Comprehensive Physiology Wiley Online Library

Lysosomes Mediate Benefits of Intermittent Fasting in Cardiometabolic Disease: The Janitor Is the Undercover Boss

Full Article on Wiley Online Library



ABSTRACT

Adaptive responses that counter starvation have evolved over millennia to permit organismal survival, including changes at the level of individual organelles, cells, tissues, and organ systems. In the past century, a shift has occurred away from disease caused by insufficient nutrient supply toward overnutrition, leading to obesity and diabetes, atherosclerosis, and cardiometabolic disease. The burden of these diseases has spurred interest in fasting strategies that harness physiological responses to starvation, thus limiting tissue injury during metabolic stress. Insights gained from animal and human studies suggest that intermittent fasting and chronic caloric restriction extend lifespan, decrease risk factors for cardiometabolic and inflammatory disease, limit tissue injury during myocardial stress, and activate a cardioprotective metabolic program. Acute fasting activates autophagy, an intricately orchestrated lysosomal degradative process that sequesters cellular constituents for degradation, and is critical for cardiac homeostasis during fasting. Lysosomes are dynamic cellular organelles that function as incinerators to permit autophagy, as well as degradation of extracellular material internalized by endocytosis, macropinocytosis, and phagocytosis. The last decade has witnessed an explosion of knowledge that has shaped our understanding of lysosomes as central regulators of cellular metabolism and the fasting response. Intriguingly, lysosomes also store nutrients for release during starvation; and function as a nutrient sensing organelle to couple activation of mammalian target of rapamycin to nutrient availability. This article reviews the evidence for how the lysosome, in the guise of a janitor, may be the “undercover boss” directing cellular processes for beneficial effects of intermittent fasting and restoring homeostasis during feast and famine. © 2018 American Physiological Society. Compr Physiol 8:1639‐1667, 2018.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Cardiac metabolism in health and disease. During normal physiology, the vast majority of myocardial ATP production is from oxidative phosphorylation. Of this, a majority is from β‐oxidation of fatty acids, with a smaller contribution from glucose oxidation. By contrast, glycolysis, ketone body oxidation, and amino acid metabolism contribute very small amounts to overall ATP generation. During pathological states, cardiac myocytes display alternate substrate preferences for energy generation (reviewed in (212,214); please see text under the section “Cardiac energetics, substrate metabolism and pathophysiology under fasting states” for details). (A) Metabolic changes during nutrient deprivation vary depending the duration (ST = short term and LT = long term). During short‐term fasting, fatty acid oxidation is unchanged while glucose metabolism is suppressed. In addition, marked increases in ketone body oxidation and amino acid metabolism contribute to myocardial energetics. During prolonged fasting, however, lipid oxidation is increased and serves as the main energetic source. (B) During ischemic syndromes (I = ischemia and IR = ischemia‐reperfusion), energetic choices are driven by oxygen availability. While ischemia is associated with reduction in fatty acid and glucose oxidation, glycolysis serves as a far more significant source of ATP. When reperfused, glycolysis rates decline and glucose oxidation is still suppressed, while fatty acid oxidation increases. In most ischemic syndromes, TCA (tricarboxylic acid cycle)‐independent amino acid metabolism is augmented. (C) Cardiac myocyte dysfunction in cardiomyopathy is often accompanied by abnormalities in myocardial metabolism and energetics. Prominent among these is a switch to a preference for glucose oxidation as opposed to lipid oxidation. (D) During aging, similar to cardiomyopathy, fatty acid oxidation is decreased with increases in glucose oxidation and glycolysis driving myocardial energetics. (E) Fatty acid utilization is increased in the diabetic myocardium (with type 2 diabetes due to insulin resistance) with reduction in glucose utilization. The diabetic heart increasingly utilizes ketone bodies as a nutrient source. “↑” indicates upregulation, whereas “↓” indicates downregulation.
Figure 2. Figure 2. Mechanisms for execution of autophagy as a lysosomal degradative pathway. Nutrient deprivation and organelle damage trigger macroautophagy in cardiac myocytes [reviewed in (67)]. Assembly of the phagophore (the initiation of the double membrane) is activated by class III phosphatidylinositol 3‐kinase (PtdIns3K) catalyzed generation of phosphatydil inositol 3‐phosphate (PtdIns3P) to recruit PtdIns3P‐binding proteins. A protein complex is formed comprised of Vps15 homolog phosphoinositide‐3‐kinase, regulatory subunit 4 (PIK3R4), the Vps34 homolog phosphatidylinositol 3‐kinase, catalytic subunit type 3 (PIK3C3), and the Vps30/Atg6 homolog Beclin 1. This interacts with AMBRA1 (not shown) and ATG14, and also contain the BECN1‐interacting protein UV radiation resistance associated (UVRAG, not shown) and SH3‐domain GRB2‐like endophilin B1 (SH3GLB1/Bif‐1, not shown) to promote autophagosome formation. Autophagosome formation has been shown to be initiated at multiple sites including the mitochondria‐associated membrane, endoplasmic reticulum, the ER‐golgi interface and the plasma membrane. PtdIns3P at the ER triggers the recruitment of the PtdIns3P‐binding protein ZFYVE1/DFCP1 (zinc finger, FYVE domain containing 1) protein and WD repeat domain, phosphoinositide interacting 2 protein (WIPI2) to from a PtdIns3P‐enriched ER‐associated structure termed the omegasome for its Ω‐like shape. Elongation and expansion of the phagophore membrane takes place by the Atg12‐Atg5‐Atg16 and Atg8 conjugation systems, which are ubiquitin like conjugation systems. The Atg12‐Atg5‐Atg16 complex catalyzes the lipidation of Atg8 by Atg4‐mediated cleavage followed by covalent linkage to phosphatidylethanolamine. Mammalian Atg8 homologs, namely, the microtubule‐associated protein 1 light chain 3 (MAP1LC3/LC3) and GABA (A) receptor‐associated protein (GABARAP) subfamily proteins are involved in membrane expansion and also interact with adaptor proteins that bind ubiquitinated proteins (as cargo) to sequester them within the autophagosomes. Adaptor proteins such as p62, NBR1, NDP52, VCP, and optineurin interact with ubiquitinated proteins through a ubiquitin‐binding domain (UBA) and with LC3 family of proteins via a LC3‐interacting region (LIR). After closure of the double membrane, autophagosome‐lysosome fusion occurs in a manner dependent upon lysosomal pH (requires acidified lysosomes) and regulated by Rab7, SNARE, and HOPS complex proteins. This is followed by intralysosomal degradation of autophagosome contents with recycling of basic building blocks back to the cytosol, completing the process of autophagy. Lysosomes harbor the LYNUS complex on their cytosolic face, which is comprised of mTOR complexed with the lysosomal proton pump, ragulator, Rag GTPases, GAP (GTPase activating) and GEF (GTP exchange factor) proteins, and Rheb GTPase. The LYNUS complex activates mTOR during the nutrient replete state which phosphorylates the TFEB family of transcription factors [reviewed in (161,187)]. Phosphorylated TFEB on lysosome membrane exists in equilibrium with a pool that is bound to cytosolic 14‐3‐3 proteins. Activated mTOR inactivates autophagy via phosphorylation of ULK1 and ATG13. Nutrient depletion induces the LYNUS complex to unravel, whereby mTOR becomes inactive relieving the constitutive phosphorylation of TFEB. Simultaneously, release of lysosome calcium via activation of the mucolipin channel activates calcineurin, which dephosphorylates TFEB to unmask its nuclear localization signal. Nuclear TFEB activates transcription of autophagy and lysosome genes, and induces autophagosome formation and lysosome biogenesis to upregulate flux through the macroautophagy pathway. These lysosomal degradative processes generate nutrients that provide energy and support various metabolic functions [reviewed in (97)], and destroy invading organisms in immune cells via the process of xenophagy and enable antigen presentation to mount an immune response [reviewed in (135)].
Figure 3. Figure 3. Regulation of autophagy‐lysosome machinery. Various cell‐intrinsic and ‐extrinsic inputs regulate the autophagy‐lysosome machinery. Organelle damage and impaired protein quality control with dysfunction of the ubiquitin‐proteasome system activates selective autophagy to remove the damaged organelles (mitochondria, ER, golgi, and lysososomes) and protein aggregates [reviewed in (68)]. Aging induces ROS upregulation and accumulation of DNA damage, which induce senescence in cardiac progenitors through telomere attrition and DNA damage [reviewed in (194)]. While SIRT1 expression is increased, experimental evidence indicates that further stimulation of SIRT1 function enhances autophagy via activation of FOXO1. Serum levels of GDF11, a TGFβ family member, decreases with age in the serum and increasing GDF11 levels beneficially affects age‐related cardiac phenotypes. While GDF11 stimulates autophagy in skeletal muscle, its effects on the heart have not been studied. miR‐216a increases in the aging heart and is known to downregulate Beclin‐1, a protein essential for autophagy induction. Nicotinamide phosphoribosyl transferase (Nampt), a key enzyme in the salvage pathway of NAD+ synthesis in cardiomyocytes is downregulated with aging, and restoring Nampt and NAD+ levels restores autophagic flux in the heart. Multiple transcriptional pathways intricately regulate the autophagy‐lysosome machinery [reviewed in (60)]. The FOXO family of transcription factors are autophagy activators and are activated by phosphorylation effected by AMPK, JNK, and MST1 kinases. However, Akt‐mediated phosphorylation of FOXO1 at a different site inactivates FOXO1 and holds it in the cytosol. Activation of the stress sensor ERN1/IRE1α signaling also holds FOXO activity in check. TP53 (tumor protein p53), a transcription factor that regulates cell cycle activates autophagy when localized to the nucleus by directly activating transcriprion of various ATG genes, but inhibits autophagy when in the cytosol. Lysosomal lipases are held in check by mxl‐3 in C. elegans and by its orthologue MAX in mice. Both are transcriptional repressors that bind to the CLEAR response element where TFEB binds to inhibit lipophagy and lipolysis for generation of energy [reviewed in (97)]. CREB, or cAMP response element‐binding protein is held in check by its interaction with FXR, farnesoid X receptor in the fed state. Fasting induces FXR degradation and binding of CREB to CRTC2, its coactivator to induce TFEB transcription. Fasting also causes LYNUS machinery to deactivate mTOR, which results in dephosphorylation and activation of TFEB. TFEB stimulates transcription of autophagy and lysosome pathway genes. Circadian rhythm plays a central role in regulation of the fasting feeding responses and controls autophagy‐lysosome machinery. Emerging evidence points to counterregulation of TFEB family of transcription factors by circadian rhythm regulators either indirectly via mTOR activity or directly via TFEB binding to Per genes [reviewed in (154)]. Numerous metabolic influences also regulate autophagy in the heart, as shown [and reviewed in (97)]. Please see section titled “Cardiac energetics, substrate metabolism and pathophysiology under fasting states” for a detailed discussion of cardiac metabolism under stress. “↑” indicates, whereas “↓” indicates inhibition.
