AARS2 ameliorates myocardial ischemia via fine-tuning PKM2-mediated metabolism

  1. Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, College of Future Technology, Academy for Advanced Interdisciplinary Studies, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
  2. School of Basic Medical Sciences and The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
  3. Obstetrics and Gynecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Ke Cheng
    Yongchuan Hospital of Chongqing Medical University, Chongqing, China
  • Senior Editor
    Olujimi Ajijola
    University of California, Los Angeles, Los Angeles, United States of America

Reviewer #1 (Public Review):

In this study, the authors introduced an essential role of AARS2 in maintaining cardiac function. They also investigated the underlying mechanism that through regulating alanine and PKM2 translation are regulated by AARS2. Accordingly, a therapeutic strategy for cardiomyopathy and MI was provided. Several points need to be addressed to make this article more comprehensive:

(1) Include apoptotic caspases in Figure 2B, and Figure 4 B and E as well.

(2) It would be better to show the change of apoptosis-related proteins upon the knocking down of AARS2 by small interfering RNA (siRNA).

(3) In Figure 5, the authors performed Mass Spectrometry to assess metabolites of homogenates. I was wondering if the change of other metabolites could be provided in the form of a heatmap.

(4) The amounts of lactate should be accessed using a lactate assay kit to validate the Mass Spectrometry results.

(5) How about the expression pattern of PKM2 before and after mouse MI. Furtherly, the correlation between AARS2 and PKM2?

(6) In Figure 5, how about the change of apoptosis-related proteins after administration of PKM2 activator TEPP-46?

Reviewer #2 (Public Review):

Summary:

The authors aimed to elucidate the role of AARS2, an alanyl-tRNA synthase, in mouse hearts, specifically its impact on cardiac function, fibrosis, apoptosis, and metabolic pathways under conditions of myocardial infarction (MI). By investigating the effects of both deletion and overexpression of AARS2 in cardiomyocytes, the study aims to determine how AARS2 influences cardiac health and survival during ischemic stress.

The authors successfully achieved their aims by demonstrating the critical role of AARS2 in maintaining cardiomyocyte function under ischemic conditions. The evidence presented, including genetic manipulation results, functional assays, and mechanistic studies, robustly supports the conclusion that AARS2 facilitates cardiomyocyte survival through PKM2-mediated metabolic reprogramming. The study convincingly links AARS2 overexpression to improved cardiac outcomes post-MI, validating the proposed protective AARS2-PKM2 signaling pathway.

This work may have a significant impact on the field of cardiac biology and ischemia research. By identifying AARS2 as a key player in cardiomyocyte survival and metabolic regulation, the study opens new avenues for therapeutic interventions targeting this pathway. The methods used, particularly the cardiomyocyte-specific genetic models and ribosome profiling, are valuable tools that can be employed by other researchers to investigate similar questions in cardiac physiology and pathology.

Understanding the metabolic adaptations in cardiomyocytes during ischemia is crucial for developing effective treatments for MI. This study highlights the importance of metabolic flexibility and the role of specific enzymes like AARS2 in facilitating such adaptations. The identification of the AARS2-PKM2 axis adds a new layer to our understanding of cardiac metabolism, suggesting that enhancing glycolysis can be a viable strategy to protect the heart from ischemic damage.

Strengths:

(1) Comprehensive Genetic Models: The use of cardiomyocyte-specific AARS2 knockout and overexpression mouse models allowed for precise assessment of AARS2's role in cardiac cells.

(2) Functional Assays: Detailed phenotypic analyses, including measurements of cardiac function, fibrosis, and apoptosis, provided evidence for the physiological impact of AARS2 manipulation.

(3) Mechanistic Insights: This study used ribosome profiling (Ribo-Seq) to uncover changes in protein translation, specifically highlighting the role of PKM2 in metabolic reprogramming.

(4) Therapeutic Relevance: The use of the PKM2 activator TEPP-46 to reverse the effects of AARS2 deficiency presents a potential therapeutic avenue, underscoring the practical implications of the findings.

Weaknesses:

(1) Species Limitation: The study is limited to mouse and rat models, and while these are highly informative, further validation in human cells or tissues would strengthen the translational relevance.

(2) Temporal Dynamics: The study does not extensively address the temporal dynamics of AARS2 expression and PKM2 activity during the progression of MI and recovery, which could offer deeper insights into the timing and regulation of these processes.

Reviewer #3 (Public Review):

In the present study, the author revealed that cardiomyocyte-specific deletion of mouse AARS2 exhibited evident cardiomyopathy with impaired cardiac function, notable cardiac fibrosis, and cardiomyocyte apoptosis. Cardiomyocyte-specific AARS2 overexpression in mice improved cardiac function and reduced cardiac fibrosis after myocardial infarction (MI), without affecting cardiomyocyte proliferation and coronary angiogenesis. Mechanistically, AARS2 overexpression suppressed cardiomyocyte apoptosis and mitochondrial reactive oxide species production, and changed cellular metabolism from oxidative phosphorylation toward glycolysis in cardiomyocytes, thus leading to cardiomyocyte survival from ischemia and hypoxia stress. Ribo-Seq revealed that AARS2 overexpression increased pyruvate kinase M2 (PKM2) protein translation and the ratio of PKM2 dimers to tetramers that promote glycolysis. Additionally, PKM2 activator TEPP-46 reversed cardiomyocyte apoptosis and cardiac fibrosis caused by AARS2 deficiency. Thus, this study demonstrates that AARS2 plays an essential role in protecting cardiomyocytes from ischemic pressure via fine-tuning PKM2-mediated energy metabolism, and presents a novel cardiac protective AARS2-PKM2 signaling during the pathogenesis of MI. This study provides some new knowledge in the field, and there are still some questions that need to be addressed in order to better support the authors' views.

(1) WGA staining showed obvious cardiomyocyte hypertrophy in the AARS2 cKO heart. Whether AARS affects cardiac hypertrophy needs to be further tested.

(2) The authors observed that AARS2 can improve myocardial infarction, and whether AARS2 has an effect on other heart diseases.

(3) Studies have shown that hypoxia conditions can lead to mitochondrial dysfunction, including abnormal division and fusion. AARS2 also affects mitochondrial division and fusion and interacts with mitochondrial proteins, including FIS and DRP1, the authors are suggested to verify.

(4) The authors only examined the role of AARS2 in cardiomyocytes, and fibroblasts are also an important cell type in the heart. Authors should examine the expression and function of AARS2 in fibroblasts.

(5) Overexpression of AARS2 can inhibit the production of mtROS, and has a protective effect on myocardial ischemia and H/ R-induced injury, and the occurrence of iron death is also closely related to ROS, whether AARS protects myocardial by regulating the occurrence of iron death?

(6) Please revise the English grammar and writing style of the manuscript, spelling and grammatical errors should be excluded.

(7) Recent studies have shown that a decrease in oxygen levels leads to an increase in AARS2, and lactic acid rises rapidly without being oxidized. Both of these factors inhibit oxidative phosphorylation and muscle ATP production by increasing mitochondrial lactate acylation, thereby inhibiting exercise capacity and preventing the accumulation of reactive oxygen species ROS. The key role of protein lactate acylation modification in regulating oxidative phosphorylation of mitochondria, and the importance of metabolites such as lactate regulating cell function through feedback mechanisms, i.e. cells adapt to low oxygen through metabolic regulation to reduce ROS production and oxidative damage, and therefore whether AARS2 in the heart also acts in this way.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation