Skip to main content

ORIGINAL RESEARCH article

Front. Aging Neurosci., 16 February 2018
Sec. Cellular and Molecular Mechanisms of Brain-aging
Volume 10 - 2018 | https://doi.org/10.3389/fnagi.2018.00032

Increased Degradation Rates in the Components of the Mitochondrial Oxidative Phosphorylation Chain in the Cerebellum of Old Mice

Aurel Popa-Wagner1,2,3*† Raluca E. Sandu2† Coman Cristin4 Adriana Uzoni5 Kevin A. Welle6 Jennifer R. Hryhorenko6 Sina Ghaemmaghami6
  • 1Department of Neurology, Chair of Vascular Neurology and Dementia, Essen University Hospital, Essen, Germany
  • 2Neurobiology of Aging Group, University of Medicine and Pharmacy Craiova, Craiova, Romania
  • 3School of Medicine, Griffith University, Southport, QLD, Australia
  • 4Institutul Naţional de Cercetare şi Dezvoltare pentru Microbiologie şi Imunologie (Cantacuzino), Bucharest, Romania
  • 5Department of Psychiatry, University of Medicine Rostock, Rostock, Germany
  • 6Department of Biology, University of Rochester, Rochester, NY, United States

Brain structures differ in the magnitude of age-related neuron loss with the cerebellum being more affected. An underlying cause could be an age-related decline in mitochondrial bioenergetics. Successful aging of mitochondria reflects a balanced turnover of proteins involved in mitochondrial biogenesis and mitophagy. Thus, an imbalance in mitochondrial turnover can contribute to the diminution of cellular function seen during aging. Mitochondrial biogenesis and mitophagy are mediated by a set of proteins including MFN1, MFN2, OPA1, DRP1, FIS1 as well as DMN1l and DNM1, all of which are required for mitochondrial fission. Using N15 labeling, we report that the turnover rates for DMN1l and FIS1 go in opposite directions in the cerebellum of 22-month-old C57BL6j mice as compared to 3-month-old mice. Previous studies have reported decreased turnover rates for the mitochondrial respiratory complexes of aged rodents. In contrast, we found increased turnover rates for mitochondrial proteins of the oxidative phosphorylation chain in the aged mice as compared to young mice. Furthermore, the turnover rate of the components that are most affected by aging –complex III components (ubiquinol cytochrome C oxidoreductase) and complex IV components (cytochrome C oxidase)– was significantly increased in the senescent cerebellum. However, the turnover rates of proteins involved in mitophagy (i.e., the proteasomal and lysosomal degradation of damaged mitochondria) were not significantly altered with age. Overall, our results suggest that an age-related imbalance in the turnover rates of proteins involved in mitochondrial biogenesis and mitophagy (DMN1l, FIS1) in conjunction with an age-related imbalance in the turnover rates of proteins of the complexes III and IV of the electron transfer chain, might impair cerebellar mitochondrial bioenergetics in old mice.

Introduction

Brain structures differ in the magnitude of age-related neuronal loss. For example, during normal aging in humans and non-human mammals, the number of neurons in the hippocampus remains relatively stable whereas in the cerebellum, a clear age-related neuron loss is observed (Kennard et al., 2013), suggesting that cerebellum-essential tasks show age-related deficits at earlier ages than do hippocampus-essential tasks (Woodruff-Pak and Finkbiner, 1995; Woodruff-Pak et al., 2010).

There is a strong relationship between energy metabolism, aging, and longevity (Lehmann et al., 2008, 2013). For example, calorie restriction correlates with extended life expectancies and fewer age-associated diseases in a wide range of organisms (Heilbronn and Ravussin, 2003). This link is also supported by the free radical hypothesis that links oxidative damage production to energy metabolism and the disposable soma theory which hypothesizes a tradeoff between energy invested in maintenance and reproduction (van Leeuwen et al., 2010). Therefore, mitochondria are important in aging both due to ROS production that contribute to cell damage, and the accumulation of mutations in mitochondrial DNA, which in turn, results in mitochondrial dysfunction (Batlevi and La Spada, 2011; Stefanatos and Sanz, 2017). Thus, comparative studies showed that the stability of mitochondrial DNA, reflected by GC content and mitochondria-associated variables such as resting metabolic rate and body temperature, are determinants (predictors) of mammalian longevity (Lehmann et al., 2008, 2013; Yanai et al., 2016, 2017).

Mitochondria could play a key role in the pathophysiology of aging (Chan, 2006; Navarro et al., 2012; Gonzalez-Freire et al., 2015; Grimm and Eckert, 2017; Herst et al., 2017). In the brain, a recent report showed an age- and brain region-specific differences in mitochondrial bioenergetics in Brown Norway rats (Pandya et al., 2016).

Human cerebral cortex, human cerebellum, and chimpanzee cerebral cortex, each undergo different patterns of age-related gene expression alterations (Fraser et al., 2005). An underlying cause of age-related alterations in gene expression could be an age-related genomic response to altered mitochondrial bioenergetics.

In the rat cerebellum, the number of efficient organelles decreases with advancing age and this may be of critical importance for the proper function of nerve cells when the energy demand is high (Fattoretti et al., 1996). Likewise, mitochondrial metabolic competence, that is, the capacity of mitochondria to provide adequate amounts of energy, declines in the old monkey cerebellum (Bertoni-Freddari et al., 2008).

Recent evidence suggests that complex I activity decreases in the cortex, brainstem and cerebellum of middle aged mice and much more in the cerebellum of the pcd5j mouse, a cerebellar ataxic neurodegeneration model (Pollard et al., 2016). However, the directionality and extent of mitochondrial proteomic alterations underlying the decreased mitochondrial metabolic competence are not clear (Dai et al., 2014; Martin et al., 2014; Stauch et al., 2014; Karunadharma et al., 2015). In the current study, by using 15N labeling of proteins in young and old mice, we report highly heterogenous turnover rates for cerebellar mitochondrial proteins.