Figure 4. Figure 4. Proposed mechanism for the effects of intermittent fasting on LYNUS‐TFEB‐mTORC1 axis in cardiac myocyte pathophysiology. Cardiac myocytes in pathological states are likely to be affected by the functioning of the lysosomal nutrient‐sensing complex [see reference (161) for a detailed review of signaling via the LYNUS complex, and text under the subsection titled “Lysosomes and nutrient sensing”]. Unopposed activation of mTORC1 as a result of nutrient oversupply, aging, lysosomal dysfunction, and diabetes mellitus, is expected to result in a sustained synthetic state. This would result in overload of the ER‐translational machinery precipitating ER stress and the unfolded protein response due to accumulation of misfolded entities. This pathological synthetic state is expected to provoke decreased recycling of organelles as well sarcomere renewal and repair. Furthermore, the overall metabolic reserve of the cell would be reduced due to a decrease in the ability of cells to activate catabolic pathways. Similarly, during starvation or with continuous mTOR antagonism, the translation machinery is expected to be suppressed while proteolysis is increased. This would drive breakdown of organelles and sarcomeres with suppression of organelle and sarcomere biogenesis. In addition, progressive depletion of cellular components by catabolic pathways is expected to result in a similar decrease in metabolic reserve. In contrast, intermittent fasting drives cyclic activation of both TFEB and mTORC1, which is expected to optimize protein turnover. This is accompanied by decreased ER stress, increased organelle turnover, where accelerated organelle/sarcomere breakdown is accompanied by increase biogenesis and optimal availability of cellular components and metabolic pathways to increase metabolic reserve. As compared to cardiac myocyte dysfunction seen with sustained activation of either mTORC1 or TFEB, acceleration of the cyclic activation of mTORC1 and TFEB is expected to results in cardiac myocyte rejuvenation. Similar mechanisms are proposed to drive the beneficial effects of exercise, intermittent rapamycin, circadian feeding, and sirtuin action.
Figure 5. Figure 5. Proposed LYNUS‐TFEB‐mTORC1 cues to entrain the circadian rhythm. Dietary factors have been noted to affect circadian physiology. In this proposed mechanism, diurnal variation in feeding behavior is accompanied by the effect of mTORC1 and TFEB on the Bmal1 and Clock heterodimer resulting in synchronization of the circadian clock with nutrient supply. High TFEB levels at waking (“Day” in diurnal species) enhance the Bmal1/CLOCK heterodimer, thus driving circadian genes. In contrast, peak mTORC1 activation at the onset of “night” phase, when feeding is complete affects Bmal1 ubiquitination and alters the circadian gene expression, coincident with the peak synthetic phase. As opposed to these two phases, the transition from synthesis to proteolysis (and vice versa) at “0” and “12,” is also accompanied by combined effects of TFEB and mTORC1 on the Bmal1/CLOCK proteins. By adhering to a fixed pattern, it may be thus possible to synchronize the metabolic and biologic clocks, and thus maximize cellular repair and renewal while optimizing metabolism. Please also see text under the section titled “Interplay of Lysosome Function with the Metabolic Circadian Axis.”
Figure 6. Figure 6. Lysosomal regulation of life and death. Lysosomes play multiple salutary roles as well as participate in orchestrating cell death programs. Necrosis and autosis (or autophagic cell death) are both characterized by lysosomal digestion of cellular contents. Please see reference (66) for a comprehensive review of the role of lysosomes and autophagy in cell death. In contrast to this destructive role, lysosomes play critical roles in both synthesis and nonproteasomal breakdown of proteins. By virtue of the lysosomal nutrient‐sensing mechanisms, they control cellular metabolic choices as well as circadian physiology. Lysosomes are also vital for cellular innate immunity against pathogens as well as dysfunctional organelles, sarcomeres and even, pathologically aggregated proteins. Through effects on TFEB and mTORC1, lysosomes can control gene expression as well as the translational machinery. Finally, processing of both intracellular and extracellular messages, via exosomes and endosomes, is regulated via lysosomal function.


Figure 1. Cardiac metabolism in health and disease. During normal physiology, the vast majority of myocardial ATP production is from oxidative phosphorylation. Of this, a majority is from β‐oxidation of fatty acids, with a smaller contribution from glucose oxidation. By contrast, glycolysis, ketone body oxidation, and amino acid metabolism contribute very small amounts to overall ATP generation. During pathological states, cardiac myocytes display alternate substrate preferences for energy generation (reviewed in (212,214); please see text under the section “Cardiac energetics, substrate metabolism and pathophysiology under fasting states” for details). (A) Metabolic changes during nutrient deprivation vary depending the duration (ST = short term and LT = long term). During short‐term fasting, fatty acid oxidation is unchanged while glucose metabolism is suppressed. In addition, marked increases in ketone body oxidation and amino acid metabolism contribute to myocardial energetics. During prolonged fasting, however, lipid oxidation is increased and serves as the main energetic source. (B) During ischemic syndromes (I = ischemia and IR = ischemia‐reperfusion), energetic choices are driven by oxygen availability. While ischemia is associated with reduction in fatty acid and glucose oxidation, glycolysis serves as a far more significant source of ATP. When reperfused, glycolysis rates decline and glucose oxidation is still suppressed, while fatty acid oxidation increases. In most ischemic syndromes, TCA (tricarboxylic acid cycle)‐independent amino acid metabolism is augmented. (C) Cardiac myocyte dysfunction in cardiomyopathy is often accompanied by abnormalities in myocardial metabolism and energetics. Prominent among these is a switch to a preference for glucose oxidation as opposed to lipid oxidation. (D) During aging, similar to cardiomyopathy, fatty acid oxidation is decreased with increases in glucose oxidation and glycolysis driving myocardial energetics. (E) Fatty acid utilization is increased in the diabetic myocardium (with type 2 diabetes due to insulin resistance) with reduction in glucose utilization. The diabetic heart increasingly utilizes ketone bodies as a nutrient source. “↑” indicates upregulation, whereas “↓” indicates downregulation.


Figure 2. Mechanisms for execution of autophagy as a lysosomal degradative pathway. Nutrient deprivation and organelle damage trigger macroautophagy in cardiac myocytes [reviewed in (67)]. Assembly of the phagophore (the initiation of the double membrane) is activated by class III phosphatidylinositol 3‐kinase (PtdIns3K) catalyzed generation of phosphatydil inositol 3‐phosphate (PtdIns3P) to recruit PtdIns3P‐binding proteins. A protein complex is formed comprised of Vps15 homolog phosphoinositide‐3‐kinase, regulatory subunit 4 (PIK3R4), the Vps34 homolog phosphatidylinositol 3‐kinase, catalytic subunit type 3 (PIK3C3), and the Vps30/Atg6 homolog Beclin 1. This interacts with AMBRA1 (not shown) and ATG14, and also contain the BECN1‐interacting protein UV radiation resistance associated (UVRAG, not shown) and SH3‐domain GRB2‐like endophilin B1 (SH3GLB1/Bif‐1, not shown) to promote autophagosome formation. Autophagosome formation has been shown to be initiated at multiple sites including the mitochondria‐associated membrane, endoplasmic reticulum, the ER‐golgi interface and the plasma membrane. PtdIns3P at the ER triggers the recruitment of the PtdIns3P‐binding protein ZFYVE1/DFCP1 (zinc finger, FYVE domain containing 1) protein and WD repeat domain, phosphoinositide interacting 2 protein (WIPI2) to from a PtdIns3P‐enriched ER‐associated structure termed the omegasome for its Ω‐like shape. Elongation and expansion of the phagophore membrane takes place by the Atg12‐Atg5‐Atg16 and Atg8 conjugation systems, which are ubiquitin like conjugation systems. The Atg12‐Atg5‐Atg16 complex catalyzes the lipidation of Atg8 by Atg4‐mediated cleavage followed by covalent linkage to phosphatidylethanolamine. Mammalian Atg8 homologs, namely, the microtubule‐associated protein 1 light chain 3 (MAP1LC3/LC3) and GABA (A) receptor‐associated protein (GABARAP) subfamily proteins are involved in membrane expansion and also interact with adaptor proteins that bind ubiquitinated proteins (as cargo) to sequester them within the autophagosomes. Adaptor proteins such as p62, NBR1, NDP52, VCP, and optineurin interact with ubiquitinated proteins through a ubiquitin‐binding domain (UBA) and with LC3 family of proteins via a LC3‐interacting region (LIR). After closure of the double membrane, autophagosome‐lysosome fusion occurs in a manner dependent upon lysosomal pH (requires acidified lysosomes) and regulated by Rab7, SNARE, and HOPS complex proteins. This is followed by intralysosomal degradation of autophagosome contents with recycling of basic building blocks back to the cytosol, completing the process of autophagy. Lysosomes harbor the LYNUS complex on their cytosolic face, which is comprised of mTOR complexed with the lysosomal proton pump, ragulator, Rag GTPases, GAP (GTPase activating) and GEF (GTP exchange factor) proteins, and Rheb GTPase. The LYNUS complex activates mTOR during the nutrient replete state which phosphorylates the TFEB family of transcription factors [reviewed in (161,187)]. Phosphorylated TFEB on lysosome membrane exists in equilibrium with a pool that is bound to cytosolic 14‐3‐3 proteins. Activated mTOR inactivates autophagy via phosphorylation of ULK1 and ATG13. Nutrient depletion induces the LYNUS complex to unravel, whereby mTOR becomes inactive relieving the constitutive phosphorylation of TFEB. Simultaneously, release of lysosome calcium via activation of the mucolipin channel activates calcineurin, which dephosphorylates TFEB to unmask its nuclear localization signal. Nuclear TFEB activates transcription of autophagy and lysosome genes, and induces autophagosome formation and lysosome biogenesis to upregulate flux through the macroautophagy pathway. These lysosomal degradative processes generate nutrients that provide energy and support various metabolic functions [reviewed in (97)], and destroy invading organisms in immune cells via the process of xenophagy and enable antigen presentation to mount an immune response [reviewed in (135)].


