Fine-Tuning of PGC1α Expression Regulates Cardiac Function and Longevity
Graphical Abstract
Abstract
Rationale:
PGC1α (peroxisome proliferator-activated receptor gamma coactivator 1α) represents an attractive target interfering bioenergetics and mitochondrial homeostasis, yet multiple attempts have failed to upregulate PGC1α expression as a therapy, for instance, causing cardiomyopathy.
Objective:
To determine whether a fine-tuning of PGC1α expression is essential for cardiac homeostasis in a context-dependent manner.
Methods and Results:
Moderate cardiac-specific PGC1α overexpression through a ROSA26 locus knock-in strategy was utilized in WT (wild type) mice and in G3Terc−/− (third generation of telomerase deficient; hereafter as G3) mouse model, respectively. Ultrastructure, mitochondrial stress, echocardiographic, and a variety of biological approaches were applied to assess mitochondrial physiology and cardiac function. While WT mice showed a relatively consistent PGC1α expression from 3 to 12 months old, age-matched G3 mice exhibited declined PGC1α expression and compromised mitochondrial function. Cardiac-specific overexpression of PGC1α (PGC1αOE) promoted mitochondrial and cardiac function in 3-month-old WT mice but accelerated cardiac aging and significantly shortened life span in 12-month-old WT mice because of increased mitochondrial damage and reactive oxygen species insult. In contrast, cardiac-specific PGC1α knock in in G3 (G3 PGC1αOE) mice restored mitochondrial homeostasis and attenuated senescence-associated secretory phenotypes, thereby preserving cardiac performance with age and extending health span. Mechanistically, age-dependent defect in mitophagy is associated with accumulation of damaged mitochondria that leads to cardiac impairment and premature death in 12-month-old WT PGC1αOE mice. In the context of telomere dysfunction, PGC1α induction replenished energy supply through restoring the compromised mitochondrial biogenesis and thus is beneficial to old G3 heart.
Conclusions:
Fine-tuning the expression of PGC1α is crucial for the cardiac homeostasis because the balance between mitochondrial biogenesis and clearance is vital for regulating mitochondrial function and homeostasis. These results reinforce the importance of carefully evaluating the PGC1α-boosting strategies in a context-dependent manner to facilitate clinical translation of novel cardioprotective therapies.
Introduction
Meet the First Author, see p 660
Cardiac degeneration and cardiovascular diseases remain intricate threats to health and longevity. Unlike other high-turnover tissues, renascence of cardiomyocytes cannot be realized because of the lack of adult cardiac stem cells in the postmitotic heart.1–4 Instead, the integrity of mitochondrial function plays an important role in maintaining cardiac homeostasis and especially, during aging. A great body of work has shown that cardiac aging, including pathologically premature aging, is closely related to mitochondrial damage.5–9 Although the causal relationship between mitochondrial compromise and cardiac aging is still inconclusive, a consensus has been reached that maintaining the mitochondrial fitness is the key to attenuate age-dependent cardiac degeneration. However, the precise mechanism of mitochondrial quality control during cardiac aging remains to be elucidated.
The PGC1α (peroxisome proliferator-activated receptor gamma coactivator 1α) plays a prominent role in regulating mitochondrial biogenesis, oxidative phosphorylation, energy metabolism, oxidative stress response, and other functions across a wide range of tissues.10 Clinical significance of PGC1α lies in the derangements in PGC1α level, as well as Ppargc1a polymorphisms, and their associations with the clinical expression of diseases, for example, diabetes mellitus,11,12 cancer,13–15 and cardiomyopathy.16–18 The regulatory effects of PGC1α in pathogenesis have been manifested in different animal models; however, attempts to boost PGC1α level as a therapeutic strategy often meet contradictory results. For instance, PGC1α knockout in muscle led to reduced endurance capacity and exhibited fiber damage,19 while transgenic expression of PGC1α increased the content of slow-twitch muscle fibers, concurrent with enhanced exercise performance and peak oxygen uptake, but causing insulin resistance in animals fed high-fat diet.20 In heart, PGC1α-null mice did not present a baseline phenotype but worsened stress response.21 Conversely, high-level overexpression of PGC1α caused cardiomyopathy.10,22,23 Thus, these results implicate that regulation of PGC1α dosage is of particular importance and context dependent.
Age-associated telomere attrition represents one intrinsic driver of aging, contributing to the dysfunction of multiple organs and the occurrence of age-related diseases.24 The Terc−/− (telomerase deficient) mice have been utilized as a sophisticated and reliable aging model that resembles human aging process with chronic telomere dysfunction. Notably, heart degeneration in mice with shortened telomeres has been linked to p53-mediated suppression of PGC1α and consequent mitochondrial dysfunction.9 Accordingly, mice with dysfunctional mitochondria also exhibited a premature aging phenotype accompanied by a distinct senescence-associated secretory phenotype.25 In this scenario, it is worth constructing a mouse model of mild PGC1α knock in and deciphering whether cardiac PGC1α knock in causes distinct biological effects in telomere-intact and telomere dysfunctional mice, respectively, in which telomere dysfunctional heart has lower PGC1α expression in comparison to the telomere-intact counterpart.
To address the questions above, we utilized a ROSA26 locus knock-in approach to mildly overexpress PGC1α specifically in cardiomyocytes of WT (wild type) and G3Terc−/− (third generation [G3] of Terc−/−) mice. Functional characterizations of the 2 cohorts revealed that PGC1α promoted mitochondrial and cardiac function in 3-month-old WT and G3 mice but accelerated cardiac aging and significantly shortened life span in 12-month-old WT mice partially because of age-dependent defect in mitophagy. In contrast, in the context of telomere dysfunction, PGC1α induction replenished energy supply through restoring the compromised mitochondrial biogenesis and thus was beneficial to old G3 heart. Collectively, our results highlight the importance of fine-tuning PGC1α expression to maintain mitochondrial functionality and cardiac homeostasis during aging.