Materials and Methods

Animals and Experimental Design

The subjects of these experiments were young (3-month-old) and aged (18-month-old) male C57BL/6J mice kept under standard laboratory conditions with free access to food and water. For 15N-labeling, mice were fed with natural algae for 7 days. After this habituation period, a diet of 15N-enriched algae was introduced (t = 0) and mice were maintained on this isotopically enriched diet for the remainder of the experiment. At 15N-incorporation time points of 0, 1, 2, 4, 8, and 16 days, young (N = 2/time point) and aged (N = 3/time point) were deeply anesthetized and perfused with buffered saline and the brain tissue was collected. All protocols and procedures were performed in accordance with the guidelines of the Romania Experimental Animals Protection Law and approved by the Animal Experimentation Ethics Board of the University of Medicine and Pharmacy Craiova, Romania.

Sample Preparation

For protein analysis, dissected cerebellums were homogenized in sample buffer containing 8 M urea, 2 M thiourea, 4% CHAPS, 65 mM DTT, 40 mM Tris and protease inhibitor cocktail (Roth, Karlsruhe, Germany). Total protein was quantified by the Bradford method.

Ten micrograms of each extract was ran on an Invitrogen 4–12% Bis-Tris gel for 5 min at 200V and stained with SimplyBlue SafeStain. Gel bands were diced into small pieces and places in Lowbind tubes. Gel pieces were rinsed with ammonium bicarbonate and acetonitrile and dried in a Speed Vac for 5 min. One hundred microliters of 10 mM DTT in 50 mM ammonium bicarbonate was added to gel pieces and incubated at 55°C for 1 h. Supernatant was removed and 100 μl of 50 mM 2-iodoacetamide in 50 mM ammonium bicarbonate was added to the gel pieces and incubated in the dark for 30 min. After rinsing with ammonium bicarbonate and acetonitrile, the gel pieces were dried in a Speed Vac for 5 min. Trypsin solutions were made by reconstituting Trypsin Gold (100 ug) in 400 ul of 50 mM acetic acid. Trypsin aliquots were diluted to 10 ng/uL with 50 mM ammonium bicarbonate and added to gel pieces. The gels were incubated at room temperature for 1 h for rehydration and then placed at 37°C overnight. Solutions were transferred into a clean 0.5 ml Lobind Tube and 50% ACN/0.1% TFA was added to cover gel pieces and shaken for 25 min. Extracted supernatants were pooled and freeze dried. The samples were resuspended in 50 μL of 0.1% TFA, and proceeded to mass spectrometry analysis.

LC-MS/MS Analysis

Six microliters of each sample was analyzed on a Q Exactive Plus mass spectrometer (Thermo Scientific) coupled with an Easy nLC-1000 pump (Thermo Scientific). Columns were hand-pulled using 100 um fused silica, which was packed with 30 cm of 1.8 um, 120 Angstrom C18 beads (Sepax). Mobile phase A was 0.1% formic acid in water, and mobile phase B was 0.1% formic acid in acetonitrile. Peptides were separated using a gradient of 8–30% B over 145 min, 30–50% B over 10 min, and 50–70% B over 4 min, holding at 70% B for 5 min. The gradient returned to 0% B in 4 min, and held there for 10 min to prepare for the next injection. The flow rate throughout the run was held at 300 uL/min. A data-dependent top 10 MS2 method was used for peptide detection and fragmentation. For each cycle, one full scan from m/z 400–1700 was acquired in the Orbitrap at a resolution of 70,000 at m/z = 200, with an AGC target of 3e6, and a maximum injection time of 50 ms. After each full scan, the top 10 most intense ions were selected for fragmentation in the HCD cell, followed by detection in the Orbitrap at a resolution of 35,000 at m/z = 200. The AGC target was set to 1e5, with a maximum injection time of 150 ms, an isolation width of 1.5 Da, and a normalized collision energy of 27. A dynamic exclusion window of 25 s was used, while excluding unassigned, +1, and greater than +5 charge states. The polysiloxane 445.12003 m/z was used for lockmass. For Stable Isotope Labeling in Cell Culture (SILAC) LC-MS/MS analysis on the TMT-labeled, un-mixed samples, all settings remained the same except for the isolation width, which was adjusted up to 1.0 Da, with an offset of 0.3.

Data Analysis

MS/MS peaklists were extracted using the program PAVA from the LC-MS/MS raw data files corresponding to the unlabeled tissues. Peptide identification was conducted by Protein Prospector against the Mus Musculus Uniprot database (downloaded June 27, 2016). To this database, a randomized version was concatenated to allow determination of false discovery rates. The search parameters were the following: species = Mus musculus; enzyme specificity = trypsin; allowed missed cleavages = 1; fixed modification = carbamidomethylation; variable modifications = acetylation on protein N terminus, glutamine on peptide N-terminal to glutamic acid, methionine loss from protein N terminus, methionine loss from protein N terminus and acetylation, methionine oxidation; maximal number of variable modifications = 2; parent mass tolerance = 50 ppm; fragment mass tolerance = 0.6 Da; peptide expectation cutoff = 0.05. The match of sequences from the decoy database (normal + random) indicated that the false discovery rates for this expectation cutoff value were less than 0.2% for all analyzed datasets. A complete list of protein identifications and supporting statistics are provided in Supplementary Table S1.

Using the Protein Prospector database search results, the following information was gathered for each peptide ion with an expectation value less than 0.05: monoisotopic mass to charge ratio (m/z), charge (z), retention time (RT), assigned protein and sequence. The data were tabulated as a text file. Using the program PAVA(1), MS1 spectra were extracted from all raw data files obtained from the labeling time-course. The MS1 spectra were centroided and the resulting peaklists were used for further analysis. Using a script written in Java (mssplice) (Guan et al., 2011), the following set of analyses were sequentially conducted for each tabulated peptide obtained from the database search:

(1) Numbers of nitrogen atoms were determined for each peptide sequence.

(2) The possible set of isotopic m/z values for each peptide was determined, assuming a range of 0.0–1.0 15N incorporation ratios.