Figure 3. Regulation of autophagy‐lysosome machinery. Various cell‐intrinsic and ‐extrinsic inputs regulate the autophagy‐lysosome machinery. Organelle damage and impaired protein quality control with dysfunction of the ubiquitin‐proteasome system activates selective autophagy to remove the damaged organelles (mitochondria, ER, golgi, and lysososomes) and protein aggregates [reviewed in (68)]. Aging induces ROS upregulation and accumulation of DNA damage, which induce senescence in cardiac progenitors through telomere attrition and DNA damage [reviewed in (194)]. While SIRT1 expression is increased, experimental evidence indicates that further stimulation of SIRT1 function enhances autophagy via activation of FOXO1. Serum levels of GDF11, a TGFβ family member, decreases with age in the serum and increasing GDF11 levels beneficially affects age‐related cardiac phenotypes. While GDF11 stimulates autophagy in skeletal muscle, its effects on the heart have not been studied. miR‐216a increases in the aging heart and is known to downregulate Beclin‐1, a protein essential for autophagy induction. Nicotinamide phosphoribosyl transferase (Nampt), a key enzyme in the salvage pathway of NAD+ synthesis in cardiomyocytes is downregulated with aging, and restoring Nampt and NAD+ levels restores autophagic flux in the heart. Multiple transcriptional pathways intricately regulate the autophagy‐lysosome machinery [reviewed in (60)]. The FOXO family of transcription factors are autophagy activators and are activated by phosphorylation effected by AMPK, JNK, and MST1 kinases. However, Akt‐mediated phosphorylation of FOXO1 at a different site inactivates FOXO1 and holds it in the cytosol. Activation of the stress sensor ERN1/IRE1α signaling also holds FOXO activity in check. TP53 (tumor protein p53), a transcription factor that regulates cell cycle activates autophagy when localized to the nucleus by directly activating transcriprion of various ATG genes, but inhibits autophagy when in the cytosol. Lysosomal lipases are held in check by mxl‐3 in C. elegans and by its orthologue MAX in mice. Both are transcriptional repressors that bind to the CLEAR response element where TFEB binds to inhibit lipophagy and lipolysis for generation of energy [reviewed in (97)]. CREB, or cAMP response element‐binding protein is held in check by its interaction with FXR, farnesoid X receptor in the fed state. Fasting induces FXR degradation and binding of CREB to CRTC2, its coactivator to induce TFEB transcription. Fasting also causes LYNUS machinery to deactivate mTOR, which results in dephosphorylation and activation of TFEB. TFEB stimulates transcription of autophagy and lysosome pathway genes. Circadian rhythm plays a central role in regulation of the fasting feeding responses and controls autophagy‐lysosome machinery. Emerging evidence points to counterregulation of TFEB family of transcription factors by circadian rhythm regulators either indirectly via mTOR activity or directly via TFEB binding to Per genes [reviewed in (154)]. Numerous metabolic influences also regulate autophagy in the heart, as shown [and reviewed in (97)]. Please see section titled “Cardiac energetics, substrate metabolism and pathophysiology under fasting states” for a detailed discussion of cardiac metabolism under stress. “↑” indicates, whereas “↓” indicates inhibition.


Figure 4. Proposed mechanism for the effects of intermittent fasting on LYNUS‐TFEB‐mTORC1 axis in cardiac myocyte pathophysiology. Cardiac myocytes in pathological states are likely to be affected by the functioning of the lysosomal nutrient‐sensing complex [see reference (161) for a detailed review of signaling via the LYNUS complex, and text under the subsection titled “Lysosomes and nutrient sensing”]. Unopposed activation of mTORC1 as a result of nutrient oversupply, aging, lysosomal dysfunction, and diabetes mellitus, is expected to result in a sustained synthetic state. This would result in overload of the ER‐translational machinery precipitating ER stress and the unfolded protein response due to accumulation of misfolded entities. This pathological synthetic state is expected to provoke decreased recycling of organelles as well sarcomere renewal and repair. Furthermore, the overall metabolic reserve of the cell would be reduced due to a decrease in the ability of cells to activate catabolic pathways. Similarly, during starvation or with continuous mTOR antagonism, the translation machinery is expected to be suppressed while proteolysis is increased. This would drive breakdown of organelles and sarcomeres with suppression of organelle and sarcomere biogenesis. In addition, progressive depletion of cellular components by catabolic pathways is expected to result in a similar decrease in metabolic reserve. In contrast, intermittent fasting drives cyclic activation of both TFEB and mTORC1, which is expected to optimize protein turnover. This is accompanied by decreased ER stress, increased organelle turnover, where accelerated organelle/sarcomere breakdown is accompanied by increase biogenesis and optimal availability of cellular components and metabolic pathways to increase metabolic reserve. As compared to cardiac myocyte dysfunction seen with sustained activation of either mTORC1 or TFEB, acceleration of the cyclic activation of mTORC1 and TFEB is expected to results in cardiac myocyte rejuvenation. Similar mechanisms are proposed to drive the beneficial effects of exercise, intermittent rapamycin, circadian feeding, and sirtuin action.


Figure 5. Proposed LYNUS‐TFEB‐mTORC1 cues to entrain the circadian rhythm. Dietary factors have been noted to affect circadian physiology. In this proposed mechanism, diurnal variation in feeding behavior is accompanied by the effect of mTORC1 and TFEB on the Bmal1 and Clock heterodimer resulting in synchronization of the circadian clock with nutrient supply. High TFEB levels at waking (“Day” in diurnal species) enhance the Bmal1/CLOCK heterodimer, thus driving circadian genes. In contrast, peak mTORC1 activation at the onset of “night” phase, when feeding is complete affects Bmal1 ubiquitination and alters the circadian gene expression, coincident with the peak synthetic phase. As opposed to these two phases, the transition from synthesis to proteolysis (and vice versa) at “0” and “12,” is also accompanied by combined effects of TFEB and mTORC1 on the Bmal1/CLOCK proteins. By adhering to a fixed pattern, it may be thus possible to synchronize the metabolic and biologic clocks, and thus maximize cellular repair and renewal while optimizing metabolism. Please also see text under the section titled “Interplay of Lysosome Function with the Metabolic Circadian Axis.”


Figure 6. Lysosomal regulation of life and death. Lysosomes play multiple salutary roles as well as participate in orchestrating cell death programs. Necrosis and autosis (or autophagic cell death) are both characterized by lysosomal digestion of cellular contents. Please see reference (66) for a comprehensive review of the role of lysosomes and autophagy in cell death. In contrast to this destructive role, lysosomes play critical roles in both synthesis and nonproteasomal breakdown of proteins. By virtue of the lysosomal nutrient‐sensing mechanisms, they control cellular metabolic choices as well as circadian physiology. Lysosomes are also vital for cellular innate immunity against pathogens as well as dysfunctional organelles, sarcomeres and even, pathologically aggregated proteins. Through effects on TFEB and mTORC1, lysosomes can control gene expression as well as the translational machinery. Finally, processing of both intracellular and extracellular messages, via exosomes and endosomes, is regulated via lysosomal function.
References
 1.Abu‐Remaileh M, Wyant GA, Kim C, Laqtom NN, Abbasi M, Chan SH, Freinkman E, Sabatini DM. Lysosomal metabolomics reveals V‐ATPase‐ and mTOR‐dependent regulation of amino acid efflux from lysosomes. Science 358: 807‐813, 2017.
 2.Acosta‐Rodriguez VA, de Groot MHM, Rijo‐Ferreira F, Green CB, Takahashi JS. Mice under caloric restriction self‐impose a temporal restriction of food intake as revealed by an automated feeder system. Cell Metab 26: 267‐277 e262, 2017.
 3.Adeva‐Andany MM, Gonzalez‐Lucan M, Donapetry‐Garcia C, Fernandez‐Fernandez C, Ameneiros‐Rodriguez E. Glycogen metabolism in humans. BBA Clin 5: 85‐100, 2016.
 4.Ahmet I, Wan R, Mattson MP, Lakatta EG, Talan M. Cardioprotection by intermittent fasting in rats. Circulation 112: 3115‐3121, 2005.
 5.Amamou T, Normandin E, Pouliot J, Dionne IJ, Brochu M, Riesco E. Effect of a high‐protein energy‐restricted diet combined with resistance training on metabolic profile in older individuals with metabolic impairments. J Nutr Health Aging 21: 67‐74, 2017.
 6.An L, Hu XW, Zhang S, Hu X, Song Z, Naz A, Zi Z, Wu J, Li C, Zou Y, He L, Zhu H. UVRAG deficiency exacerbates doxorubicin‐induced cardiotoxicity. Sci Rep 7: 43251, 2017.
 7.Anding AL, Baehrecke EH. Cleaning house: Selective autophagy of organelles. Dev Cell 41: 10‐22, 2017.
 8.Anson RM, Guo Z, de CR, Iyun T, Rios M, Hagepanos A, Ingram DK, Lane MA, Mattson MP. Intermittent fasting dissociates beneficial effects of dietary restriction on glucose metabolism and neuronal resistance to injury from calorie intake. Proc Natl Acad Sci U S A 100: 6216‐6220, 2003.
 9.Appaix F, Kuznetsov AV, Usson Y, Kay L, Andrienko T, Olivares J, Kaambre T, Sikk P, Margreiter R, Saks V. Possible role of cytoskeleton in intracellular arrangement and regulation of mitochondria. Exp Physiol 88: 175‐190, 2003.
 10.Arad M, Maron BJ, Gorham JM, Johnson WH, Jr., Saul JP, Perez‐Atayde AR, Spirito P, Wright GB, Kanter RJ, Seidman CE, Seidman JG. Glycogen storage diseases presenting as hypertrophic cardiomyopathy. N Engl J Med 352: 362‐372, 2005.
 11.Arad M, Seidman CE, Seidman JG. AMP‐activated protein kinase in the heart: Role during health and disease. Circ Res 100: 474‐488, 2007.
 12.Arriola Apelo SI, Neuman JC, Baar EL, Syed FA, Cummings NE, Brar HK, Pumper CP, Kimple ME, Lamming DW. Alternative rapamycin treatment regimens mitigate the impact of rapamycin on glucose homeostasis and the immune system. Aging Cell 15: 28‐38, 2016.
 13.Arriola Apelo SI, Pumper CP, Baar EL, Cummings NE, Lamming DW. Intermittent administration of rapamycin extends the life span of female C57BL/6J mice. J Gerontol A Biol Sci Med Sci 71: 876‐881, 2016.
 14.Asher G, Sassone‐Corsi P. Time for food: The intimate interplay between nutrition, metabolism, and the circadian clock. Cell 161: 84‐92, 2015.
 15.Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC, Koves T, Gardell SJ, Kruger M, Hoppel CL, Lewandowski ED, Crawford PA, Muoio DM, Kelly DP. The failing heart relies on ketone bodies as a fuel. Circulation 133: 698‐705, 2016.
 16.Balaban RS, Kantor HL, Katz LA, Briggs RW. Relation between work and phosphate metabolite in the in vivo paced mammalian heart. Science 232: 1121‐1123, 1986.
 17.Bales CW, Kraus WE. Caloric restriction: Implications for human cardiometabolic health. J Cardiopulm Rehabil Prev 33: 201‐208, 2013.
 18.Bao J, Zheng L, Zhang Q, Li X, Zhang X, Li Z, Bai X, Zhang Z, Huo W, Zhao X, Shang S, Wang Q, Zhang C, Ji J. Deacetylation of TFEB promotes fibrillar Abeta degradation by upregulating lysosomal biogenesis in microglia. Protein Cell 7: 417‐433, 2016.
 19.Barnosky AR, Hoddy KK, Unterman TG, Varady KA. Intermittent fasting vs daily calorie restriction for type 2 diabetes prevention: A review of human findings. Transl Res 164: 302‐311, 2014.
 20.Batacan RB, Jr., Duncan MJ, Dalbo VJ, Connolly KJ, Fenning AS. Light‐intensity and high‐intensity interval training improve cardiometabolic health in rats. Appl Physiol Nutr Metab 41: 945‐952, 2016.
 21.Bedi KC, Jr., Snyder NW, Brandimarto J, Aziz M, Mesaros C, Worth AJ, Wang LL, Javaheri A, Blair IA, Margulies KB, Rame JE. Evidence for intramyocardial disruption of lipid metabolism and increased myocardial ketone utilization in advanced human heart failure. Circulation 133: 706‐716, 2016.
 22.Beli E, Yan Y, Moldovan L, Vieira CP, Gao R, Duan Y, Prasad R, Bhatwadekar A, White FA, Townsend S, Chan L, Ryan CN, Morton D, Moldovan EG, Chu FI, Oudit GY, Derendorf H, Adorini L, Wang XX, Evans‐Molina C, Mirmira RG, Boulton ME, Yoder MC, Li Q, Levi M, Busik JV, Grant MB. Restructuring of the gut microbiome by intermittent fasting prevents retinopathy and prolongs survival in db/db mice. Diabetes 2018. pii: db180158. doi: 10.2337/db18‐0158.
 23.Billia F, Hauck L, Konecny F, Rao V, Shen J, Mak TW. PTEN‐inducible kinase 1 (PINK1)/Park6 is indispensable for normal heart function. Proc Natl Acad Sci U S A 108: 9572‐9577, 2011.
 24.Brandhorst S, Choi IY, Wei M, Cheng CW, Sedrakyan S, Navarrete G, Dubeau L, Yap LP, Park R, Vinciguerra M, Di Biase S, Mirzaei H, Mirisola MG, Childress P, Ji L, Groshen S, Penna F, Odetti P, Perin L, Conti PS, Ikeno Y, Kennedy BK, Cohen P, Morgan TE, Dorff TB, Longo VD. A periodic diet that mimics fasting promotes multi‐system regeneration, enhanced cognitive performance, and healthspan. Cell Metab 22: 86‐99, 2015.
 25.Bravo‐San Pedro JM, Kroemer G, Galluzzi L. Autophagy and mitophagy in cardiovascular disease. Circ Res 120: 1812‐1824, 2017.
 26.Brewer RA, Collins HE, Berry RD, Brahma MK, Tirado BA, Peliciari‐Garcia RA, Stanley HL, Wende AR, Taegtmeyer H, Rajasekaran NS, Darley‐Usmar V, Zhang J, Frank SJ, Chatham JC, Young ME. Temporal partitioning of adaptive responses of the murine heart to fasting. Life Sci 197: 30‐39, 2018.
 27.Brisson L, Banski P, Sboarina M, Dethier C, Danhier P, Fontenille MJ, Van Hee VF, Vazeille T, Tardy M, Falces J, Bouzin C, Porporato PE, Frederick R, Michiels C, Copetti T, Sonveaux P. Lactate dehydrogenase B controls lysosome activity and autophagy in cancer. Cancer Cell 30: 418‐431, 2016.
 28.Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, Hu LS, Cheng HL, Jedrychowski MP, Gygi SP, Sinclair DA, Alt FW, Greenberg ME. Stress‐dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303: 2011‐2015, 2004.
 29.Buss SJ, Muenz S, Riffel JH, Malekar P, Hagenmueller M, Weiss CS, Bea F, Bekeredjian R, Schinke‐Braun M, Izumo S, Katus HA, Hardt SE. Beneficial effects of mammalian target of rapamycin inhibition on left ventricular remodeling after myocardial infarction. J Am Coll Cardiol 54: 2435‐2446, 2009.
 30.Camandola S, Mattson MP. Brain metabolism in health, aging, and neurodegeneration. EMBO J 36: 1474‐1492, 2017.
 31.Cao R, Robinson B, Xu H, Gkogkas C, Khoutorsky A, Alain T, Yanagiya A, Nevarko T, Liu AC, Amir S, Sonenberg N. Translational control of entrainment and synchrony of the suprachiasmatic circadian clock by mTOR/4E‐BP1 signaling. Neuron 79: 712‐724, 2013.
 32.Carlson AJ, Hoelzel F. Apparent prolongation of the life span of rats by intermittent fasting. J Nutri 31: 363‐375, 1946.
 33.Castellano BM, Thelen AM, Moldavski O, Feltes M, van der Welle RE, Mydock‐McGrane L, Jiang X, van Eijkeren RJ, Davis OB, Louie SM, Perera RM, Covey DF, Nomura DK, Ory DS, Zoncu R. Lysosomal cholesterol activates mTORC1 via an SLC38A9‐Niemann‐Pick C1 signaling complex. Science 355: 1306‐1311, 2017.
 34.Chaix A, Zarrinpar A, Panda S. The circadian coordination of cell biology. J Cell Biol 215: 15‐25, 2016.
 35.Chambers KT, Leone TC, Sambandam N, Kovacs A, Wagg CS, Lopaschuk GD, Finck BN, Kelly DP. Chronic inhibition of pyruvate dehydrogenase in heart triggers an adaptive metabolic response. J Biol Chem 286: 11155‐11162, 2011.
 36.Chandramouli C, Varma U, Stevens EM, Xiao RP, Stapleton DI, Mellor KM, Delbridge LM. Myocardial glycogen dynamics: New perspectives on disease mechanisms. Clin Exp Pharmacol Physiol 42: 415‐425, 2015.
 37.Chantranupong L, Wolfson RL, Sabatini DM. Nutrient‐sensing mechanisms across evolution. Cell 161: 67‐83, 2015.
 38.Cheng CW, Villani V, Buono R, Wei M, Kumar S, Yilmaz OH, Cohen P, Sneddon JB, Perin L, Longo VD. Fasting‐mimicking diet promotes Ngn3‐driven beta‐cell regeneration to reverse diabetes. Cell 168: 775‐788 e712, 2017.
 39.Chiao YA, Kolwicz SC, Basisty N, Gagnidze A, Zhang J, Gu H, Djukovic D, Beyer RP, Raftery D, MacCoss M, Tian R, Rabinovitch PS. Rapamycin transiently induces mitochondrial remodeling to reprogram energy metabolism in old hearts. Aging (Albany NY) 8: 314‐327, 2016.
 40.Chin RM, Fu X, Pai MY, Vergnes L, Hwang H, Deng G, Diep S, Lomenick B, Meli VS, Monsalve GC, Hu E, Whelan SA, Wang JX, Jung G, Solis GM, Fazlollahi F, Kaweeteerawat C, Quach A, Nili M, Krall AS, Godwin HA, Chang HR, Faull KF, Guo F, Jiang M, Trauger SA, Saghatelian A, Braas D, Christofk HR, Clarke CF, Teitell MA, Petrascheck M, Reue K, Jung ME, Frand AR, Huang J. The metabolite alpha‐ketoglutarate extends lifespan by inhibiting ATP synthase and TOR. Nature 510: 397‐401, 2014.
 41.Colman RJ, Anderson RM, Johnson SC, Kastman EK, Kosmatka KJ, Beasley TM, Allison DB, Cruzen C, Simmons HA, Kemnitz JW, Weindruch R. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325: 201‐204, 2009.
 42.Cotter DG, Schugar RC, Wentz AE, d'Avignon DA, Crawford PA. Successful adaptation to ketosis by mice with tissue‐specific deficiency of ketone body oxidation. Am J Physiol Endocrinol Metab 304: E363‐E374, 2013.
 43.Dai DF, Karunadharma PP, Chiao YA, Basisty N, Crispin D, Hsieh EJ, Chen T, Gu H, Djukovic D, Raftery D, Beyer RP, MacCoss MJ, Rabinovitch PS. Altered proteome turnover and remodeling by short‐term caloric restriction or rapamycin rejuvenate the aging heart. Aging Cell 13: 529‐539, 2014.
 44.Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury‐Olela F, Schibler U. Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev 14: 2950‐2961, 2000.
 45.Dassanayaka S, Jones SP. O‐GlcNAc and the cardiovascular system. Pharmacol Ther 142: 62‐71, 2014.
 46.Decker RS, Poole AR, Crie JS, Dingle JT, Wildenthal K. Lysosomal alterations in hypoxic and reoxygenated hearts. II. Immunohistochemical and biochemical changes in cathepsin D. Am J Pathol 98: 445‐456, 1980.
 47.Decker RS, Poole AR, Wildenthal K. Distribution of lysosomal cathepsin D in normal, ischemic, and starved rabbit cardiac myocytes. Circ Res 46: 485‐494, 1980.
 48.Decker RS, Wildenthal K. Lysosomal alterations in hypoxic and reoxygenated hearts. I. Ultrastructural and cytochemical changes. Am J Pathol 98: 425‐444, 1980.
 49.Delbridge LMD, Mellor KM, Taylor DJ, Gottlieb RA. Myocardial stress and autophagy: Mechanisms and potential therapies. Nat Rev Cardiol 14: 412‐425, 2017.
 50.Di Malta C, Siciliano D, Calcagni A, Monfregola J, Punzi S, Pastore N, Eastes AN, Davis O, De Cegli R, Zampelli A, Di Giovannantonio LG, Nusco E, Platt N, Guida A, Ogmundsdottir MH, Lanfrancone L, Perera RM, Zoncu R, Pelicci PG, Settembre C, Ballabio A. Transcriptional activation of RagD GTPase controls mTORC1 and promotes cancer growth. Science 356: 1188‐1192, 2017.
 51.Diwan A. Regulation of the transcription factor EB‐PGC1alpha axis by beclin‐1 controls mitochondrial quality and cardiomyocyte death under stress. Autophagy 35: 956‐976, 2015.
 52.Efeyan A, Zoncu R, Chang S, Gumper I, Snitkin H, Wolfson RL, Kirak O, Sabatini DD, Sabatini DM. Regulation of mTORC1 by the Rag GTPases is necessary for neonatal autophagy and survival. Nature 493: 679‐683, 2013.
 53.Egan D, Kim J, Shaw RJ, Guan KL. The autophagy initiating kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR. Autophagy 7: 643‐644, 2011.
 54.Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, Asara JM, Fitzpatrick J, Dillin A, Viollet B, Kundu M, Hansen M, Shaw RJ. Phosphorylation of ULK1 (hATG1) by AMP‐activated protein kinase connects energy sensing to mitophagy. Science 331: 456‐461, 2011.
 55.Eisenberg T, Abdellatif M, Schroeder S, Primessnig U, Stekovic S, Pendl T, Harger A, Schipke J, Zimmermann A, Schmidt A, Tong M, Ruckenstuhl C, Dammbrueck C, Gross AS, Herbst V, Magnes C, Trausinger G, Narath S, Meinitzer A, Hu Z, Kirsch A, Eller K, Carmona‐Gutierrez D, Buttner S, Pietrocola F, Knittelfelder O, Schrepfer E, Rockenfeller P, Simonini C, Rahn A, Horsch M, Moreth K, Beckers J, Fuchs H, Gailus‐Durner V, Neff F, Janik D, Rathkolb B, Rozman J, de Angelis MH, Moustafa T, Haemmerle G, Mayr M, Willeit P, von Frieling‐Salewsky M, Pieske B, Scorrano L, Pieber T, Pechlaner R, Willeit J, Sigrist SJ, Linke WA, Muhlfeld C, Sadoshima J, Dengjel J, Kiechl S, Kroemer G, Sedej S, Madeo F. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med 22: 1428‐1438, 2016.
 56.Eisenberg T, Schroeder S, Andryushkova A, Pendl T, Kuttner V, Bhukel A, Marino G, Pietrocola F, Harger A, Zimmermann A, Moustafa T, Sprenger A, Jany E, Buttner S, Carmona‐Gutierrez D, Ruckenstuhl C, Ring J, Reichelt W, Schimmel K, Leeb T, Moser C, Schatz S, Kamolz LP, Magnes C, Sinner F, Sedej S, Frohlich KU, Juhasz G, Pieber TR, Dengjel J, Sigrist SJ, Kroemer G, Madeo F. Nucleocytosolic depletion of the energy metabolite acetyl‐coenzyme a stimulates autophagy and prolongs lifespan. Cell Metab 19: 431‐444, 2014.
 57.Elkhwanky MS, Hakkola J. Extranuclear sirtuins and metabolic stress. Antioxid Redox Signal 28(8): 662‐676, 2017.
 58.Ellenbroek JH, van Dijck L, Tons HA, Rabelink TJ, Carlotti F, Ballieux BE, de Koning EJ. Long‐term ketogenic diet causes glucose intolerance and reduced beta‐ and alpha‐cell mass but no weight loss in mice. Am J Physiol Endocrinol Metab 306: E552‐E558, 2014.
 59.Feng Y, Yao Z, Klionsky DJ. How to control self‐digestion: Transcriptional, post‐transcriptional, and post‐translational regulation of autophagy. Trends Cell Biol 25: 354‐363, 2015.
 60.Ferreira CR, Gahl WA. Lysosomal storage diseases. Transl Sci Rare Dis 2: 1‐71, 2017.
 61.Fillmore N, Lopaschuk GD. Targeting mitochondrial oxidative metabolism as an approach to treat heart failure. Biochim Biophys Acta 1833: 857‐865, 2013.
 62.Flynn JM, O'Leary MN, Zambataro CA, Academia EC, Presley MP, Garrett BJ, Zykovich A, Mooney SD, Strong R, Rosen CJ, Kapahi P, Nelson MD, Kennedy BK, Melov S. Late‐life rapamycin treatment reverses age‐related heart dysfunction. Aging Cell 12: 851‐862, 2013.
 63.Fontana L, Meyer TE, Klein S, Holloszy JO. Long‐term calorie restriction is highly effective in reducing the risk for atherosclerosis in humans. Proc Natl Acad Sci U S A 101: 6659‐6663, 2004.
 64.Fontana L, Vinciguerra M, Longo VD. Growth factors, nutrient signaling, and cardiovascular aging. Circ Res 110: 1139‐1150, 2012.
 65.Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Annicchiarico‐Petruzzelli M, Antonov AV, Arama E, Baehrecke EH, Barlev NA, Bazan NG, Bernassola F, Bertrand MJM, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Boya P, Brenner C, Campanella M, Candi E, Carmona‐Gutierrez D, Cecconi F, Chan FK, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Cohen GM, Conrad M, Cubillos‐Ruiz JR, Czabotar PE, D'Angiolella V, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, DeBerardinis RJ, Deshmukh M, Di Daniele N, Di Virgilio F, Dixit VM, Dixon SJ, Duckett CS, Dynlacht BD, El‐Deiry WS, Elrod JW, Fimia GM, Fulda S, Garcia‐Saez AJ, Garg AD, Garrido C, Gavathiotis E, Golstein P, Gottlieb E, Green DR, Greene LA, Gronemeyer H, Gross A, Hajnoczky G, Hardwick JM, Harris IS, Hengartner MO, Hetz C, Ichijo H, Jaattela M, Joseph B, Jost PJ, Juin PP, Kaiser WJ, Karin M, Kaufmann T, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Knight RA, Kumar S, Lee SW, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, Lopez‐Otin C, Lowe SW, Luedde T, Lugli E, MacFarlane M, Madeo F, Malewicz M, Malorni W, Manic G, Marine JC, Martin SJ, Martinou JC, Medema JP, Mehlen P, Meier P, Melino S, Miao EA, Molkentin JD, Moll UM, Munoz‐Pinedo C, Nagata S, Nunez G, Oberst A, Oren M, Overholtzer M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pereira DM, Pervaiz S, Peter ME, Piacentini M, Pinton P, Prehn JHM, Puthalakath H, Rabinovich GA, Rehm M, Rizzuto R, Rodrigues CMP, Rubinsztein DC, Rudel T, Ryan KM, Sayan E, Scorrano L, Shao F, Shi Y, Silke J, Simon HU, Sistigu A, Stockwell BR, Strasser A, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Thorburn A, Tsujimoto Y, Turk B, Vanden Berghe T, Vandenabeele P, Vander Heiden MG, Villunger A, Virgin HW, Vousden KH, Vucic D, Wagner EF, Walczak H, Wallach D, Wang Y, Wells JA, Wood W, Yuan J, Zakeri Z, Zhivotovsky B, Zitvogel L, Melino G, Kroemer G. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 25: 486‐541, 2018.
 66.Gatica D, Chiong M, Lavandero S, Klionsky DJ. Molecular mechanisms of autophagy in the cardiovascular system. Circ Res 116: 456‐467, 2015.
 67.Gatica D, Lahiri V, Klionsky DJ. Cargo recognition and degradation by selective autophagy. Nat Cell Biol 20: 233‐242, 2018.
 68.Giebultowicz J, Kapahi P. Circadian clocks and metabolism: The nutrient‐sensing AKT and TOR pathways make the link. Curr Biol 20: R608‐R609, 2010.
 69.Glatz JF, Nabben M, Heather LC, Bonen A, Luiken JJ. Regulation of the subcellular trafficking of CD36, a major determinant of cardiac fatty acid utilization. Biochim Biophys Acta 1861: 1461‐1471, 2016.
 70.Godar RJ, Ma X, Liu H, Murphy JT, Weinheimer CJ, Kovacs A, Crosby SD, Saftig P, Diwan A. Repetitive stimulation of autophagy‐lysosome machinery by intermittent fasting preconditions the myocardium to ischemia‐reperfusion injury. Autophagy 11: 1537‐1560, 2015.
 71.Goldhamer A, Lisle D, Parpia B, Anderson SV, Campbell TC. Medically supervised water‐only fasting in the treatment of hypertension. J Manipulative Physiol Ther 24: 335‐339, 2001.
 72.Goldhamer AC, Lisle DJ, Sultana P, Anderson SV, Parpia B, Hughes B, Campbell TC. Medically supervised water‐only fasting in the treatment of borderline hypertension. J Altern Complement Med 8: 643‐650, 2002.
 73.Gong G, Song M, Csordas G, Kelly DP, Matkovich SJ, Dorn GW, II. Parkin‐mediated mitophagy directs perinatal cardiac metabolic maturation in mice. Science 350: aad2459, 2015.
 74.Goodrick CL, Ingram DK, Reynolds MA, Freeman JR, Cider N. Effects of intermittent feeding upon body weight and lifespan in inbred mice: Interaction of genotype and age. Mech Ageing Dev 55: 69‐87, 1990.
 75.Gow ML, Garnett SP, Baur LA, Lister NB. The effectiveness of different diet strategies to reduce type 2 diabetes risk in youth. Nutrients 8(8): pii: E486, 2016.
 76.Guo B, Liang Q, Li L, Hu Z, Wu F, Zhang P, Ma Y, Zhao B, Kovacs AL, Zhang Z, Feng D, Chen S, Zhang H. O‐GlcNAc‐modification of SNAP‐29 regulates autophagosome maturation. Nat Cell Biol 16: 1215‐1226, 2014.
 77.Guzun R, Kaambre T, Bagur R, Grichine A, Usson Y, Varikmaa M, Anmann T, Tepp K, Timohhina N, Shevchuk I, Chekulayev V, Boucher F, Dos Santos P, Schlattner U, Wallimann T, Kuznetsov AV, Dzeja P, Aliev M, Saks V. Modular organization of cardiac energy metabolism: Energy conversion, transfer and feedback regulation. Acta Physiol (Oxf) 213: 84‐106, 2015.
 78.Halestrap AP. Monocarboxylic acid transport. Compr Physiol 3: 1611‐1643, 2013.
 79.Hammerling BC, Najor RH, Cortez MQ, Shires SE, Leon LJ, Gonzalez ER, Boassa D, Phan S, Thor A, Jimenez RE, Li H, Kitsis RN, Dorn Ii GW, Sadoshima J, Ellisman MH, Gustafsson AB. A Rab5 endosomal pathway mediates Parkin‐dependent mitochondrial clearance. Nat Commun 8: 14050, 2017.
 80.Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki‐Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441: 885‐889, 2006.
 81.Hariharan N, Maejima Y, Nakae J, Paik J, Depinho RA, Sadoshima J. Deacetylation of FoxO by Sirt1 plays an essential role in mediating starvation‐induced autophagy in cardiac myocytes. Circ Res 107: 1470‐1482, 2010.
 82.Harvie MN, Pegington M, Mattson MP, Frystyk J, Dillon B, Evans G, Cuzick J, Jebb SA, Martin B, Cutler RG, Son TG, Maudsley S, Carlson OD, Egan JM, Flyvbjerg A, Howell A. The effects of intermittent or continuous energy restriction on weight loss and metabolic disease risk markers: A randomized trial in young overweight women. Int J Obes (Lond) 35: 714‐727, 2011.
 83.Hatori M, Vollmers C, Zarrinpar A, DiTacchio L, Bushong EA, Gill S, Leblanc M, Chaix A, Joens M, Fitzpatrick JA, Ellisman MH, Panda S. Time‐restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high‐fat diet. Cell Metab 15: 848‐860, 2012.
 84.He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43: 67‐93, 2009.
 85.Heilbronn LK, Smith SR, Martin CK, Anton SD, Ravussin E. Alternate‐day fasting in nonobese subjects: Effects on body weight, body composition, and energy metabolism. Am J Clin Nutr 81: 69‐73, 2005.
 86.Honjoh S, Yamamoto T, Uno M, Nishida E. Signalling through RHEB‐1 mediates intermittent fasting‐induced longevity in C. elegans. Nature 457: 726‐730, 2009.
 87.Horne BD, May HT, Anderson JL, Kfoury AG, Bailey BM, McClure BS, Renlund DG, Lappe DL, Carlquist JF, Fisher PW, Pearson RR, Bair TL, Adams TD, Muhlestein JB, Intermountain Heart Collaborative S. Usefulness of routine periodic fasting to lower risk of coronary artery disease in patients undergoing coronary angiography. Am J Cardiol 102: 814‐819, 2008.
 88.Horne BD, Muhlestein JB, May HT, Carlquist JF, Lappe DL, Bair TL, Anderson JL, Intermountain Heart Collaborative Study G. Relation of routine, periodic fasting to risk of diabetes mellitus, and coronary artery disease in patients undergoing coronary angiography. Am J Cardiol 109: 1558‐1562, 2012.
 89.Ikeda Y, Shirakabe A, Maejima Y, Zhai P, Sciarretta S, Toli J, Nomura M, Mihara K, Egashira K, Ohishi M, Abdellatif M, Sadoshima J. Endogenous Drp1 mediates mitochondrial autophagy and protects the heart against energy stress. Circ Res 116: 264‐278, 2015.
 90.Berg JM, Tymoczko JL, Stryer L. Biochemistry, 5th edition. W H Freeman, 2002. ISBN‐10: 0‐7167‐3051‐0.
 91.Jia G, Hill MA, Sowers JR. Diabetic cardiomyopathy: An update of mechanisms contributing to this clinical entity. Circ Res 122: 624‐638, 2018.