Methods
All data from these experiments are available from the corresponding author on reasonable request.
Animals
All studies with mice were approved by the Institutional Animal Use and Care Committee of Hangzhou Normal University. C57BL/6J mice (referred as WT) were purchased from the Laboratory Animal Center of Hangzhou Normal University. Cardiac-specific αMHC (α-myosin heavy chain) Cre recombinase (αMHC Cre+/−) mice, Terc+/− (T+/−) heterozygous and Terc−/− (T−/−) mice, PGC1αF/+ mice were all backcrossed for 10 generations onto a C57/BL6 background. The PGC1αF/+ mice were intercrossed with αMHC Cre+/− mice to generate PGC1αF/+-αMHC Cre+/− (WT PGC1αOE) mice, which were further crossed with T+/− mice to generate T+/− PGC1αOE mice. The T+/− PGC1αOE mice and T+/− mice were further crossed to generate the first generation of Terc−/− mice (G1Terc−/− mice) or G1Terc−/− PGC1αOE mice, which were crossed successively to produce the G3Terc−/− (G3 of Terc−/− mice), PGC1αOE (G3 PGC1αOE), and control littermates (ie, G3, αMHC Cre+/− G3, and PGC1αF/+ G3 mice). No developmental or survival differences were found among the control littermates examined; thus these 3 groups all designated G3 hereafter.
Electron Microscopy
Heart samples were fixed in 20-fold volumes of 2.5% glutaraldehyde in 0.1 M PBS solution for 48 hours, followed by 3× of rinse in 0.1 M PBS for 10 minutes each, and then fixed in 1% OsO4 for 1 hour at room temperature and rinsed in distilled H2O for 3×. The samples then were transferred into 1.5 mL-microcentrifuge tubes containing 2% uranyl acetate for 30 minutes, followed by gradient rinse in 50%, 70%, 90%, and 100% ethanol for 10 minutes and 2× of 100% acetone rinse for 15 minutes. The specimens were infiltrated, embedded, polymerized, sectioned, and stained as described previously.26
Statistics
Statistical analyses were performed using Prism 7 (GraphPad Software, Inc). Unpaired Student t test (2 tailed) was used to compare 2 normally distributed data sets. One-way ANOVA was used, where appropriate, to compare >2 data sets. P<0.05 was considered to be statistically significant. All data were shown as mean±SE of mean.
For further information of the methodology, please see the Online Data Supplement.
Results
Cardiac Deterioration in 12-Month-Old G3 Mice and 24-Month-Old WT Mice
Previous study demonstrated that late generation of Terc−/− mice developed an impaired cardiac function because of repressed PGC1α expression.9 Indeed, distinct PGC1α expression patterns were found between WT and G3 hearts during aging. In contrast to relatively stable PGC1α level between 3- and 12-month-old WT hearts, a significant reduced PGC1α expression (P<0.05) was seen in 12-month-old G3 heart versus its young counterpart (Figure 1A), whereas WT hearts exhibited a decreased PGC1α level at 24 months old (P<0.05; Online Figure I), coincided with augmented reactive oxygen species (ROS) production (P<0.01; Online Figure I), increased myh7-to-myh6 ratio (P<0.001; Online Figure I), and hypertropic cardiac dysfunction (P<0.01; Online Figure I). Notably, 12-month-old G3 heart exhibited increased heart-weight-to-body-weight ratio (P<0.001; Figure 1B) and ROS production (P<0.001; Figure 1C) versus that of age-matched WT heart. The functional defects of G3 heart were further substantiated by impaired mitochondrial respiration (Figure 1D), concurrent with augmented cardiac stress indices and elevated inflammatory related gene expression (Figure 1E). In line with this, we documented a significant fibrosis (Figure 1F) in 12-month-old G3 heart compared with WT heart. Intriguingly, we also observed an increased NADH-to-NAD+ (nicotinamide adenine dinucleotide) ratio (P<0.01; Online Figure II) and more protein aggregates (Online Figure II) in aged G3 cardiomyocytes. These data indicate that downregulation of PGC1α is associated with accelerated cardiac deterioration in aged WT and G3 mice.
PGC1αOE Improves Mitochondrial and Cardiac Function in Young WT and G3 Hearts
The above profiles prompted assessment of whether elevating PGC1α level could attenuate aging-dependent structural and functional deterioration in old WT and G3 heart. Given that previous studies have shown cardiomyopathy could be induced via a boost of PGC1α expression,22,23 it is of particular interest to compare the biological effect of mild cardiac-specific PGC1α overexpression between WT and telomere dysfunctional mice within a physiological range. To this end, we engineered a cardiac-specific PGC1α knock-in WT (WT PGC1αOE) and Terc−/− mouse (G3 PGC1αOE), respectively, where PGC1α was under transcriptional control of the endogenous ROSA26 promoter (Online Figure III). This strategy moderately increased PGC1α expression in WT and G3 hearts (Online Figure III), and electron microscopy (Figure 2A and 2B) and mitochondrial staining (Online Figure IV) revealed that cardiac PGC1α knock in increased mitochondrial mass with normal mitochondrial morphology in both 3-month-old WT and G3 hearts, although G3 hearts exhibited a slightly higher ROS production (Figure 2C). In line with this, a significant enhanced basal and maximal oxygen consumption rate (Figure 2D) was found in both WT PGC1αOE and G3 PGC1αOE cardiomyocytes versus their respective counterparts. Interestingly, such moderate elevation of PGC1α expression is sufficient to improve cardiac function (Figure 2E through 2G) and exercise tolerance (Figure 2H). These results demonstrate that in young stage, moderate PGC1αOE is favorable to enhance mitochondrial biogenesis and function without causing adverse effect seen in previous PGC1αOE models.