(3) Using a series of overlapping 30-s RT windows, ranging from 2 min prior to 2 min passed the experimentally determined RT, MS1 spectra ranging from the monoisotopic m/z to the maximum possible isotopic m/z (assuming 100% 15N incorporation) were collected and averaged. The total intensity of all possible peptide isotopic m/z values were summed and plotted as a function of RT to generate a chromatogram for each peptide. The chromatogram was fitted with a Gaussian function to determine the peak width. MS1 spectra within the determined RT peak width and the m/z window were summed and the aggregated spectra were used for further analysis.

(4) The extracted MS1 peptide spectra were fitted with a probabilistic combinatorial isotopic distribution model that considers the spectra as the sum of two isotopic populations (labeled and unlabeled). The 15N enrichment of the unlabeled population was set as a constant to its natural abundance. The 15N enrichment of the labeled population and the ratio of labeled and unlabeled populations were considered as the two fitted variables. Least-square fitting to the model was used to obtain the following information: the fraction 15N incorporation of the labeled population, the ratio of labeled and unlabeled populations, goodness of fits (coefficient of determination – R2) to predicted isotopomer distributions for labeled and unlabeled populations. Only spectra that had R2 values of greater than 0.8 for both populations were used for further analysis.

(5) For each peptide ion where more than four unique time-points passed the above criteria, the fraction labeled values were fitted to a single exponential equation to determine the clearance rate.

(6) Peptide level labeled population measurements were aggregated for each homologous group of proteins. Peptides with sequences that were shared among multiple homologous groups were not considered.

(7) The aggregated data were fitted with a single exponential equation to determine protein clearance rates. A complete list of measured turnover rates are provided in the Supplementary Table S1.

Results

Proteome-wide analyses of turnover by mass spectrometry have been greatly propelled by the development of SILAC – a set of standardized protocols, reagents and analysis tools aimed at quantifying the incorporation of 15N/13C labeled amino acids into proteins (Ong and Mann, 2007). Conducting dynamic (i.e., time-resolved) SILAC experiments are now fairly routine and have enabled global analyses of protein turnover in a number of cellular systems (Cambridge et al., 2011; Schwanhausser et al., 2011). In a recent study (Price et al., 2010), we modified a 15N labeling strategy developed by Wu et al. (2004) to measure in vivo turnover rates in various mouse tissues. The procedure involves the complete isotopic labeling of blue green algae (Spirulina platensis) with 15N labeled salts and utilizing it as a dietary source of nitrogen for mice. By quantifying mass shifts and isotopomer distributions, we are able to obtain turnover rates for thousands of proteins across multiple tissues. In this study, we measured rate constants for fractional isotopic labeling to analyze age-induced changes in global rates of degradation in mice. Thus, the measured rate constants correspond to the first order rate constant for degradation. Differences in synthesis rates do not influence this rate constant (Claydon and Beynon, 2012).

The assembly and maintenance of respiratory complexes is central to both successful aging and protection from age-related diseases. Thus, an imbalance in mitochondrial turnover can contribute to a loss of function seen during aging.

A hierarchical gene ontology (GO) enrichment analysis of proteins with enhanced degradation using the algorithm GOrilla (Eden et al., 2009) in old mice revealed most significant changes in the mitochondrial part and in particular in the mitochondrial protein complex (Figure 1). The complete results of the GO analysis are tabulated in Supplementary Table S2.

FIGURE 1
www.frontiersin.org

FIGURE 1. Gene ontology (GO) analysis of proteins with enhanced degradation in old mice. Log2 ratios of degradation rates between old and young mice cerebellum samples were measured (see Materials and Methods). A hierarchical GO enrichment analysis was conducted for proteins with enhanced degradation in the old mice. The statistical significance (P-value) of gene enrichment of differentially expressed genes within each GO accession is indicated by the color shading. The hierarchical relationship between accessions is indicated by arrows. The analysis was conducted using the algorithm GOrilla (2). The table provides additional information about each accession, including the false discovery rate and enrichment level. This figure is limited to the cellular component GO category. The complete results of the GO analysis are tabulated in Supplementary Table S2. A list of proteins belonging to the GO term “mitochondrial protein complex” that are enriched among the subset of proteins with enhanced degradation rates in old mice is provided.

Mitochondrial mass is modulated during the lifespan of the cell, in accordance with the physiological state. This modulation involves mitochondrial biogenesis including metabolites supply, fusion and fission but also and mitochondrial degradation, including autophagy, lysosome and proteasome processes. However, of these, only the turnover rates of proteins of the oxidative phosphorylation complexes were significantly changed in the cerebellum of the old mice (Figure 2). In particular, we noted significantly increased degradation rates in the components of the complex I (NADH dehydrogenase), complex III (ubiquinol cytochrome C oxidoreductase), complex IV (cytochrome C oxidase), and complex V (ATP synthase) in the cerebellum of old mice. For complex 2, a single subunit was quantifiable.

FIGURE 2
www.frontiersin.org

FIGURE 2. Distribution of log2 ratios of degradation rates between old and young mice for a select group of functional categories related to protein homeostasis. Log2 ratios of degradation rates between old and young mice cerebellum samples were measured (see Materials and Methods). Box plots indicate the distribution of log2 ratios degradation rates for each category. The box indicates the interquartile range (IQR) and the line indicates the median. Far outliers (>1.5IQR) were excluded.

In the following, we give an account of changes in the degradation rates of specific proteins of the oxidative phosphorylation complexes (Table 1). The dynamic balance of fusion and fission of mitochondria determines their morphology and allows their immediate adaptation to energetic needs. It also preserves mitochondria functionality and morphology by restoring or removing damaged organelles or precipitates cells in apoptosis in cases of severe defects. Thus, we noted that the turnover rate of dynamin-1-like protein (Dmn1l), a protein that mediates mitochondria fission, was three-fold increased in the cerebellum of old mice. However, the turnover rate of dynamin 1,0 (Dnm1), a protein also involved in mitochondrial fission, did not change significantly with increasing age. Finally, the turnover rate of mitochondrial fission 1 protein (Fis1), also involved in mitochondrial fission, was decreased two-fold in the cerebellum of the old mice.

TABLE 1
www.frontiersin.org

TABLE 1. Changes in the degradation rates of specific proteins of the oxidative phosphorylation complexes.

Aging of the mammalian brain is associated with decreased electron transfer in mitochondria isolated from old animals and a continuous decrease of the capacity to produce ATP by oxidative phosphorylation (Boveris and Navarro, 2008). Decreased ATP production could be due to either a failure of the biochemical pathway to supply mitochondria with pyruvate and reducing power or a decreased efficiency of electron transfer in mitochondria due to age-related changes in the components of the oxidative chain. We found that turnover rates in the enzymes and proteins required for mitochondria supply with oxidative phosphorylation substrates did not change significantly with age (Table 1).

Previous studies have shown that mitochondria isolated from brain, liver, heart, and kidney of old mice showed decreased electron transfer activity in complexes I and IV, whereas complexes II and III were largely unaffected (Navarro and Boveris, 2004, 2005, 2007). In terms of turnover rates, we found a significant increase in the turnover rates of the complex IV components (cytochrome C oxidase) and to a lesser extent in the complex III (ubiquinol cytochrome C oxidoreductase) whereas turnover rates in the complexes I, II, and V were largely unaffected by increasing age (Figure 3). Within complex IV, increases in the degradation rate were found for cytochrome oxidase subunit VIIA (4.3-fold) of the old mice cerebellum, followed by cytochrome C, an electron carrier protein that transfers electrons to the cytochrome oxidase complex and cytochrome oxidase subunit VIC and cytochrome oxidase subunit VA (both, 2.7-fold) and to a lesser extent cytochrome oxidase subunit VB (1.9-fold). One protein showed, nevertheless, decreased turnover rates in the mitochondria from old cerebellum, i.e., cytochrome oxidase subunit IV (-0.97).

FIGURE 3
www.frontiersin.org

FIGURE 3. Distribution of degradation rates for old and young mice for components of the mitochondrial transport chain. Box plots indicate the distribution of degradation rates for all subunits quantified for each complex. The box indicates the interquartile range (IQR) and the line indicates the median. Far outliers (>1.5IQR) were excluded. P-values for comparisons between old and young mice were conducted by two-sided Mann–Whitney U test, considering values from different subunits as independent measurements of kdegradation (d-1) for a given complex. For complex 2, a single subunit was quantifiable, and the data is shown as that single measurement rather than a distribution.

The second largest changes in turnover rates of proteins of the mitochondrial respiratory chain were noted for complex III, and especially for ubiquinol cytochrome C reductase subunit VII (10.6-fold), ubiquinol cytochrome C reductase hinge protein (2.5-fold) and ubiquinol cytochrome C reductase Rieske iron-sulfur polypeptide 1 (2.12-fold). Negative turnover rates in the old cerebellum were found for ubiquinol cytochrome C reductase binding protein (-1.63-fold) and ubiquinol cytochrome C reductase core protein (-0.86-fold).

Discussion

Age-related changes in cerebellar mitochondria are reflected in alterations of their morphology, function, and biochemical properties. Further, an imbalance in mitochondrial turnover can contribute to a loss of function seen during aging. In this work, using a different labeling strategy of mitochondrial proteins, we report increased turnover rates for mitochondrial proteins of the complexes III and IV of the electron transport chain in the cerebellum of old mice. Nevertheless, we noted that complexes III and IV were more dramatically affected in comparison with complexes I, II, and V.

Results on the mitochondrial proteins turnover with increasing age have yielded conflicting results (Ingram and Chakrabarti, 2016). A slower rate of mitochondrial turnover with increasing age caused by reduced mitochondrial biogenesis and/or inefficient mitochondrial degradation has been documented (Terman et al., 2010; Martin et al., 2014). Further, by quantitative proteomics there was an age-dependent decrease in the levels of proteins involved in mitochondrial function, electron transport chain, citric acid cycle, and fatty acid metabolism as well as an increased abundance of proteins involved in glycolysis and oxidative stress response (Dai et al., 2014).

A recent study addressed directly the turnover rates of mitochondrial proteins across various tissues in young and old mice. Thus, using metabolic [2H3]-leucine heavy isotope labeling, it was found that the turnover rates were highly heterogeneous between subunits of the respiratory complexes (Karunadharma et al., 2015).

As it has been shown for muscle, the changes in protein homeostasis in the aged mitochondria suggest a breakdown of coordination of biosynthesis and protein recycling which might result in increased degradation rates for some components of the electron transfer chain. Thus, the loss of stoichiometry and lower mitochondrial efficiency due to the elevation of mitochondrial protein expression, particularly in complex I, is indicative of lack of a coordinated response of the mitochondrial proteome to age-induced stress in susceptible tissue (Kruse et al., 2016).

Functional mitochondria is maintained by timely replacement of its components, in particular the proteins of the electron transfer chain, that are exposed to aggressive ROS generated during oxidative phosphorylation (Wang et al., 2013). Therefore, an increased turnover rate for components of the respiratory chain in the old cerebellum might also reflect an increased need to replace worn out components of the electron transfer chain which accumulate with increasing age in long-lived cells like neurons (Wang et al., 2013).

Mitochondria are extremely dynamic organelles engaged in a constant process of fission and fusion that constantly rejuvenate mitochondria (Detmer and Chan, 2007). Ideally, successful aging of mitochondria shall reflect a balance between mitochondria biogenesis and mitophagy. This process is mediated by a set of proteins including (i) mitofusins 1 and 2 (Mfn1, Mfn2) which mediate the outer membrane fusion, (ii) optic atrophy 1 (OPA1) which mediates inner membrane fusion, (iii) dynamin-related protein 1 (Drp1) and Fis1 as well as (iv) dynamin-1-like protein (Dmn1l) and dynamin 1,0 (Dnm1), all being required for mitochondria fission (Detmer and Chan, 2007; Batlevi and La Spada, 2011).