 92.Johnson JB, Summer W, Cutler RG, Martin B, Hyun DH, Dixit VD, Pearson M, Nassar M, Telljohann R, Maudsley S, Carlson O, John S, Laub DR, Mattson MP. Alternate day calorie restriction improves clinical findings and reduces markers of oxidative stress and inflammation in overweight adults with moderate asthma. Free Radic Biol Med 42: 665‐674, 2007.
 93.Kamphoven JH, Stubenitsky R, Reuser AJ, Van Der Ploeg AT, Verdouw PD, Duncker DJ. Cardiac remodeling and contractile function in acid alpha‐glucosidase knockout mice. Physiol Genomics 5: 171‐179, 2001.
 94.Kanamori H, Takemura G, Maruyama R, Goto K, Tsujimoto A, Ogino A, Li L, Kawamura I, Takeyama T, Kawaguchi T, Nagashima K, Fujiwara T, Fujiwara H, Seishima M, Minatoguchi S. Functional significance and morphological characterization of starvation‐induced autophagy in the adult heart. Am J Pathol 174: 1705‐1714, 2009.
 95.Katare RG, Kakinuma Y, Arikawa M, Yamasaki F, Sato T. Chronic intermittent fasting improves the survival following large myocardial ischemia by activation of BDNF/VEGF/PI3K signaling pathway. J Mol Cell Cardiol 46: 405‐412, 2009.
 96.Kaur J, Debnath J. Autophagy at the crossroads of catabolism and anabolism. Nat Rev Mol Cell Biol 16: 461‐472, 2015.
 97.Kaushik S, Cuervo AM. AMPK‐dependent phosphorylation of lipid droplet protein PLIN2 triggers its degradation by CMA. Autophagy 12: 432‐438, 2016.
 98.Kemper JK, Xiao Z, Ponugoti B, Miao J, Fang S, Kanamaluru D, Tsang S, Wu SY, Chiang CM, Veenstra TD. FXR acetylation is normally dynamically regulated by p300 and SIRT1 but constitutively elevated in metabolic disease states. Cell Metab 10: 392‐404, 2009.
 99.Kessler CS, Stange R, Schlenkermann M, Jeitler M, Michalsen A, Selle A, Raucci F, Steckhan N. A nonrandomized controlled clinical pilot trial on 8 wk of intermittent fasting (24 h/wk). Nutrition 46: 143‐152 e142, 2018.
 100.Kim TT, Dyck JR. The role of CD36 in the regulation of myocardial lipid metabolism. Biochim Biophys Acta 1861: 1450‐1460, 2016.
 101.Kim YC, Park HW, Sciarretta S, Mo JS, Jewell JL, Russell RC, Wu X, Sadoshima J, Guan KL. Rag GTPases are cardioprotective by regulating lysosomal function. Nat Commun 5: 4241, 2014.
 102.Kingma SD, Bodamer OA, Wijburg FA. Epidemiology and diagnosis of lysosomal storage disorders; challenges of screening. Best Pract Res Clin Endocrinol Metab 29: 145‐157, 2015.
 103.Klempel MC, Kroeger CM, Bhutani S, Trepanowski JF, Varady KA. Intermittent fasting combined with calorie restriction is effective for weight loss and cardio‐protection in obese women. Nutr J 11: 98, 2012.
 104.Kolwicz SC, Jr., Purohit S, Tian R. Cardiac metabolism and its interactions with contraction, growth, and survival of cardiomyocytes. Circ Res 113: 603‐616, 2013.
 105.Kubli DA, Cortez MQ, Moyzis AG, Najor RH, Lee Y, Gustafsson AB. PINK1 is dispensable for mitochondrial recruitment of Parkin and activation of mitophagy in cardiac myocytes. PloS One 10: e0130707, 2015.
 106.Kubli DA, Zhang X, Lee Y, Hanna RA, Quinsay MN, Nguyen CK, Jimenez R, Petrosyan S, Murphy AN, Gustafsson AB. Parkin protein deficiency exacerbates cardiac injury and reduces survival following myocardial infarction. J Biol Chem 288: 915‐926, 2013.
 107.Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, Tokuhisa T, Mizushima N. The role of autophagy during the early neonatal starvation period. Nature 432: 1032‐1036, 2004.
 108.Kumar S, Kaur G. Intermittent fasting dietary restriction regimen negatively influences reproduction in young rats: A study of hypothalamo‐hypophysial‐gonadal axis. PloS One 8: e52416, 2013.
 109.Lamia KA, Sachdeva UM, DiTacchio L, Williams EC, Alvarez JG, Egan DF, Vasquez DS, Juguilon H, Panda S, Shaw RJ, Thompson CB, Evans RM. AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science 326: 437‐440, 2009.
 110.Lange LG, Sobel BE. Mitochondrial dysfunction induced by fatty acid ethyl esters, myocardial metabolites of ethanol. J Clin Invest 72: 724‐731, 1983.
 111.Lee JM, Wagner M, Xiao R, Kim KH, Feng D, Lazar MA, Moore DD. Nutrient‐sensing nuclear receptors coordinate autophagy. Nature 516: 112‐115, 2014.
 112.Li DL, Wang ZV, Ding G, Tan W, Luo X, Criollo A, Xie M, Jiang N, May H, Kyrychenko V, Schneider JW, Gillette TG, Hill JA. Doxorubicin blocks cardiomyocyte autophagic flux by inhibiting lysosome acidification. Circulation 133: 1668‐1687, 2016.
 113.Li G, Xie C, Lu S, Nichols RG, Tian Y, Li L, Patel D, Ma Y, Brocker CN, Yan T, Krausz KW, Xiang R, Gavrilova O, Patterson AD, Gonzalez FJ. Intermittent fasting promotes white adipose browning and decreases obesity by shaping the gut microbiota. Cell Metab 26: 801, 2017.
 114.Lim CY, Zoncu R. The lysosome as a command‐and‐control center for cellular metabolism. J Cell Biol 214: 653‐664, 2016.
 115.Lim JA, Li L, Shirihai OS, Trudeau KM, Puertollano R, Raben N. Modulation of mTOR signaling as a strategy for the treatment of Pompe disease. EMBO Mol Med 9: 353‐370, 2017.
 116.Lipton JO, Yuan ED, Boyle LM, Ebrahimi‐Fakhari D, Kwiatkowski E, Nathan A, Guttler T, Davis F, Asara JM, Sahin M. The circadian protein BMAL1 regulates translation in response to S6K1‐mediated phosphorylation. Cell 161: 1138‐1151, 2015.
 117.Liu H, Javaheri A, Godar RJ, Murphy J, Ma X, Rohatgi N, Mahadevan J, Hyrc K, Saftig P, Marshall C, McDaniel ML, Remedi MS, Razani B, Urano F, Diwan A. Intermittent fasting preserves beta‐cell mass in obesity‐induced diabetes via the autophagy‐lysosome pathway. Autophagy13(11): 1952‐1968, 2017.
 118.Luo W, Ma S, Yang Y, Wang C, Zhang D, Zhang Q, Liu Y, Liu Z. TFEB regulates PER3 expression via glucose‐dependent effects on CLOCK/BMAL1. Int J Biochem Cell Biol 78: 31‐42, 2016.
 119.Ma S, Gladyshev VN. Molecular signatures of longevity: Insights from cross‐species comparative studies. Semin Cell Dev Biol 70: 190‐203, 2017.
 120.Ma X, Liu H, Foyil SR, Godar RJ, Weinheimer CJ, Diwan A. Autophagy is impaired in cardiac ischemia‐reperfusion injury. Autophagy 8(9): 1394‐1396, 2012.
 121.Ma X, Liu H, Foyil SR, Godar RJ, Weinheimer CJ, Hill JA, Diwan A. Impaired autophagosome clearance contributes to cardiomyocyte death in ischemia/reperfusion injury. Circulation 125: 3170‐3181, 2012.
 122.Ma X, Liu H, Murphy JT, Foyil SR, Godar RJ, Abuirqeba H, Weinheimer CJ, Barger PM, Diwan A. Regulation of TFEB‐PGC1alpha axis by BECLIN‐1 controls mitochondrial quality and cardiomyocyte death under stress. Mol Cell Biol 35(6): 956‐976, 2015.
 123.Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, Ko CH, Ivanova G, Omura C, Mo S, Vitaterna MH, Lopez JP, Philipson LH, Bradfield CA, Crosby SD, JeBailey L, Wang X, Takahashi JS, Bass J. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466: 627‐631, 2010.
 124.Marino G, Pietrocola F, Eisenberg T, Kong Y, Malik SA, Andryushkova A, Schroeder S, Pendl T, Harger A, Niso‐Santano M, Zamzami N, Scoazec M, Durand S, Enot DP, Fernandez AF, Martins I, Kepp O, Senovilla L, Bauvy C, Morselli E, Vacchelli E, Bennetzen M, Magnes C, Sinner F, Pieber T, Lopez‐Otin C, Maiuri MC, Codogno P, Andersen JS, Hill JA, Madeo F, Kroemer G. Regulation of autophagy by cytosolic acetyl‐coenzyme A. Mol Cell 53: 710‐725, 2014.
 125.Martin B, Pearson M, Kebejian L, Golden E, Keselman A, Bender M, Carlson O, Egan J, Ladenheim B, Cadet JL, Becker KG, Wood W, Duffy K, Vinayakumar P, Maudsley S, Mattson MP. Sex‐dependent metabolic, neuroendocrine, and cognitive responses to dietary energy restriction and excess. Endocrinology 148: 4318‐4333, 2007.
 126.Martinez‐Lopez N, Tarabra E, Toledo M, Garcia‐Macia M, Sahu S, Coletto L, Batista‐Gonzalez A, Barzilai N, Pessin JE, Schwartz GJ, Kersten S, Singh R. System‐wide benefits of intermeal fasting by autophagy. Cell Metab 26: 856‐871 e855, 2017.
 127.Mattson MP, Longo VD, Harvie M. Impact of intermittent fasting on health and disease processes. Ageing Res Rev 39: 46‐58, 2017.
 128.McGinnis GR, Tang Y, Brewer RA, Brahma MK, Stanley HL, Shanmugam G, Rajasekaran NS, Rowe GC, Frank SJ, Wende AR, Abel ED, Taegtmeyer H, Litovsky S, Darley‐Usmar V, Zhang J, Chatham JC, Young ME. Genetic disruption of the cardiomyocyte circadian clock differentially influences insulin‐mediated processes in the heart. J Mol Cell Cardiol 110: 80‐95, 2017.
 129.Medina DL, Di Paola S, Peluso I, Armani A, De Stefani D, Venditti R, Montefusco S, Scotto‐Rosato A, Prezioso C, Forrester A, Settembre C, Wang W, Gao Q, Xu H, Sandri M, Rizzuto R, De Matteis MA, Ballabio A. Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat Cell Biol 17: 288‐299, 2015.
 130.Meikle PJ, Hopwood JJ, Clague AE, Carey WF. Prevalence of lysosomal storage disorders. JAMA 281: 249‐254, 1999.
 131.Meyer TE, Kovacs SJ, Ehsani AA, Klein S, Holloszy JO, Fontana L. Long‐term caloric restriction ameliorates the decline in diastolic function in humans. J Am Coll Cardiol 47: 398‐402, 2006.