Deteriorated Cardiac Function and Reduced Life Span in WT PGC1αOE Mice
We next examined whether aforementioned salutary effects in young mice can maintain to an old age, which is crucial for cardiac homeostasis because the abundant mitochondria residing in the heart accumulate more damage during aging. We first subjected WT and WT PGC1αOE mice to echocardiographic assessment (Figure 3A). Surprisingly, 12-month-old WT PGC1αOE mice exhibited an accelerated cardiac dysfunction as evidenced by lowered ejection fraction and fractional shortening (Figure 3B), concurrent with thinner systolic interventricular septum diameter and left ventricular posterior wall thickness, while a dilated left ventricular internal dimension (Figure 3C). These altered parameters echoed with a significant increase in heart-weight-to-body-weight ratio (P<0.01; Figure 3D), as well as the cardiac stress-related gene expressions (Figure 3E). The survival analysis indicated that a dramatically shortened life span occurred in WT PGC1αOE mice with the median survival of only 1 year (P<0.001; Figure 3F).
Deteriorated Mitochondrial Function in WT PGC1αOE Heart During Aging
We investigated the mechanism of PGC1αOE-induced cardiac dysfunction and premature death in aged mice. Electron microscopy analyses revealed that 12-month-old WT PGC1αOE heart contained a portion of distorted mitochondria, with enlarged cristae compartments in comparison to that of the age-matched WT mice (Figure 4A). Mitochondrial ROS level was also significantly increased in 12-month-old WT PGC1αOE cardiomyocytes (P<0.01; Figure 4B), accompanied by a reduced oxygen consumption rate (Figure 4C), although the ATP production was unchanged (Figure 4D). In line with these findings, spare respiration capacity and coupling efficiency were both decreased, while an augmented proton leak was seen in 12-month-old WT PGC1αOE cardiomyocytes (Figure 4E). These mitochondrial defects linked to reduced autophagy and citrate cycle activity in 12-month-old WT PGC1αOE heart as analyzed by RNA sequencing (Online Figure V). Indeed, Western blotting of autophagy-related proteins, for example, p62 and LC3B-II, confirmed a reduced mitochondrial autophagy (aka mitophagy) in old WT PGC1αOE heart versus young counterpart (Figure 4F). To verify whether mitophagy is required to maintain the mitochondrial fitness in WT PGC1αOE heart, we utilized thapsigargin—an autophagy blocker27—to treat 3-month-old WT and WT PGC1αOE cardiomyocytes and found a significant increased ROS production in WT PGC1αOE but not in WT cardiomyocytes (Figure 4G). Similar finding was seen using another autophagy inhibitor 3-methyladenine (Figure 4H). In line with this, an 8-week 3-methyladenine treatment significantly accelerated cardiac dysfunction in WT PGC1aOE mice but not in WT controls (Online Figure VI). In vitro mitochondrial functional characterization also confirmed that 3-methyladenine treatment lowered ATP production in 3-month-old WT PGC1aOE cardiomyocytes, whereas minor changes were observed in WT control cardiomyocytes (Online Figure VI), further strengthening the causality linking mitochondrial alterations with cardiac dysfunction in WT PGC1aOE mice. In addition, given that rapamycin has shown the ability to induce mitophagy,28 we further explored the mitophagic capacity by incubating 12-month-old cardiomyocytes with 200 nmol/L rapamycin. While rapamycin-treated WT cardiomyocytes showed a positive response after a 12-hour incubation, WT PGC1αOE cardiomyocytes failed to stimulate or even inhibition of autophagy to some extent (Figure 4I). Taken together, these findings suggest that impaired mitophagic capacity in old WT PGC1αOE heart may account for its cardiac compromise and premature death.
Cardiac PGC1α Induction Preserves Cardiac Function and Extends Health Span in G3 Mice
In contrast to the deteriorating effect of PGC1αOE in 12-month-old WT mice, a normalized heart-weight-to-body-weight ratio was seen in G3 PGC1αOE mice (Figure 5A), along with the PGC1α expression restored to a level comparable to WT mice (Online Figure VII). We next subjected aged G3 and G3 PGC1αOE mice to echocardiographic assessment. G3 PGC1αOE mice exhibited an improved cardiac function compared with G3 mice (Figure 5B), including enhanced left ventricular ejection fraction and fraction shortening (Figure 5C), as well as reduced left ventricular dilatation and left ventricular posterior wall thickness (Figure 5D). Consistently, analyses of RNA sequencing results revealed downregulation of dilated cardiomyopathy–related pathways after cardiac PGC1α induction (Online Figure VIII; Online Table I). Despite the absence of proliferative or apoptotic changes in all heart samples examined, a spectrum of cardiac degenerative markers were significantly declined in G3 PGC1αOE hearts (Figure 5E), suggesting PGC1α attenuated cardiac dysfunction of G3 mice during aging. Indeed, aged (≈350 days old) G3 mice displayed many typical degenerative indications, such as senile plaques, kyphosis, hair loss (Online Figure IX), and accelerated body mass loss (Online Figure IX), whereas age-matched G3 PGC1αOE mice appeared healthier with an overall improved fitness. Indeed, a 15-minute run test demonstrated that aged G3 PGC1αOE mice were as exercise tolerant as WT mice, both significantly better than G3 mice (Figure 5F). Consistent with above findings, a highly significant increase in median life span was observed in G3 PGC1αOE cohort compared with that of G3 cohort (Figure 5G; Online Figure X), indicating that cardiac PGC1α knock in extends health span in G3 mice.
