAR (h) and IF (i) reported as the ratios of AAR to left ventricular area (LV) and IF to AAR (n= 5–8 male mice per group; **p < 0.01 versus WT). j Measurement of serum LDH release after IR (n= 10–13 male mice per group; *p < 0.05 versus WT, #p < 0.01 after IR versus basal). k Echocardiographic analysis of cardiac function 48 h after IR injury. EF, ejection fraction; FS, fractional shortening (mean ± s.e.m.; n = 4 male mice per group; *p < 0.05 versus WT)

References

    1. Neubauer S. The failing heart–an engine out of fuel. N. Engl. J. Med. 2007;356:1140–1151. doi: 10.1056/NEJMra063052.
    1. Ingwall JS. ATP and the Heart. Norwell, MA: Kluwer Academic Publishers; 2002.
    1. Droge W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002;82:47–95. doi: 10.1152/physrev.00018.2001.
    1. Beckman KB, Ames BN. The free radical theory of aging matures. Physiol. Rev. 1998;78:547–581. doi: 10.1152/physrev.1998.78.2.547.
    1. DiMauro S, Schon EA. Mitochondrial respiratory-chain diseases. N. Engl. J. Med. 2003;348:2656–2668. doi: 10.1056/NEJMra022567.
    1. Ingwall JS. Transgenesis and cardiac energetics: new insights into cardiac metabolism. J. Mol. Cell Cardiol. 2004;37:613–623. doi: 10.1016/j.yjmcc.2004.05.020.
    1. Maack C, O’Rourke B. Excitation-contraction coupling and mitochondrial energetics. Basic Res. Cardiol. 2007;102:369–392. doi: 10.1007/s00395-007-0666-z.
    1. Nicholls, D. G. & Ferguson, S. L. Bioenergetics. (Academic Press, London, 2002).
    1. Wu M, et al. Structure of mammalian respiratory supercomplex I1III2IV1. Cell. 2016;167:1598–1609 e1510. doi: 10.1016/j.cell.2016.11.012.
    1. Sousa, J. S., Mills, D. J., Vonck, J. & Kuhlbrandt, W. Functional asymmetry and electron flow in the bovine respirasome. Elife5, 10.7554/eLife.21290 (2016).
    1. Letts JA, Fiedorczuk K, Sazanov LA. The architecture of respiratory supercomplexes. Nature. 2016;537:644–648. doi: 10.1038/nature19774.
    1. Gu J, et al. The architecture of the mammalian respirasome. Nature. 2016;537:639–643. doi: 10.1038/nature19359.
    1. Guo R, et al. Architecture of human mitochondrial respiratory megacomplex I2III2IV2. Cell. 2017;170:1247–1257. doi: 10.1016/j.cell.2017.07.050.
    1. Gong Hongri, Li Jun, Xu Ao, Tang Yanting, Ji Wenxin, Gao Ruogu, Wang Shuhui, Yu Lu, Tian Changlin, Li Jingwen, Yen Hsin-Yung, Man Lam Sin, Shui Guanghou, Yang Xiuna, Sun Yuna, Li Xuemei, Jia Minze, Yang Cheng, Jiang Biao, Lou Zhiyong, Robinson Carol V., Wong Luet-Lok, Guddat Luke W., Sun Fei, Wang Quan, Rao Zihe. An electron transfer path connects subunits of a mycobacterial respiratory supercomplex. Science. 2018;362(6418):eaat8923. doi: 10.1126/science.aat8923.
    1. Porras CA, Bai Y. Respiratory supercomplexes: plasticity and implications. Front. Biosci. 2015;20:621–634. doi: 10.2741/4327.
    1. Milenkovic D, Blaza JN, Larsson NG, Hirst J. The enigma of the respiratory chain supercomplex. Cell Metab. 2017;25:765–776. doi: 10.1016/j.cmet.2017.03.009.
    1. Maranzana E, et al. Mitochondrial respiratory supercomplex association limits production of reactive oxygen species from complex I. Antioxid. Redox Signal. 2013;19:1469–1480. doi: 10.1089/ars.2012.4845.
    1. Lopez-Fabuel I, et al. Complex I assembly into supercomplexes determines differential mitochondrial ROS production in neurons and astrocytes. Proc. Natl Acad. Sci. USA. 2016;113:13063–13068. doi: 10.1073/pnas.1613701113.
    1. Schagger H, et al. Significance of respirasomes for the assembly/stability of human respiratory chain complex I. J. Biol. Chem. 2004;279:36349–36353. doi: 10.1074/jbc.M404033200.
    1. Acin-Perez R, et al. Respiratory complex III is required to maintain complex I in mammalian mitochondria. Mol. cell. 2004;13:805–815. doi: 10.1016/S1097-2765(04)00124-8.