Proteins that control mitochondrial fusion/fission dynamics are ubiquitous. Therefore, their disturbances are associated with several diseases. This is particularly relevant for the brain because of the constant need for high levels of energy in neuronal cells. Age-related degeneration of cerebellar Purkinje cells has been linked to various age-related disabilities including dysfunction of postural control, cerebellar ataxia, essential tremor, Alzheimer’s disease and Lewy Body disease (Di Bella et al., 2010; Zhang et al., 2010). Clinically, mitochondrial dysfunction has emerged as a potential mechanism underlying Parkinson’s disease (PD). More specifically, complexes I and II deficiency has been reported in homogenates of postmortem substantia nigra samples of PD patients (Schapira et al., 1990; Grünewald et al., 2016). Further, familial PD cases were found to harbor mutations in proteins involved in the removal of damaged mitochondria (Kumar et al., 2011).

Current evidence suggests that unbalanced mitochondrial dynamics may contribute to Huntington disease (HD), too (Oliveira and Lightowlers, 2010; Costa and Scorrano, 2012). Thus, HD patients exhibit elevated mRNA and protein levels of the fission-associated genes, DRP1 and FIS1, together with decreased levels of mitofusins in striatal and cortical regions (Kim et al., 2010; Shirendeb et al., 2012). It should be noted that Drp1 deletion in the cerebellum results in (i) mitochondrial swelling, increased oxidative damage, (ii) ubiquitination of mitochondrial proteins, (iii) accumulation of autophagy markers and, (iv) impaired activity of the complex IV activity, ultimately leading to neurodegeneration (Kageyama et al., 2012).

Similarly, increased mitochondrial fragmentation and decreased levels for Mfn1 and Mfn2 in cultured cortical neurons and cortical neurons derived from a newly developed HD mouse model (BACHD) have been reported. Furthermore, increased Drp1 and Fis1 mRNA levels were found in HD mice (Shirendeb et al., 2012). Of note, germline mutations in Mfn2 cause Charcot–Marie–Tooth hereditary-like neuropathy type 2A (CMT2A), while germline mutations in OPA1 causes the autosomal dominant optic atrophy (ADOA) which is the most common genetic cause of optic atrophy in humans (Knott and Bossy-Wetzel, 2008).

Mitochondrial fission is also important for neuronal function, as dominant-negative Drp1 mutation can cause a lethal infantile neurodegenerative phenotype (Waterham et al., 2007). Thus, Drp1 knock-out mice display embryonic lethality, increased mitophagy and dilated cardiomyopathy, associated with increased levels of necrotic cell death, as well as abnormal brain development and failure to develop synapses (Ishihara et al., 2009; Wakabayashi et al., 2009; Song et al., 2015). Mitochondrial fission has also been linked to apoptotic cell death of neurons (Knott and Bossy-Wetzel, 2008). On the other hand, inhibition of mitochondrial fission with dominant-negative Drp1 or by Fis1 RNA knock-down, prevents mitophagy and results in accumulation of damaged mitochondria (Twig et al., 2008).

Mitophagy has been linked to aging, as impaired macroautophagy over time promotes mitochondrial dysfunction associated with the aging process. Further, an impaired macroautophagy over time promotes mitochondrial dysfunction associated with the aging process (Batlevi and La Spada, 2011).

It has been reported that aging is associated with an increased level of oxidized proteins and an imbalance in proteostasis due to impairment of the ubiquitin/proteasome system (Maharjan et al., 2014; Gonzalez-Freire et al., 2015). However, our results indicate that the turnover rates of proteins involved in mitophagy, proteasome and lysosomal degradation of damaged mitochondria are not significantly changed in the cerebellum of old mice.

Study Limitations

In this study, we report age-related changes in the degradation rate of proteins of the complexes III and IV of the electron transport chain in the old cerebellum. However, since the degradation rates of proteins are specific for different brain regions, the full significance of increased degradation rates for cerebellar mitochondrial proteins shall be clarified by additional studies on the other brain regions.

Conclusion

Previous studies have reported decreased turnover rates for mitochondrial proteins with increasing age in rodents. In the current study, by using N15 labeling, we found highly heterogeneous turnover rates for mitochondrial proteins, but contrary to prior findings, our results do not indicate decreased turnover rates for mitochondrial proteins in the aged mouse cerebellum. On the contrary, we found significantly increased turnover rates for several proteins of the electron transport chain, especially for components of the complex III (ubiquinol cytochrome C oxidoreductase) and complex IV (cytochrome C oxidase). Our results also indicate that the turnover rates of proteins involved in mitophagy are not significantly altered in the cerebellum of old mice.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This study was partially supported by grants from CNCS – UEFISCDI, PN-II-ID-PCE-2011-3-0848, “Age-related deterioration of genetic and biological pathways and their significance for brain tissue regeneration and functional recuperation after injuries” and PCCA no 80/2012, “Cellular Therapeutic Approaches for Regenerative Stroke Therapies” to AP-W.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fnagi.2018.00032/full#supplementary-material

TABLE S1 | A complete list of protein identifications and measured turnover rates for the young and aged mouse cerebellum.

TABLE S2 | The complete results of the GO analysis for the young and aged mouse cerebellum.

References

Batlevi, Y., and La Spada, A. R. (2011). Mitochondrial autophagy in neural function, neurodegenerative disease, neuron cell death, and aging. Neurobiol. Dis. 43, 46–51. doi: 10.1016/j.nbd.2010.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Bertoni-Freddari, C., Balietti, M., Giorgetti, B., Grossi, Y., Casoli, T., Di Stefano, G., et al. (2008). Selective decline of the metabolic competence of oversized synaptic mitochondria in the old monkey cerebellum. Rejuvenation Res. 11, 387–391. doi: 10.1089/rej.2008.0659

PubMed Abstract | CrossRef Full Text | Google Scholar

Boveris, A., and Navarro, A. (2008). Brain mitochondrial dysfunction in aging. IUBMB Life 60, 308–314. doi: 10.1002/iub.46

PubMed Abstract | CrossRef Full Text | Google Scholar

Cambridge, S. B., Gnad, F., Nguyen, C., Bermejo, J. L., Kruger, M., and Mann, M. (2011). Systems-wide proteomic analysis in mammalian cells reveals conserved, functional protein turnover. J. Proteome Res. 10, 5275–5284. doi: 10.1021/pr101183k

PubMed Abstract | CrossRef Full Text | Google Scholar

Chan, D. C. (2006). Mitochondria: dynamic organelles in disease, aging, and development. Cell 125, 1241–1252.

Google Scholar

Claydon, A. J., and Beynon, R. (2012). Proteome dynamics: revisiting turnover with a global perspective. Mol. Cell. Proteomics 11, 1551–1565. doi: 10.1074/mcp.O112.022186

PubMed Abstract | CrossRef Full Text | Google Scholar

Costa, V., and Scorrano, L. (2012). Shaping the role of mitochondria in the pathogenesis of Huntington’s disease. EMBO J. 31, 1853–1864. doi: 10.1038/emboj.2012.65

PubMed Abstract | CrossRef Full Text | Google Scholar

Dai, D. F., Karunadharma, P. P., Chiao, Y. A., Basisty, N., Crispin, D., Hsieh, E. J., et al. (2014). Altered proteome turnover and remodeling by short-term caloric restriction or rapamycin rejuvenate the aging heart. Aging Cell 13, 529–539. doi: 10.1111/acel.12203

PubMed Abstract | CrossRef Full Text | Google Scholar

Detmer, S. A., and Chan, D. C. (2007). Functions and dysfunctions of mitochondrial dynamics. Nat. Rev. Mol. Cell Biol. 8, 870–879. doi: 10.1038/nrm2275

PubMed Abstract | CrossRef Full Text | Google Scholar

Di Bella, D., Lazzaro, F., Brusco, A., Plumari, M., Battaglia, G., Pastore, A., et al. (2010). Mutations in the mitochondrial protease gene AFG3L2 cause dominant hereditary ataxia SCA28. Nat. Genet. 42, 313–321. doi: 10.1038/ng.544

PubMed Abstract | CrossRef Full Text | Google Scholar

Eden, E., Navon, R., Steinfeld, I., Lipson, D., and Yakhini, Z. (2009). GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinformatics 10:48. doi: 10.1186/1471-2105-10-48

PubMed Abstract | CrossRef Full Text | Google Scholar

Fattoretti, P., Bertoni-Freddari, C., Caselli, U., Paoloni, R., and Meier-Ruge, W. (1996). Morphologic changes in cerebellar mitochondria during aging. Anal. Quant. Cytol. Histol. 18, 205–208.

PubMed Abstract | Google Scholar

Fraser, H. B., Khaitovich, P., Plotkin, J. B., Pääbo, S., and Eisen, M. B. (2005). Aging and gene expression in the primate brain. PLOS Biol. 3:e274. doi: 10.1371/journal.pbio.0030274

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonzalez-Freire, M., De Cabo, R., Bernier, M., Sollott, S. J., Fabbri, E., Navas, P., et al. (2015). Reconsidering the role of mitochondria in aging. J. Gerontol. A Biol. Sci. Med. Sci. 70, 1334–1342. doi: 10.1093/gerona/glv070

PubMed Abstract | CrossRef Full Text | Google Scholar

Grimm, A., and Eckert, A. (2017). Brain aging and neurodegeneration: from a mitochondrial point of view. J. Neurochem. 143, 418–431. doi: 10.1111/jnc.14037

PubMed Abstract | CrossRef Full Text | Google Scholar

Grünewald, A., Rygiel, K. A., Hepplewhite, P. D., Morris, C. M., Picard, M., and Turnbull, D. M. (2016). Mitochondrial DNA depletion in respiratory chain–deficient Parkinson disease neurons. Ann. Neurol. 79, 366–378. doi: 10.1002/ana.24571

PubMed Abstract | CrossRef Full Text | Google Scholar

Guan, S., Price, J. C., Prusiner, S. B., Ghaemmaghami, S., and Burlingame, A. L. (2011). A data processing pipeline for mammalian proteome dynamics studies using stable isotope metabolic labeling. Mol. Cell. Proteomics 10:M111010728. doi: 10.1074/mcp.M111.010728

PubMed Abstract | CrossRef Full Text | Google Scholar

Heilbronn, L. K., and Ravussin, E. (2003). Calorie restriction and aging: review of the literature and implications for studies in humans. Am. J. Clin. Nutr. 78, 361–369.

Google Scholar

Herst, P. M., Rowe, M. R., Carson, G. M., and Berridge, M. V. (2017). Functional mitochondria in health and disease. Front. Endocrinol. 8:296. doi: 10.3389/fendo.2017.00296

CrossRef Full Text | Google Scholar

Ingram, T., and Chakrabarti, L. (2016). Proteomic profiling of mitochondria: what does it tell us about the ageing brain? Aging 8, 3161–3179. doi: 10.18632/aging.101131

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishihara, N., Nomura, M., Jofuku, A., Kato, H., Suzuki, S. O., Masuda, K., et al. (2009). Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat. Cell Biol. 11, 958–966. doi: 10.1038/ncb1907

PubMed Abstract | CrossRef Full Text | Google Scholar

Kennard, J. A., Brown, K. L., and Woodruff-Pak, D. S. (2013). Aging in the cerebellum and hippocampus and associated behaviors over the adult life span of CB6F1 mice. Neuroscience 247, 335–350. doi: 10.1016/j.neuroscience.2013.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Kageyama, Y., Zhang, Z., Roda, R., Fukaya, M., Wakabayashi, J., Wakabayashi, N., et al. (2012). Mitochondrial division ensures the survival of postmitotic neurons by suppressing oxidative damage. J. Cell Biol. 197, 535–551. doi: 10.1083/jcb.201110034