 132.Mialet‐Perez J, Vindis C. Autophagy in health and disease: Focus on the cardiovascular system. Essays Biochem 61: 721‐732, 2017.
 133.Michalsen A, Riegert M, Ludtke R, Backer M, Langhorst J, Schwickert M, Dobos GJ. Mediterranean diet or extended fasting's influence on changing the intestinal microflora, immunoglobulin A secretion and clinical outcome in patients with rheumatoid arthritis and fibromyalgia: An observational study. BMC Complement Altern Med 5: 22, 2005.
 134.Mitchell G, Isberg RR. Innate immunity to intracellular pathogens: Balancing microbial elimination and inflammation. Cell Host Microbe 22: 166‐175, 2017.
 135.Mizushima N, Komatsu M. Autophagy: Renovation of cells and tissues. Cell 147: 728‐741, 2011.
 136.Mizushima N, Noda T, Yoshimori T, Tanaka Y, Ishii T, George MD, Klionsky DJ, Ohsumi M, Ohsumi Y. A protein conjugation system essential for autophagy. Nature 395: 395‐398, 1998.
 137.Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell 15: 1101‐1111, 2004.
 138.Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, Bultsma Y, McBurney M, Guarente L. Mammalian SIRT1 represses forkhead transcription factors. Cell 116: 551‐563, 2004.
 139.Muller H, de Toledo FW, Resch KL. Fasting followed by vegetarian diet in patients with rheumatoid arthritis: A systematic review. Scand J Rheumatol 30: 1‐10, 2001.
 140.Nah J, Miyamoto S, Sadoshima J. Mitophagy as a protective mechanism against myocardial stress. Compr Physiol 7: 1407‐1424, 2017.
 141.Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I, Matsumura Y, Asahi M, Nishida K, Hori M, Mizushima N, Otsu K. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med 13: 619‐624, 2007.
 142.Nakatogawa H, Suzuki K, Kamada Y, Ohsumi Y. Dynamics and diversity in autophagy mechanisms: Lessons from yeast. Nat Rev Mol Cell Biol 10: 458‐467, 2009.
 143.Newman JC, Covarrubias AJ, Zhao M, Yu X, Gut P, Ng CP, Huang Y, Haldar S, Verdin E. Ketogenic diet reduces midlife mortality and improves memory in aging mice. Cell Metab 26: 547‐557 e548, 2017.
 144.Nezich CL, Wang C, Fogel AI, Youle RJ. MiT/TFE transcription factors are activated during mitophagy downstream of Parkin and Atg5. J Cell Biol 210: 435‐450, 2015.
 145.Nishino I, Fu J, Tanji K, Yamada T, Shimojo S, Koori T, Mora M, Riggs JE, Oh SJ, Koga Y, Sue CM, Yamamoto A, Murakami N, Shanske S, Byrne E, Bonilla E, Nonaka I, DiMauro S, Hirano M. Primary LAMP‐2 deficiency causes X‐linked vacuolar cardiomyopathy and myopathy (Danon disease). Nature 406: 906‐910, 2000.
 146.O'Neill JS, Reddy AB. Circadian clocks in human red blood cells. Nature 469: 498‐503, 2011.
 147.O'Neill JS, van Ooijen G, Dixon LE, Troein C, Corellou F, Bouget FY, Reddy AB, Millar AJ. Circadian rhythms persist without transcription in a eukaryote. Nature 469: 554‐558, 2011.
 148.O'Rourke EJ, Ruvkun G. MXL‐3 and HLH‐30 transcriptionally link lipolysis and autophagy to nutrient availability. Nat Cell Biol 15: 668‐676, 2013.
 149.Oakhill JS, Steel R, Chen ZP, Scott JW, Ling N, Tam S, Kemp BE. AMPK is a direct adenylate charge‐regulated protein kinase. Science 332: 1433‐1435, 2011.
 150.Oka S, Alcendor R, Zhai P, Park JY, Shao D, Cho J, Yamamoto T, Tian B, Sadoshima J. PPARalpha‐Sirt1 complex mediates cardiac hypertrophy and failure through suppression of the ERR transcriptional pathway. Cell Metab 14: 598‐611, 2011.
 151.Ouwens DM, Boer C, Fodor M, de Galan P, Heine RJ, Maassen JA, Diamant M. Cardiac dysfunction induced by high‐fat diet is associated with altered myocardial insulin signalling in rats. Diabetologia 48: 1229‐1237, 2005.
 152.Palmieri M, Impey S, Kang H, di RA, Pelz C, Sardiello M, Ballabio A. Characterization of the CLEAR network reveals an integrated control of cellular clearance pathways. Hum Mol Genet 20: 3852‐3866, 2011.
 153.Panda S. Circadian physiology of metabolism. Science 354: 1008‐1015, 2016.
 154.Panda S, Antoch MP, Miller BH, Su AI, Schook AB, Straume M, Schultz PG, Kay SA, Takahashi JS, Hogenesch JB. Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109: 307‐320, 2002.
 155.Park S, Yoo KM, Hyun JS, Kang S. Intermittent fasting reduces body fat but exacerbates hepatic insulin resistance in young rats regardless of high protein and fat diets. J Nutr Biochem 40: 14‐22, 2017.
 156.Peliciari‐Garcia RA, Goel M, Aristorenas JA, Shah K, He L, Yang Q, Shalev A, Bailey SM, Prabhu SD, Chatham JC, Gamble KL, Young ME. Altered myocardial metabolic adaptation to increased fatty acid availability in cardiomyocyte‐specific CLOCK mutant mice. Biochim Biophys Acta 1861: 1579‐1595, 2016.
 157.Pendergast JS, Nakamura W, Friday RC, Hatanaka F, Takumi T, Yamazaki S. Robust food anticipatory activity in BMAL1‐deficient mice. PloS One 4: e4860, 2009.
 158.Pendergast JS, Wendroth RH, Stenner RC, Keil CD, Yamazaki S. mPeriod2 (Brdm1) and other single period mutant mice have normal food anticipatory activity. Sci Rep 7: 15510, 2017.
 159.Perelis M, Marcheva B, Ramsey KM, Schipma MJ, Hutchison AL, Taguchi A, Peek CB, Hong H, Huang W, Omura C, Allred AL, Bradfield CA, Dinner AR, Barish GD, Bass J. Pancreatic beta cell enhancers regulate rhythmic transcription of genes controlling insulin secretion. Science 350: aac4250, 2015.
 160.Perera RM, Zoncu R. The lysosome as a regulatory hub. Annu Rev Cell Dev Biol 32: 223‐253, 2016.
 161.Platt FM, Boland B, van der Spoel AC. The cell biology of disease: Lysosomal storage disorders: The cellular impact of lysosomal dysfunction. J Cell Biol 199: 723‐734, 2012.
 162.Polito VA, Li H, Martini‐Stoica H, Wang B, Yang L, Xu Y, Swartzlander DB, Palmieri M, di Ronza A, Lee VM, Sardiello M, Ballabio A, Zheng H. Selective clearance of aberrant tau proteins and rescue of neurotoxicity by transcription factor EB. EMBO Mol Med 6: 1142‐1160, 2014.
 163.Purushotham A, Schug TT, Xu Q, Surapureddi S, Guo X, Li X. Hepatocyte‐specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab 9: 327‐338, 2009.
 164.Ravikumar B, Moreau K, Jahreiss L, Puri C, Rubinsztein DC. Plasma membrane contributes to the formation of pre‐autophagosomal structures. Nat Cell Biol 12: 747‐757, 2010.
 165.Razani B, Zhang H, Schulze PC, Schilling JD, Verbsky J, Lodhi IJ, Topkara VK, Feng C, Coleman T, Kovacs A, Kelly DP, Saffitz JE, Dorn GW, II, Nichols CG, Semenkovich CF. Fatty acid synthase modulates homeostatic responses to myocardial stress. J Biol Chem 286: 30949‐30961, 2011.
 166.Reichelt ME, Mellor KM, Curl CL, Stapleton D, Delbridge LM. Myocardial glycophagy‐‐‐A specific glycogen handling response to metabolic stress is accentuated in the female heart. J Mol Cell Cardiol 65: 67‐75, 2013.
 167.Riehle C, Wende AR, Sena S, Pires KM, Pereira RO, Zhu Y, Bugger H, Frank D, Bevins J, Chen D, Perry CN, Dong XC, Valdez S, Rech M, Sheng X, Weimer BC, Gottlieb RA, White MF, Abel ED. Insulin receptor substrate signaling suppresses neonatal autophagy in the heart. J Clin Invest 123: 5319‐5333, 2013.
 168.Riordan MM, Weiss EP, Meyer TE, Ehsani AA, Racette SB, Villareal DT, Fontana L, Holloszy JO, Kovacs SJ. The effects of caloric restriction‐ and exercise‐induced weight loss on left ventricular diastolic function. Am J Physiol Heart Circ Physiol 294: H1174‐H1182, 2008.
 169.Roberts MN, Wallace MA, Tomilov AA, Zhou Z, Marcotte GR, Tran D, Perez G, Gutierrez‐Casado E, Koike S, Knotts TA, Imai DM, Griffey SM, Kim K, Hagopian K, Haj FG, Baar K, Cortopassi GA, Ramsey JJ, Lopez‐Dominguez JA. A ketogenic diet extends longevity and healthspan in adult mice. Cell Metab 26: 539‐546 e535, 2017.
 170.Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC‐1alpha and SIRT1. Nature 434: 113‐118, 2005.
 171.Rowley WR, Bezold C, Arikan Y, Byrne E, Krohe S. Diabetes 2030: Insights from yesterday, today, and future trends. Popul Health Manag 20: 6‐12, 2017.
 172.Sabatini DD, Adesnik M. Christian de Duve: Explorer of the cell who discovered new organelles by using a centrifuge. Proc Natl Acad Sci U S A 110: 13234‐13235, 2013.
 173.Sacks FM, Bray GA, Carey VJ, Smith SR, Ryan DH, Anton SD, McManus K, Champagne CM, Bishop LM, Laranjo N, Leboff MS, Rood JC, de Jonge L, Greenway FL, Loria CM, Obarzanek E, Williamson DA. Comparison of weight‐loss diets with different compositions of fat, protein, and carbohydrates. N Engl J Med 360: 859‐873, 2009.
 174.Samovski D, Sun J, Pietka T, Gross RW, Eckel RH, Su X, Stahl PD, Abumrad NA. Regulation of AMPK activation by CD36 links fatty acid uptake to beta‐oxidation. Diabetes 64: 353‐359, 2015.
 175.Sardiello M, Palmieri M, di RA, Medina DL, Valenza M, Gennarino VA, Di MC, Donaudy F, Embrione V, Polishchuk RS, Banfi S, Parenti G, Cattaneo E, Ballabio A. A gene network regulating lysosomal biogenesis and function. Science 325: 473‐477, 2009.
 176.Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell 168: 960‐976, 2017.
 177.Schroeder S, Pendl T, Zimmermann A, Eisenberg T, Carmona‐Gutierrez D, Ruckenstuhl C, Marino G, Pietrocola F, Harger A, Magnes C, Sinner F, Pieber TR, Dengjel J, Sigrist SJ, Kroemer G, Madeo F. Acetyl‐coenzyme A: A metabolic master regulator of autophagy and longevity. Autophagy 10: 1335‐1337, 2014.