Sustained Mitochondrial Function and Calmed Inflammation in G3 PGC1αOE Mice During Aging
To further explore the underlying mechanism of PGC1αOE-mediated beneficial effects in G3 mice, we performed additional analyses. RNA sequencing of aged G3 and G3 PGC1αOE hearts using differential gene expression analysis revealed an upregulation of oxidative phosphorylation pathways in comparison to G3 hearts (Online Figure XI; Online Table I). Moreover, transmission electron microscopy showed reinstated mitochondrial morphology and restored mtDNA content in aged G3 PGC1αOE hearts (Figure 6A and 6B), concurrent with improved mitochondrial respiration (Figure 6C), lowered ROS production (Figure 6D), and calmed inflammatory cytokine secretion (Figure 6E through 6G). These results suggest that PGC1α knock in ameliorates telomere deficiency–induced mitochondrial dysfunction. Altogether, these data reinforce that therapeutic regulation of PGC1α in cardiac aging should aim at achieving moderate induction of PGC1α within a therapeutically beneficial window.
Discussion
Here, we document distinct biological effects of cardiac-specific PGC1α overexpression in WT and a later generation of Terc−/− mice, respectively, in which telomere dysfunctional heart exhibits a lower basal PGC1α expression in comparison to the telomere-intact counterpart. Notably, we found accelerated cardiac degeneration and significantly shortened life span in WT PGC1αOE mice, while a favorable longevity-extending effect in G3 PGC1αOE mice despite the fact that the PGC1α knock in was restricted to the heart. We believe that the contrasting consequence in our models is at least partially due to the dose effect of PGC1α on tuning mitochondrial biogenesis and clearance. That is, full functional mitochondrial quality control is engaged in WT heart of normal PGC1α expression. In aged G3 heart of low PGC1α expression, however, energy deficit occurs because of the fact that damaged mitochondria exceed mitochondrial biogenesis. Much more complex is that mitochondrial biogenesis exceeds energy demand in WT PGC1αOE heart, where mitophagy is engaged to eliminate excessive mitochondria during young, but impaired mitophagy and subsequently increased oxidative stress worsen the cardiac function of 12-month-old WT PGC1αOE mice (Figure 7).
Aging-induced mitochondrial dysfunction leads to an increase in ROS production and reduced oxidative phosphorylation, thereby decreasing ATP synthesis and cell respiration. Interestingly, mitochondrial function of Terc−/− iPSCs (induced pluripotent stem cells) and their differentiated derivatives was severely impaired, while mitochondrial function in Terc−/− ntESCs (nuclear transferred embryonic stem cells) was considerably improved, with PGC1α a possible target.29 Other experimental attempts in different systems, such as tissue-specific overexpression of PGC1α in Drosophila stem and progenitor cells within the digestive tract, extended the life span of this organism.30 However, forced expression of PGC1α driven by a cardiac αMHC promoter or an inducible Tet-On system in the heart developed cardiomyopathy,22,23 indicating that a fine-tuned expression of PGC1α and relevant downstream signaling molecules is essential for mitochondrial quality control and proper mitochondrial functioning. In our PGC1α knock-in G3 mouse model, we unveiled multiple beneficial aspects of PGC1α induction in the absence of previous documented cardiomyopathy. This difference could be due first to the fact that we used a ROSA26 locus knock-in approach to overexpress mildly one copy of PGC1α; hence, the mild elevation is completely within the physiological range. Second, while the aged G3 heart showed decreased PGC1α expression, the G3 PGC1αOE heart exhibited a PGC1α level comparable to that of WT hearts. In the current study, we did not overexpress 2 copies of PGC1α in G3 hearts as we were limited by the mating strategy. Further investigation of the dose effect of PGC1α on cardiac function and maximal life span in G3 mice would be of great interest.
Given the relatively low mitochondrial dynamics proven in heart,31,32 a logical regulatory hub for the maintenance of its mitochondrial homeostasis lies in the mitophagy, the scavenger of redundant or damaged mitochondria.5,33 Mitophagy is responsible for both coordinating the metabolic reprogramming of heart during maturation34 and for the suppression of aging-associated inflammation through leaking mtDNA-induced cGAS (cyclic GMP-AMP synthase)/STING (stimulator of interferon genes) activation.35 Several pieces of evidence have implicated that regulation of mitophagy could affect cardiac function and life span of the organism. For instance, investigators have reported the salutary effect of natural polyamine spermidine supplement in promoting cardiac performance and extending life span in mice via activating mitophagy.36 In contrast, ablation of autophagy-related gene leads to cardiomyopathy.37,38 In the present study, we saw an elevated mitophagy in WT PGC1αOE heart at 3 months old but reduced mitophagy at the age of 12 months old, which coincides with the changes in mitochondrial respiration and cardiac function of WT PGC1αOE mice. Despite we cannot exclude that ROS elevation, contractility alterations related to mitochondrial number expansion, and other possibilities lead to cardiomyopathy in WT PGC1αOE mice, current finding implicates mitophagy may be essential for maintaining the equilibrium of PGC1α-mediated mitochondrial biogenesis and ROS production within a physiological-tolerant range. If so, a well-orchestrated activation of both PGC1α and mitophagy in heart is worth testing in eliminating adverse effects seen in old WT PGC1αOE mice and possibly in previous cardiac PGC1α overexpression models.