PubMed Abstract | CrossRef Full Text | Google Scholar

Karunadharma, P. P., Basisty, N., Chiao, Y. A., Dai, D. F., Drake, R., Levy, N., et al. (2015). Respiratory chain protein turnover rates in mice are highly heterogeneous but strikingly conserved across tissues, ages, and treatments. FASEB J. 29, 3582–3592. doi: 10.1096/fj.15-272666

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J., Moody, J. P., Edgerly, C. K., Bordiuk, O. L., Cormier, K., Smith, K., et al. (2010). Mitochondrial loss, dysfunction and altered dynamics in Huntington’s disease. Hum. Mol. Genet. 19, 3919–3935. doi: 10.1093/hmg/ddq306

PubMed Abstract | CrossRef Full Text | Google Scholar

Knott, A. B., and Bossy-Wetzel, E. (2008). Impairing the mitochondrial fission and fusion balance: a new mechanism of neurodegeneration. Ann. N. Y. Acad. Sci. 1147, 283–292. doi: 10.1196/annals.1427.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Kruse, S. E., Karunadharma, P. P., Basisty, N., Johnson, R., Beyer, R. P., MacCoss, M. J., et al. (2016). Age modifies respiratory complex I and protein homeostasis in a muscle type-specific manner. Aging Cell 15, 89–99. doi: 10.1111/acel.12412

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, K. R., Djarmati-Westenberger, A., and Grünewald, A. (2011). Genetics of Parkinson’s disease. Semin. Neurol. 31, 433–440. doi: 10.1055/s-0031-1299782

PubMed Abstract | CrossRef Full Text | Google Scholar

Lehmann, G., Muradian, K. K., and Fraifeld, V. E. (2013). Telomere length and body temperature-independent determinants of mammalian longevity? Front. Genet. 4:111. doi: 10.3389/fgene.2013.00111

PubMed Abstract | CrossRef Full Text | Google Scholar

Lehmann, G., Segal, E., Muradian, K. K., and Fraifeld, V. E. (2008). Do mitochondrial DNA and metabolic rate complement each other in determination of the mammalian maximum longevity? Rejuvenation Res. 11, 409–417. doi: 10.1089/rej.2008.0676

PubMed Abstract | CrossRef Full Text | Google Scholar

Maharjan, S., Oku, M., Tsuda, M., Hoseki, J., and Sakai, Y. (2014). Mitochondrial impairment triggers cytosolic oxidative stress and cell death following proteasome inhibition. Sci. Rep. 4:5896. doi: 10.1038/srep05896

PubMed Abstract | CrossRef Full Text | Google Scholar

Martin, O. J., Lai, L., Soundarapandian, M. M., Leone, T. C., Zorzano, A., Keller, M. P., et al. (2014). A role for peroxisome proliferator-activated receptor γ coactivator-1 in the control of mitochondrial dynamics during postnatal cardiac growth. Circ. Res. 114, 626–636. doi: 10.1161/CIRCRESAHA.114.302562

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., and Boveris, A. (2004). Rat brain and liver mitochondria develop oxidative stress and lose enzymatic activities on aging. Am. J. Physiol. Regul. Integr. Comp. Physiol. 287, R1244–R1249. doi: 10.1152/ajpregu.00226.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., and Boveris, A. (2005). Hypoxia exacerbates macrophage mitochondrial damage in endotoxic shock. Am. J. Physiol. Regul. Integr. Comp. Physiol. 288, R354–R355. doi: 10.1152/ajpregu.00726.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., and Boveris, A. (2007). The mitochondrial energy transduction system and the aging process. Am. J. Physiol. Cell Physiol. 292, C670–C686. doi: 10.1152/ajpcell.00213.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., Gomez, C., Lopez-Cepero, J. M., and Boveris, A. (2012). Beneficial effects of moderate exercise on mice aging: survival, behavior, oxidative stress, and mitochondrial electron transfer. Am. J. Physiol. Regul. Integr. Comp. Physiol. 21, 406–420. doi: 10.1093/hmg/ddr475

PubMed Abstract | CrossRef Full Text | Google Scholar

Oliveira, J. M., and Lightowlers, R. N. (2010). Could successful (mitochondrial) networking help prevent Huntington’s disease? EMBO Mol. Med. 2, 487–489. doi: 10.1002/emmm.201000104

PubMed Abstract | CrossRef Full Text | Google Scholar

Ong, S. E., and Mann, M. (2007). Stable isotope labeling by amino acids in cell culture for quantitative proteomics. Methods Mol. Biol. 359, 37–52.

Google Scholar

Pandya, J. D., Royland, J. E., MacPhail, R. C., Sullivan, P. G., and Kodavanti, P. R. (2016). Age- and brain region-specific differences in mitochondrial bioenergetics in Brown Norway rats. Neurobiol. Aging 42, 25–34. doi: 10.1016/j.neurobiolaging.2016.02.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Pollard, A. K., Craig, E. L., and Chakrabarti, L. (2016). Mitochondrial complex 1 activity measured by spectrophotometry is reduced across all brain regions in ageing and more specifically in neurodegeneration. PLOS ONE 11:e0157405. doi: 10.1371/journal.pone.0157405

PubMed Abstract | CrossRef Full Text | Google Scholar

Price, J. C., Guan, S., Burlingame, A., Prusiner, S. B., and Ghaemmaghami, S. (2010). Analysis of proteome dynamics in the mouse brain. Proc. Natl. Acad. Sci. U.S.A. 107, 14508–14513. doi: 10.1073/pnas.1006551107

PubMed Abstract | CrossRef Full Text | Google Scholar

Schapira, A. H., Cooper, J. M., Dexter, D., Clark, J. B., Jenner, P., and Marsden, C. D. (1990). Mitochondrial complex I deficiency in Parkinson’s disease. J. Neurochem. 54, 823–827. doi: 10.1111/j.1471-4159.1990.tb02325.x