    1. Diaz F, Fukui H, Garcia S, Moraes CT. Cytochrome c oxidase is required for the assembly/stability of respiratory complex I in mouse fibroblasts. Mol. Cell. Biol. 2006;26:4872–4881. doi: 10.1128/MCB.01767-05.
    1. Lapuente-Brun E, et al. Supercomplex assembly determines electron flux in the mitochondrial electron transport chain. Science. 2013;340:1567–1570. doi: 10.1126/science.1230381.
    1. Guaras A, et al. The CoQH2/CoQ ratio serves as a sensor of respiratory chain efficiency. Cell Rep. 2016;15:197–209. doi: 10.1016/j.celrep.2016.03.009.
    1. Acin-Perez R, Enriquez JA. The function of the respiratory supercomplexes: the plasticity model. Biochim. Biophys. Acta. 2014;1837:444–450. doi: 10.1016/j.bbabio.2013.12.009.
    1. Greggio C, et al. Enhanced respiratory chain supercomplex formation in response to exercise in human skeletal muscle. Cell Metab. 2017;25:301–311. doi: 10.1016/j.cmet.2016.11.004.
    1. Rosca MG, et al. Cardiac mitochondria in heart failure: decrease in respirasomes and oxidative phosphorylation. Cardiovasc. Res. 2008;80:30–39. doi: 10.1093/cvr/cvn184.
    1. Runswick MJ, Fearnley IM, Skehel JM, Walker JE. Presence of an acyl carrier protein in NADH:ubiquinone oxidoreductase from bovine heart mitochondria. FEBS Lett. 1991;286:121–124. doi: 10.1016/0014-5793(91)80955-3.
    1. Hiltunen JK, et al. Mitochondrial fatty acid synthesis–an adopted set of enzymes making a pathway of major importance for the cellular metabolism. Prog. Lipid Res. 2010;49:27–45. doi: 10.1016/j.plipres.2009.08.001.
    1. Feng D, Witkowski A, Smith S. Down-regulation of mitochondrial acyl carrier protein in mammalian cells compromises protein lipoylation and respiratory complex I and results in cell death. J. Biol. Chem. 2009;284:11436–11445. doi: 10.1074/jbc.M806991200.
    1. Vinothkumar KR, Zhu J, Hirst J. Architecture of mammalian respiratory complex I. Nature. 2014;515:80–84. doi: 10.1038/nature13686.
    1. Floyd BJ, et al. Mitochondrial protein interaction mapping identifies regulators of respiratory chain function. Mol. cell. 2016;63:621–632. doi: 10.1016/j.molcel.2016.06.033.
    1. Van Vranken, J. G., et al. The mitochondrial acyl carrier protein (ACP) coordinates mitochondrial fatty acid synthesis with iron sulfur cluster biogenesis. Elife5, 10.7554/eLife.17828 (2016).
    1. Stroud DA, et al. Accessory subunits are integral for assembly and function of human mitochondrial complex I. Nature. 2016;538:123–126. doi: 10.1038/nature19754.
    1. Zheng M, et al. Cardiac-specific ablation of Cypher leads to a severe form of dilated cardiomyopathy with premature death. Hum. Mol. Genet. 2009;18:701–713. doi: 10.1093/hmg/ddn400.
    1. Balaban RS, Kantor HL, Katz LA, Briggs RW. Relation between work and phosphate metabolite in the in vivo paced mammalian heart. Science. 1986;232:1121–1123. doi: 10.1126/science.3704638.
    1. Neely JR, Rovetto MJ, Whitmer JT, Morgan HE. Effects of ischemia on function and metabolism of the isolated working rat heart. Am. J. Physiol. 1973;225:651–658. doi: 10.1152/ajplegacy.1973.225.3.651.
    1. Matthews PM, Bland JL, Gadian DG, Radda GK. The steady-state rate of ATP synthesis in the perfused rat heart measured by 31P NMR saturation transfer. Biochem. Biophys. Res. Commun. 1981;103:1052–1059. doi: 10.1016/0006-291X(81)90915-3.
    1. Allue I, et al. Evidence for rapid consumption of millimolar concentrations of cytoplasmic ATP during rigor-contracture of metabolically compromised single cardiomyocytes. Biochem. J. 1996;319(Pt 2):463–469. doi: 10.1042/bj3190463.
    1. Wang, X. et al. Mitochondrial flashes regulate ATP homeostasis in the heart. Elife6, 10.7554/eLife.23908 (2017).
    1. Hiltunen JK, et al. Mitochondrial fatty acid synthesis and respiration. Biochim. Biophys. Acta. 2010;1797:1195–1202. doi: 10.1016/j.bbabio.2010.03.006.