Apart from known regulation of thermogenesis, mitochondrial biogenesis, respiration, fatty acid oxidation, and antioxidative effect, mounting studies have implicated that PGC1α may involve other physiological functions.39–42 Of note, direct evidence from Tran et al43 indicates that PGC1α is capable of driving de novo nicotinamide adenine dinucleotide biosynthesis and, therefore, enhancing stress resistance. In line with this, we also documented a restored NAD+ level concurrent with reduced inflammation and protein aggregates in G3 PGC1αOE hearts. This effect is reminiscent of the reduction of protein aggregates and improved cognitive function that occurs with NAD+ supplementation in animal models of Alzheimer disease.44 These observations warrant further investigation to clarify how NAD+ and PGC1α influence inflammation and proteostasis in the heart.
There are some limitations of the study. First, the goal of this study was to evaluate the dosage effect of PGC1α overexpression in murine models. We cannot exclude the possibility that a different phenotype of PGC1α overexpression would occur in primates and humans. Given that laboratory mice have long telomeres as compared with humans do, it is obligatory to test the biological function of PGC1α overexpression in mice with shorter telomeres to extrapolate these findings to human aging. Our study provides the proof of principle that moderately boosting PGC1α level could restore mitochondrial function and thereby rejuvenate the cardiac aging in the presence of short telomeres. Nevertheless, the cause-effect links between telomerase deficiency, mitochondrial dysfunction, and cardiomyopathy require further investigation. We cannot exclude that the cardiomyopathy could be driven by a separate primary process with secondary mitochondrial dysfunction that is partially restored by PGC1α overexpression (thus energetic and functional improvement) versus a direct connection between telomerase function, mitochondrial function, and PGC1α activity. Second, we acknowledge the pitfall in the assessment of cardiac function, including ejection fraction and fractioning shortening that rely on M-mode analysis of the short-axis view of echocardiography, depends on geometric assumptions. Third, our survival study included both male and female animals. Although we did not observe a significant difference between male versus female mice, future investigations should consider the impact of sex on the regulatory effect of PGC1α overexpression on cardiac function and longevity. Finally, a few challenges ahead remain to tackle with, before PGC1a therapy becomes clinically available. For instance, directly upregulating or downregulating the expression of miscellaneous PGC1a may cause a series of biological effects, rather than simply changing the mitochondrial volume and respiration. Therefore, it is mandatory to carefully consider the dose and time period of PGC1a-boosting strategy after a comprehensive assessment of the overall mitochondrial fitness (ie, mitochondrial abundance, quality control, mitophagy capacity) to achieve an optimal clinical translation of the PGC1a therapy.
Taken together, our study suggests that fine-tuning the expression of PGC1α is crucial for the cardiac homeostasis because the balance between mitochondrial biogenesis and clearance is vital for regulating mitochondrial homeostasis. Given the uncertainty in PGC1α level under various pathological conditions, these results reinforce the importance of carefully evaluating the PGC1α-boosting strategies in a context-dependent manner to facilitate clinical translation of novel cardioprotective therapies.
Acknowledgments
We thank Stephanie C. Tribuna for secretarial assistance, Yaoli Deng and all staffs in the Center of Experimental Animals, Hangzhou Normal University, for animal caretaking, Li Wang, Beibei Wang, and Ping Yang in the Center of Cyro-Electron Microscopy, Zhejiang University for their technical assistance on transmission electron microscopy analyses, Jiabin Lin and Jing Zhao for mouse echocardiography, Qingtao Hu for RNA sequencing data analysis, and all members of Ju’s Lab, particularly Weiwei Yi and Xianda Chen for flow cytometry analysis, and Fan Yang for critical insights and suggestions.
Novelty and Significance
•
PGC1α (peroxisome proliferator-activated receptor gamma coactivator 1α) is involved in the regulation of mitochondrial biogenesis and function.
•
Age-associated telomere attrition is linked to cardiac dysfunction, with PGC1α as a possible target.
•
Attempts to boost PGC1α levels as a therapeutic strategy has had contradictory results.
•
Moderate cardiac PGC1α overexpression is sufficient to revitalize mitochondrial and cardiac function in G3Terc (third-generation telomerase deficient) mice.
•
However, PGC1α overexpression in telomere-intact mice leads to accelerated cardiac aging and a significantly shortened life span.
•
The fine-tuning of PGC1α level is crucial for the mitochondrial and cardiac homeostasis partially via the balance between mitochondrial biogenesis and clearance.
Although PGC1α has been studied extensively within the context of energy metabolism and mitochondrial function in the heart, multiple attempts have failed to upregulate PGC1α expression as a therapy. By using a ROSA26 locus knock-in approach to mildly overexpress PGC1α in cardiomyocytes, we observed the consequences of cardiac PGC1α overexpression in the telomere-intact and telomere dysfunctional mouse models, in which telomerase-deficient heart exhibits a lower basal PGC1α expression in comparison to the telomere-intact counterpart. While WT (wild type) mice exhibited a relatively consistent PGC1α expression and mitochondrial abundance, overexpression of PGC1α accelerated cardiac degeneration and significantly shortened life span in WT mice at least partially due to ROS insult and perturbed mitophagy. By contrast, in hearts of third-generation mice, which show reduced PGC1α expression with advanced age, cardiac-specific PGC1α knock in normalized the PGC1α level comparable to that of WT mice and attenuated mitochondrial dysfunction, thereby preserving cardiac performance and extending life span. Our study suggests that fine-tuning the expression of PGC1α is crucial for cardiac homeostasis, and synergistic activation of mitophagy in PGC1α-enhancing strategies may have a role in cardioprotective therapies.