CrossRef Full Text | Google Scholar

Schwanhausser, B., Busse, D., Li, N., Dittmar, G., Schuchhardt, J., Wolf, J., et al. (2011). Global quantification of mammalian gene expression control. Nature 473, 337–342. doi: 10.1038/nature10098

PubMed Abstract | CrossRef Full Text | Google Scholar

Shirendeb, U. P., Calkins, M. J., Manczak, M., Anekonda, V., Dufour, B., McBride, J. L., et al. (2012). Mutant Huntingtin’s interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington’s disease. Hum. Mol. Genet. 21, 406–420. doi: 10.1093/hmg/ddr475

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, M., Mihara, K., Chen, Y., Scorrano, L., and Dorn, G. W. (2015). Mitochondrial fission and fusion factors reciprocally orchestrate mitophagic culling in mouse hearts and cultured fibroblasts. Cell Metab. 21, 273–285. doi: 10.1016/j.cmet.2014.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Stauch, K. L., Purnell, P. R., and Fox, H. S. (2014). Aging synaptic mitochondria exhibit dynamic proteomic changes while maintaining bioenergetic function. Aging 6, 320–334.

PubMed Abstract | Google Scholar

Stefanatos, R., and Sanz, A. (2017). The role of mitochondrial ROS in the aging brain. FEBS Lett. doi: 10.1002/1873-3468.12902 [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Terman, A., Kurz, T., Navratil, M., Arriaga, E. A., and Brunk, U. T. (2010). Mitochondrial turnover and aging of long-lived postmitotic cells: the mitochondrial-lysosomal axis theory of aging. Antioxid. Redox Signal. 12, 503–535. doi: 10.1089/ars.2009.2598

PubMed Abstract | CrossRef Full Text | Google Scholar

Twig, G., Elorza, A., Molina, A. J. A., Mohamed, H., Wikstrom, J. D., Walzer, G., et al. (2008). Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J. 27, 433–446. doi: 10.1038/sj.emboj.7601963

PubMed Abstract | CrossRef Full Text | Google Scholar

van Leeuwen, I. M. M., Vera, J., and Wolkenhauer, O. (2010). Dynamic energy budget approaches for modelling organismal ageing. Philos. Trans. R. Soc. Lond. B Biol. Sci. 365, 3443–3454. doi: 10.1098/rstb.2010.0071

PubMed Abstract | CrossRef Full Text | Google Scholar

Wakabayashi, J., Zhang, Z., Wakabayashi, N., Tamura, Y., Fukaya, M., Kensler, T. W., et al. (2009). The dynamin-related GTPase Drp1 is required for embryonic and brain development in mice. J. Cell Biol. 186, 805–816. doi: 10.1083/jcb.200903065

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C. H., Wu, S. B., Wu, Y. T., and Wei, Y. H. (2013). Oxidative stress response elicited by mitochondrial dysfunction: implication in the pathophysiology of aging. Exp. Biol. Med. 238, 450–460. doi: 10.1177/1535370213493069

PubMed Abstract | CrossRef Full Text | Google Scholar

Waterham, H. R., Koster, J., Van Roermund, C. W., Mooyer, P. A., Wanders, R. J., and Leonard, J. V. (2007). A lethal defect of mitochondrial and peroxisomal fission. N. Engl. J. Med. 356, 1736–1741. doi: 10.1056/NEJMoa064436

PubMed Abstract | CrossRef Full Text | Google Scholar

Woodruff-Pak, D. S., and Finkbiner, R. G. (1995). Larger nondeclarative than declarative deficits in learning and memory in human aging. Psychol. Aging 10, 416–426.

PubMed Abstract | Google Scholar

Woodruff-Pak, D. S., Foy, M. R., Akopian, G. G., Lee, K. H., Zach, J., Nguyen, K. P., et al. (2010). Differential effects and rates of normal aging in cerebellum and hippocampus. Proc. Natl. Acad. Sci. U.S.A. 107, 1624–1629. doi: 10.1073/pnas.0914207107

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, C. C., MacCoss, M. J., Howell, K. E., Matthews, D. E., and Yates, J. R. III (2004). Metabolic labeling of mammalian organisms with stable isotopes for quantitative proteomic analysis. Anal. Chem. 76, 4951–4959.

Google Scholar

Yanai, H., Budovsky, A., Barzilay, T., Tacutu, R., and Fraifeld, V. E. (2017). Wide-scale comparative analysis of longevity genes and interventions. Aging Cell 16, 1267–1275. doi: 10.1111/acel.12659

PubMed Abstract | CrossRef Full Text | Google Scholar

Yanai, H., Budovsky, A., Tacutu, R., Barzilay, T., Abramovich, A., Ziesche, R., et al. (2016). Tissue repair genes: the TiRe database and its implication for skin wound healing. Oncotarget 7, 21145–21155. doi: 10.18632/oncotarget.8501

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, C., Zhu, Q., and Hua, T. (2010). Aging of cerebellar Purkinje cells. Cell Tissue Res. 341, 341–347. doi: 10.1007/s00441-010-1016-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: aging, mice, cerebellum, mitochondria, proteins, turnover

Citation: Popa-Wagner A, Sandu RE, Cristin C, Uzoni A, Welle KA, Hryhorenko JR and Ghaemmaghami S (2018) Increased Degradation Rates in the Components of the Mitochondrial Oxidative Phosphorylation Chain in the Cerebellum of Old Mice. Front. Aging Neurosci. 10:32. doi: 10.3389/fnagi.2018.00032

Received: 27 October 2017; Accepted: 26 January 2018;
Published: 16 February 2018.

Edited by:

Vadim Fraifeld, Ben-Gurion University of the Negev, Israel

Reviewed by:

Francesco Bellanti, University of Foggia, Italy
Ruslan Kuts, Israel University, Ecuador

Copyright © 2018 Popa-Wagner, Sandu, Cristin, Uzoni, Welle, Hryhorenko and Ghaemmaghami. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Aurel Popa-Wagner, aurel.popa-wagner@geriatrics-healthyageing.com; aurel.popa2011@gmail.com

These authors have contributed equally to this work.

Download