    1. Jian C, et al. Deficiency of PHB complex impairs respiratory supercomplex formation and activates mitochondrial flashes. J. Cell Sci. 2017;130:2620–2630. doi: 10.1242/jcs.198523.
    1. Mitsopoulos P, et al. Stomatin-like protein 2 is required for in vivo mitochondrial respiratory chain supercomplex formation and optimal cell function. Mol. Cell. Biol. 2015;35:1838–1847. doi: 10.1128/MCB.00047-15.
    1. Wagener N, Ackermann M, Funes S, Neupert W. A pathway of protein translocation in mitochondria mediated by the AAA-ATPase Bcs1. Mol. cell. 2011;44:191–202. doi: 10.1016/j.molcel.2011.07.036.
    1. Maio N, Rouault TA. Iron-sulfur cluster biogenesis in mammalian cells: new insights into the molecular mechanisms of cluster delivery. Biochim. Biophys. Acta. 2015;1853:1493–1512. doi: 10.1016/j.bbamcr.2014.09.009.
    1. Roche B, et al. Iron/sulfur proteins biogenesis in prokaryotes: formation, regulation and diversity. Biochim. Biophys. Acta. 2013;1827:455–469. doi: 10.1016/j.bbabio.2012.12.010.
    1. Karamanlidis G, et al. Mitochondrial complex I deficiency increases protein acetylation and accelerates heart failure. Cell Metab. 2013;18:239–250. doi: 10.1016/j.cmet.2013.07.002.
    1. Murphy E, Steenbergen C. Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. Physiol. Rev. 2008;88:581–609. doi: 10.1152/physrev.00024.2007.
    1. Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. N. Engl. J. Med. 2007;357:1121–1135. doi: 10.1056/NEJMra071667.
    1. Eltzschig HK, Eckle T. Ischemia and reperfusion–from mechanism to translation. Nat. Med. 2011;17:1391–1401. doi: 10.1038/nm.2507.
    1. Muller FL, Liu Y, Van Remmen H. Complex III releases superoxide to both sides of the inner mitochondrial membrane. J. Biol. Chem. 2004;279:49064–49073. doi: 10.1074/jbc.M407715200.
    1. Matsumoto-Ida M, et al. Real-time 2-photon imaging of mitochondrial function in perfused rat hearts subjected to ischemia/reperfusion. Circulation. 2006;114:1497–1503. doi: 10.1161/CIRCULATIONAHA.106.628834.
    1. Wang X, et al. Cardioprotection of ischemia/reperfusion injury by cholesterol-dependent MG53-mediated membrane repair. Circ. Res. 2010;107:76–83. doi: 10.1161/CIRCRESAHA.109.215822.
    1. Moreno-Lastres D, et al. Mitochondrial complex I plays an essential role in human respirasome assembly. Cell Metab. 2012;15:324–335. doi: 10.1016/j.cmet.2012.01.015.
    1. Chouchani ET, et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature. 2014;515:431–435. doi: 10.1038/nature13909.
    1. Fiedorczuk K, et al. Atomic structure of the entire mammalian mitochondrial complex I. Nature. 2016;538:406–410. doi: 10.1038/nature19794.
    1. Guerrero-Castillo S, et al. The assembly pathway of mitochondrial respiratory chain complex I. Cell Metab. 2017;25:128–139. doi: 10.1016/j.cmet.2016.09.002.
    1. Acin-Perez R, et al. Respiratory active mitochondrial supercomplexes. Mol. cell. 2008;32:529–539. doi: 10.1016/j.molcel.2008.10.021.
    1. Medeiros DM. Assessing mitochondria biogenesis. Methods. 2008;46:288–294. doi: 10.1016/j.ymeth.2008.09.026.
    1. Cheng H, Lederer WJ, Cannell MB. Calcium sparks: elementary events underlying excitation-contraction coupling in heart muscle. Science. 1993;262:740–744. doi: 10.1126/science.8235594.
    1. Shang W, et al. Cyclophilin D regulates mitochondrial flashes and metabolism in cardiac myocytes. J. Mol. Cell Cardiol. 2016;91:63–71. doi: 10.1016/j.yjmcc.2015.10.036.
    1. Shen T, et al. Mitofusin-2 is a major determinant of oxidative stress-mediated heart muscle cell apoptosis. J. Biol. Chem. 2007;282:23354–23361. doi: 10.1074/jbc.M702657200.
    1. Zhang SJ, et al. Isoform evolution in primates through independent combination of alternative RNA processing events. Mol. Biol. Evol. 2017;34:2453–2468. doi: 10.1093/molbev/msx212.
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009;4:44–57. doi: 10.1038/nprot.2008.211.
    1. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37:1–13. doi: 10.1093/nar/gkn923.

Source: PubMed

3
Abonnere