Footnote
Nonstandard Abbreviations and Acronyms
- αMHC
- α-myosin heavy chain
- G3
- third generation
- G3Terc−/−
- third-generation telomerase deficient
- PGC1α
- peroxisome proliferator-activated receptor gamma coactivator 1α
- ROS
- reactive oxygen species
- Terc−/−
- telomerase deficient
- WT
- wild type
Supplemental Material
References
1.
van Berlo JH, Kanisicak O, Maillet M, Vagnozzi RJ, Karch J, Lin SC, Middleton RC, Marbán E, Molkentin JD. c-kit+ cells minimally contribute cardiomyocytes to the heart. Nature. 2014;509:337–341. doi: 10.1038/nature13309
2.
Sultana N, Zhang L, Yan J, Chen J, Cai W, Razzaque S, Jeong D, Sheng W, Bu L, Xu M, et al. Resident c-kit(+) cells in the heart are not cardiac stem cells. Nat Commun. 2015;6:8701. doi: 10.1038/ncomms9701
3.
Liu Q, Yang R, Huang X, Zhang H, He L, Zhang L, Tian X, Nie Y, Hu S, Yan Y, et al. Genetic lineage tracing identifies in situ Kit-expressing cardiomyocytes. Cell Res. 2016;26:119–130. doi: 10.1038/cr.2015.143
4.
He L, Li Y, Li Y, Pu W, Huang X, Tian X, Wang Y, Zhang H, Liu Q, Zhang L, et al. Enhancing the precision of genetic lineage tracing using dual recombinases. Nat Med. 2017;23:1488–1498. doi: 10.1038/nm.4437
5.
Shirakabe A, Ikeda Y, Sciarretta S, Zablocki DK, Sadoshima J. Aging and autophagy in the heart. Circ Res. 2016;118:1563–1576. doi: 10.1161/CIRCRESAHA.116.307474
6.
Abdellatif M, Sedej S, Carmona-Gutierrez D, Madeo F, Kroemer G. Autophagy in cardiovascular aging. Circ Res. 2018;123:803–824. doi: 10.1161/CIRCRESAHA.118.312208
7.
Luongo TS, Lambert JP, Gross P, Nwokedi M, Lombardi AA, Shanmughapriya S, Carpenter AC, Kolmetzky D, Gao E, van Berlo JH, et al. The mitochondrial Na+/Ca2+ exchanger is essential for Ca2+ homeostasis and viability. Nature. 2017;545:93–97. doi: 10.1038/nature22082
8.
Wai T, García-Prieto J, Baker MJ, Merkwirth C, Benit P, Rustin P, Rupérez FJ, Barbas C, Ibañez B, Langer T. Imbalanced OPA1 processing and mitochondrial fragmentation cause heart failure in mice. Science. 2015;350:aad0116. doi: 10.1126/science.aad0116
9.
Sahin E, Colla S, Liesa M, Moslehi J, Müller FL, Guo M, Cooper M, Kotton D, Fabian AJ, Walkey C, et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature. 2011;470:359–365. doi: 10.1038/nature09787
10.
Finck BN, Kelly DP. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) regulatory cascade in cardiac physiology and disease. Circulation. 2007;115:2540–2548. doi: 10.1161/CIRCULATIONAHA.107.670588
11.
Soyal S, Krempler F, Oberkofler H, Patsch W. PGC-1alpha: a potent transcriptional cofactor involved in the pathogenesis of type 2 diabetes. Diabetologia. 2006;49:1477–1488. doi: 10.1007/s00125-006-0268-6
12.
Shokouhi S, Haghani K, Borji P, Bakhtiyari S. Association between PGC-1alpha gene polymorphisms and type 2 diabetes risk: a case-control study of an Iranian population. Can J Diabetes. 2015;39:65–72. doi: 10.1016/j.jcjd.2014.05.003
13.
Jiang WG, Douglas-Jones A, Mansel RE. Expression of peroxisome-proliferator activated receptor-gamma (PPARgamma) and the PPARgamma co-activator, PGC-1, in human breast cancer correlates with clinical outcomes. Int J Cancer. 2003;106:752–757. doi: 10.1002/ijc.11302
14.
Feilchenfeldt J, Bründler MA, Soravia C, Tötsch M, Meier CA. Peroxisome proliferator-activated receptors (PPARs) and associated transcription factors in colon cancer: reduced expression of PPARgamma-coactivator 1 (PGC-1). Cancer Lett. 2004;203:25–33. doi: 10.1016/j.canlet.2003.08.024
15.
Zhang Y, Ba Y, Liu C, Sun G, Ding L, Gao S, Hao J, Yu Z, Zhang J, Zen K, et al. PGC-1alpha induces apoptosis in human epithelial ovarian cancer cells through a PPARgamma-dependent pathway. Cell Res. 2007;17:363–373. doi: 10.1038/cr.2007.11
16.
Wang S, Fu C, Wang H, Shi Y, Xu X, Chen J, Song X, Sun K, Wang J, Fan X, et al. Polymorphisms of the peroxisome proliferator-activated receptor-gamma coactivator-1alpha gene are associated with hypertrophic cardiomyopathy and not with hypertension hypertrophy. Clin Chem Lab Med. 2007;45:962–967. doi: 10.1515/CCLM.2007.189
17.
Sihag S, Cresci S, Li AY, Sucharov CC, Lehman JJ. PGC-1alpha and ERRalpha target gene downregulation is a signature of the failing human heart. J Mol Cell Cardiol. 2009;46:201–212. doi: 10.1016/j.yjmcc.2008.10.025
18.
Kulikova TG, Stepanova OV, Voronova AD, Valikhov MP, Sirotkin VN, Zhirov IV, Tereshchenko SN, Masenko VP, Samko AN, Sukhikh GT. Pathological remodeling of the myocardium in chronic heart failure: role of PGC-1α. Bull Exp Biol Med. 2018;164:794–797. doi: 10.1007/s10517-018-4082-1
19.
Handschin C, Chin S, Li P, Liu F, Maratos-Flier E, Lebrasseur NK, Yan Z, Spiegelman BM. Skeletal muscle fiber-type switching, exercise intolerance, and myopathy in PGC-1alpha muscle-specific knock-out animals. J Biol Chem. 2007;282:30014–30021. doi: 10.1074/jbc.M704817200
20.
Miura S, Kai Y, Ono M, Ezaki O. Overexpression of peroxisome proliferator-activated receptor gamma coactivator-1alpha down-regulates GLUT4 mRNA in skeletal muscles. J Biol Chem. 2003;278:31385–31390. doi: 10.1074/jbc.M304312200
21.
Arany Z, He H, Lin J, Hoyer K, Handschin C, Toka O, Ahmad F, Matsui T, Chin S, Wu PH, et al. Transcriptional coactivator PGC-1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metab. 2005;1:259–271. doi: 10.1016/j.cmet.2005.03.002
22.
Russell LK, Mansfield CM, Lehman JJ, Kovacs A, Courtois M, Saffitz JE, Medeiros DM, Valencik ML, McDonald JA, Kelly DP. Cardiac-specific induction of the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha promotes mitochondrial biogenesis and reversible cardiomyopathy in a developmental stage-dependent manner. Circ Res. 2004;94:525–533. doi: 10.1161/01.RES.0000117088.36577.EB
23.
Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, Kelly DP. Peroxisome proliferator-activated receptor gamma coactivator-1 promotes cardiac mitochondrial biogenesis. J Clin Invest. 2000;106:847–856. doi: 10.1172/JCI10268
24.
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194–1217. doi: 10.1016/j.cell.2013.05.039
25.
Wiley CD, Velarde MC, Lecot P, Liu S, Sarnoski EA, Freund A, Shirakawa K, Lim HW, Davis SS, Ramanathan A, et al. Mitochondrial dysfunction induces senescence with a distinct secretory phenotype. Cell Metab. 2016;23:303–314. doi: 10.1016/j.cmet.2015.11.011
26.
He W, Cowin P, Stokes DL. Untangling desmosomal knots with electron tomography. Science. 2003;302:109–113. doi: 10.1126/science.1086957
27.
Ganley IG, Wong PM, Gammoh N, Jiang X. Distinct autophagosomal-lysosomal fusion mechanism revealed by thapsigargin-induced autophagy arrest. Mol Cell. 2011;42:731–743. doi: 10.1016/j.molcel.2011.04.024
28.
Civiletto G, Dogan SA, Cerutti R, Fagiolari G, Moggio M, Lamperti C, Beninca C, Viscomi C, Zeviani M. Rapamycin rescues mitochondrial myopathy via coordinated activation of autophagy and lysosomal biogenesis. EMBO Mol Med. 2018;10:e8799.
29.
Le R, Kou Z, Jiang Y, Li M, Huang B, Liu W, Li H, Kou X, He W, Rudolph KL, et al. Enhanced telomere rejuvenation in pluripotent cells reprogrammed via nuclear transfer relative to induced pluripotent stem cells. Cell Stem Cell. 2014;14:27–39. doi: 10.1016/j.stem.2013.11.005
30.
Rera M, Bahadorani S, Cho J, Koehler CL, Ulgherait M, Hur JH, Ansari WS, Lo T Jones DL, Walker DW. Modulation of longevity and tissue homeostasis by the Drosophila PGC-1 homolog. Cell Metab. 2011;14:623–634. doi: 10.1016/j.cmet.2011.09.013
31.
Dorn GW Vega RB, Kelly DP. Mitochondrial biogenesis and dynamics in the developing and diseased heart. Genes Dev. 2015;29:1981–1991. doi: 10.1101/gad.269894.115
32.
Vega RB, Kelly DP. Cardiac nuclear receptors: architects of mitochondrial structure and function. J Clin Invest. 2017;127:1155–1164. doi: 10.1172/JCI88888
33.
Chen Y, Dorn GW. PINK1-phosphorylated mitofusin 2 is a Parkin receptor for culling damaged mitochondria. Science. 2013;340:471–475. doi: 10.1126/science.1231031
34.
Gong G, Song M, Csordas G, Kelly DP, Matkovich SJ, Dorn GW Parkin-mediated mitophagy directs perinatal cardiac metabolic maturation in mice. Science. 2015;350:aad2459. doi: 10.1126/science.aad2459
35.
Sliter DA, Martinez J, Hao L, Chen X, Sun N, Fischer TD, Burman JL, Li Y, Zhang Z, Narendra DP, et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature. 2018;561:258–262. doi: 10.1038/s41586-018-0448-9
36.
Eisenberg T, Abdellatif M, Schroeder S, Primessnig U, Stekovic S, Pendl T, Harger A, Schipke J, Zimmermann A, Schmidt A, et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22:1428–1438. doi: 10.1038/nm.4222
37.
Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I, Matsumura Y, Asahi M, et al. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med. 2007;13:619–624. doi: 10.1038/nm1574
38.
Wang B, Nie J, Wu L, Hu Y, Wen Z, Dong L, Zou MH, Chen C, Wang DW. AMPKα2 protects against the development of heart failure by enhancing mitophagy via PINK1 phosphorylation. Circ Res. 2018;122:712–729. doi: 10.1161/CIRCRESAHA.117.312317
39.
Handschin C, Spiegelman BM. The role of exercise and PGC1alpha in inflammation and chronic disease. Nature. 2008;454:463–469. doi: 10.1038/nature07206
40.
Wu J, Ruas JL, Estall JL, Rasbach KA, Choi JH, Ye L, Boström P, Tyra HM, Crawford RW, Campbell KP, et al. The unfolded protein response mediates adaptation to exercise in skeletal muscle through a PGC-1α/ATF6α complex. Cell Metab. 2011;13:160–169. doi: 10.1016/j.cmet.2011.01.003
41.
Torrano V, Valcarcel-Jimenez L, Cortazar AR, Liu X, Urosevic J, Castillo-Martin M, Fernández-Ruiz S, Morciano G, Caro-Maldonado A, Guiu M, et al. The metabolic co-regulator PGC1α suppresses prostate cancer metastasis. Nat Cell Biol. 2016;18:645–656. doi: 10.1038/ncb3357
42.
Luo C, Lim JH, Lee Y, Granter SR, Thomas A, Vazquez F, Widlund HR, Puigserver P. A PGC1α-mediated transcriptional axis suppresses melanoma metastasis. Nature. 2016;537:422–426. doi: 10.1038/nature19347
43.
Tran MT, Zsengeller ZK, Berg AH, Khankin EV, Bhasin MK, Kim W, Clish CB, Stillman IE, Karumanchi SA, Rhee EP, et al. PGC1α drives NAD biosynthesis linking oxidative metabolism to renal protection. Nature. 2016;531:528–532. doi: 10.1038/nature17184
44.
Sorrentino V, Romani M, Mouchiroud L, Beck JS, Zhang H, D’Amico D, Moullan N, Potenza F, Schmid AW, Rietsch S, et al. Enhancing mitochondrial proteostasis reduces amyloid-β proteotoxicity. Nature. 2017;552:187–193. doi: 10.1038/nature25143
Information & Authors
Information
Published In
Copyright
© 2019 American Heart Association, Inc.
Versions
You are viewing the most recent version of this article.
History
Received: 11 June 2019
Revision received: 10 August 2019
Accepted: 14 August 2019
Published online: 15 August 2019
Published in print: 13 September 2019
Keywords
Subjects
Authors
Disclosures
None.
Sources of Funding
This work was supported by grants from the National Natural Science Foundation of China (91749203, 81525010, and 81420108017) and the National Key Research and Development Program of China (2017YFA0103302), Innovative Team Program of Guangzhou Regenerative Medicine and Health Guangdong Laboratory (2018GZR110103002), and the Program for Guangdong Introducing Innovative and Entrepreneurial Teams (2017ZT07S347) to Z. Ju, the National Natural Science Foundation of China (81400221), and Hangzhou Normal University (PF14002004017) to X. Zhu, and grants from the EPFL, Systems X (SySX.ch 2013/153), the Velux Stiftung (1019), and the Swiss National Science Foundation (31003A-140780) to J. Auwerx, and grants from the US National Institutes of Health (HL061795, HG007690, and GM107618), and the American Heart Association (D700382) to J. Loscalzo, the National Natural Science Foundation of China (81771520) and Science Technology Department of Zhejiang Province (2016C34002) and Health Bureau of Zhejiang Province (2015DTA001) to G. Mao, and grants from the Science and Technology Commission of Shanghai Municipality (17DZ2273200/16DZ2290900).
Metrics & Citations
Metrics
Citations
Download Citations
If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.
- The transcriptional repressor HEY2 regulates mitochondrial oxidative respiration to maintain cardiac homeostasis, Nature Communications, 16, 1, (2025).https://doi.org/10.1038/s41467-024-55557-4
- Adipocyte-derived small extracellular vesicles exacerbate diabetic ischemic heart injury by promoting oxidative stress and mitochondrial-mediated cardiomyocyte apoptosis, Redox Biology, 79, (103443), (2025).https://doi.org/10.1016/j.redox.2024.103443
- From metabolism to malignancy: the multifaceted role of PGC1α in cancer, Frontiers in Oncology, 14, (2024).https://doi.org/10.3389/fonc.2024.1383809
- Aging‐associated decrease of PGC ‐1α promotes pain chronification , Aging Cell, 23, 8, (2024).https://doi.org/10.1111/acel.14177
- Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases, Signal Transduction and Targeted Therapy, 9, 1, (2024).https://doi.org/10.1038/s41392-024-01756-w
- Exercise mitigates reductive stress-induced cardiac remodeling in mice, Redox Biology, 75, (103263), (2024).https://doi.org/10.1016/j.redox.2024.103263
- Targeting organ-specific mitochondrial dysfunction to improve biological aging, Pharmacology & Therapeutics, 262, (108710), (2024).https://doi.org/10.1016/j.pharmthera.2024.108710
- Cardiomyocyte LGR6 alleviates ferroptosis in diabetic cardiomyopathy via regulating mitochondrial biogenesis, Metabolism, 159, (155979), (2024).https://doi.org/10.1016/j.metabol.2024.155979
- Aging in Heart Failure, JACC: Heart Failure, 12, 5, (795-809), (2024).https://doi.org/10.1016/j.jchf.2024.02.021
- Insights into the post-translational modifications in heart failure, Ageing Research Reviews, 100, (102467), (2024).https://doi.org/10.1016/j.arr.2024.102467
- See more
Loading...
View Options
Login options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginPurchase Options
Purchase this article to access the full text.
eLetters(0)
eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.
Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.