 178.Sciarretta S, Forte M, Frati G, Sadoshima J. New insights into the role of mTOR signaling in the cardiovascular system. Circ Res 122: 489‐505, 2018.
 179.Sciarretta S, Yee D, Nagarajan N, Bianchi F, Saito T, Valenti V, Tong M, Del Re DP, Vecchione C, Schirone L, Forte M, Rubattu S, Shirakabe A, Boppana VS, Volpe M, Frati G, Zhai P, Sadoshima J. Trehalose‐induced activation of autophagy improves cardiac remodeling after myocardial infarction. J Am Coll Cardiol 71: 1999‐2010, 2018.
 180.Sciarretta S, Zhai P, Shao D, Maejima Y, Robbins J, Volpe M, Condorelli G, Sadoshima J. Rheb is a critical regulator of autophagy during myocardial ischemia: Pathophysiological implications in obesity and metabolic syndrome. Circulation 125: 1134‐1146, 2012.
 181.Sen S, Kundu BK, Wu HC, Hashmi SS, Guthrie P, Locke LW, Roy RJ, Matherne GP, Berr SS, Terwelp M, Scott B, Carranza S, Frazier OH, Glover DK, Dillmann WH, Gambello MJ, Entman ML, Taegtmeyer H. Glucose regulation of load‐induced mTOR signaling and ER stress in mammalian heart. J Am Heart Assoc 2: e004796, 2013.
 182.Seok S, Fu T, Choi SE, Li Y, Zhu R, Kumar S, Sun X, Yoon G, Kang Y, Zhong W, Ma J, Kemper B, Kemper JK. Transcriptional regulation of autophagy by an FXR‐CREB axis. Nature 516: 108‐111, 2014.
 183.Sergin I, Evans TD, Zhang X, Bhattacharya S, Stokes CJ, Song E, Ali S, Dehestani B, Holloway KB, Micevych PS, Javaheri A, Crowley JR, Ballabio A, Schilling JD, Epelman S, Weihl CC, Diwan A, Fan D, Zayed MA, Razani B. Exploiting macrophage autophagy‐lysosomal biogenesis as a therapy for atherosclerosis. Nat Commun 8: 15750, 2017.
 184.Settembre C, Ballabio A. Lysosome: Regulator of lipid degradation pathways. Trends Cell Biol 24: 743‐750, 2014.
 185.Settembre C, De CR, Mansueto G, Saha PK, Vetrini F, Visvikis O, Huynh T, Carissimo A, Palmer D, Jurgen KT, Wollenberg AC, Di BD, Chan L, Irazoqui JE, Ballabio A. TFEB controls cellular lipid metabolism through a starvation‐induced autoregulatory loop. Nat Cell Biol 15(6): 647‐658, 2013.
 186.Settembre C, Fraldi A, Medina DL, Ballabio A. Signals from the lysosome: A control centre for cellular clearance and energy metabolism. Nat Rev Mol Cell Biol 14: 283‐296, 2013.
 187.Settembre C, Zoncu R, Medina DL, Vetrini F, Erdin S, Erdin S, Huynh T, Ferron M, Karsenty G, Vellard MC, Facchinetti V, Sabatini DM, Ballabio A. A lysosome‐to‐nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB. EMBO J 31: 1095‐1108, 2012.
 188.Shao D, Tian R. Glucose transporters in cardiac metabolism and hypertrophy. Compr Physiol 6: 331‐351, 2015.
 189.Sheng Y, Lv S, Huang M, Lv Y, Yu J, Liu J, Tang T, Qi H, Di W, Ding G. Opposing effects on cardiac function by calorie restriction in different‐aged mice. Aging Cell 16: 1155‐1167, 2017.
 190.Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD, Newgard CB, Farese RV, Jr., de Cabo R, Ulrich S, Akassoglou K, Verdin E. Suppression of oxidative stress by beta‐hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339: 211‐214, 2013.
 191.Shinmura K, Tamaki K, Bolli R. Short‐term caloric restriction improves ischemic tolerance independent of opening of ATP‐sensitive K+ channels in both young and aged hearts. J Mol Cell Cardiol 39: 285‐296, 2005.
 192.Shinmura K, Tamaki K, Sano M, Murata M, Yamakawa H, Ishida H, Fukuda K. Impact of long‐term caloric restriction on cardiac senescence: Caloric restriction ameliorates cardiac diastolic dysfunction associated with aging. J Mol Cell Cardiol 50: 117‐127, 2011.
 193.Shirakabe A, Ikeda Y, Sciarretta S, Zablocki DK, Sadoshima J. Aging and autophagy in the heart. Circ Res 118: 1563‐1576, 2016.
 194.Shirakabe A, Zhai P, Ikeda Y, Saito T, Maejima Y, Hsu CP, Nomura M, Egashira K, Levine B, Sadoshima J. Drp1‐dependent mitochondrial autophagy plays a protective role against pressure overload‐induced mitochondrial dysfunction and heart failure. Circulation 133: 1249‐1263, 2016.
 195.Sica V, Galluzzi L, Bravo‐San Pedro JM, Izzo V, Maiuri MC, Kroemer G. Organelle‐specific initiation of autophagy. Mol Cell 59: 522‐539, 2015.
 196.Simonson B, Subramanya V, Chan MC, Zhang A, Franchino H, Ottaviano F, Mishra MK, Knight AC, Hunt D, Ghiran I, Khurana TS, Kontaridis MI, Rosenzweig A, Das S. DDiT4L promotes autophagy and inhibits pathological cardiac hypertrophy in response to stress. Sci Signal 10(468): pii: eaaf5967, 2017.
 197.Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, Tanaka K, Cuervo AM, Czaja MJ. Autophagy regulates lipid metabolism. Nature 458: 1131‐1135, 2009.
 198.Sletten AC, Peterson LR, Schaffer JE. Manifestations and mechanisms of myocardial lipotoxicity in obesity. J Intern Med 2018. doi: 10.1111/joim.12728.
 199.Song M, Franco A, Fleischer JA, Zhang L, Dorn GW, II. Abrogating mitochondrial dynamics in mouse hearts accelerates mitochondrial senescence. Cell Metab 26: 872‐883 e875, 2017.
 200.Song M, Gong G, Burelle Y, Gustafsson AB, Kitsis RN, Matkovich SJ, Dorn GW, II. Interdependence of Parkin‐mediated mitophagy and mitochondrial fission in adult mouse hearts. Circ Res 117(4): 346‐351, 2015.
 201.Song M, Mihara K, Chen Y, Scorrano L, Dorn GW, II. Mitochondrial fission and fusion factors reciprocally orchestrate mitophagic culling in mouse hearts and cultured fibroblasts. Cell Metab 21: 273‐286, 2015.
 202.Spampanato C, Feeney E, Li L, Cardone M, Lim JA, Annunziata F, Zare H, Polishchuk R, Puertollano R, Parenti G, Ballabio A, Raben N. Transcription factor EB (TFEB) is a new therapeutic target for Pompe disease. EMBO Mol Med 5: 691‐706, 2013.
 203.St‐Onge MP, Ard J, Baskin ML, Chiuve SE, Johnson HM, Kris‐Etherton P, Varady K, American Heart Association Obesity Committee of the Council on L, Cardiometabolic H, Council on Cardiovascular Disease in the Y, Council on Clinical C, Stroke C. Meal timing and frequency: Implications for cardiovascular disease prevention: A scientific statement from the American Heart Association. Circulation 135: e96‐e121, 2017.
 204.Stanley S, Wynne K, McGowan B, Bloom S. Hormonal regulation of food intake. Physiol Rev 85: 1131‐1158, 2005.
 205.Stein PK, Soare A, Meyer TE, Cangemi R, Holloszy JO, Fontana L. Caloric restriction may reverse age‐related autonomic decline in humans. Aging Cell 11: 644‐650, 2012.
 206.Stypmann J, Janssen PM, Prestle J, Engelen MA, Kogler H, Lullmann‐Rauch R, Eckardt L, von FK, Landgrebe J, Mleczko A, Saftig P. LAMP‐2 deficient mice show depressed cardiac contractile function without significant changes in calcium handling. Basic Res Cardiol 101: 281‐291, 2006.
 207.Sun H, Olson KC, Gao C, Prosdocimo DA, Zhou M, Wang Z, Jeyaraj D, Youn JY, Ren S, Liu Y, Rau CD, Shah S, Ilkayeva O, Gui WJ, William NS, Wynn RM, Newgard CB, Cai H, Xiao X, Chuang DT, Schulze PC, Lynch C, Jain MK, Wang Y. Catabolic defect of branched‐chain amino acids promotes heart failure. Circulation 133: 2038‐2049, 2016.
 208.Sun Y, Xu YH, Du H, Quinn B, Liou B, Stanton L, Inskeep V, Ran H, Jakubowitz P, Grilliot N, Grabowski GA. Reversal of advanced disease in lysosomal acid lipase deficient mice: A model for lysosomal acid lipase deficiency disease. Mol Genet Metab 112: 229‐241, 2014.
 209.Sutton EF, Beyl R, Early KS, Cefalu WT, Ravussin E, Peterson CM. Early time‐restricted feeding improves insulin sensitivity, blood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab 27(6): 1212‐1221.e3, 2018.
 210.Svendsen PF, Jensen FK, Holst JJ, Haugaard SB, Nilas L, Madsbad S. The effect of a very low calorie diet on insulin sensitivity, beta cell function, insulin clearance, incretin hormone secretion, androgen levels and body composition in obese young women. Scand J Clin Lab Invest 72: 410‐419, 2012.
 211.Taegtmeyer H, Lam T, Davogustto G. Cardiac metabolism in perspective. Compr Physiol 6: 1675‐1699, 2016.
 212.Taegtmeyer H, Wilson CR, Razeghi P, Sharma S. Metabolic energetics and genetics in the heart. Ann N Y Acad Sci 1047: 208‐218, 2005.
 213.Taegtmeyer H, Young ME, Lopaschuk GD, Abel ED, Brunengraber H, Darley‐Usmar V, Des Rosiers C, Gerszten R, Glatz JF, Griffin JL, Gropler RJ, Holzhuetter HG, Kizer JR, Lewandowski ED, Malloy CR, Neubauer S, Peterson LR, Portman MA, Recchia FA, Van Eyk JE, Wang TJ, American Heart Association Council on Basic Cardiovascular S. Assessing cardiac metabolism: A scientific statement from the American Heart Association. Circ Res 118: 1659‐1701, 2016.
 214.Taffet GE, Pham TT, Hartley CJ. The age‐associated alterations in late diastolic function in mice are improved by caloric restriction. J Gerontol A Biol Sci Med Sci 52: B285‐B290, 1997.
 215.Takagi A, Kume S, Kondo M, Nakazawa J, Chin‐Kanasaki M, Araki H, Araki S, Koya D, Haneda M, Chano T, Matsusaka T, Nagao K, Adachi Y, Chan L, Maegawa H, Uzu T. Mammalian autophagy is essential for hepatic and renal ketogenesis during starvation. Sci Rep 6: 18944, 2016.
 216.Tanaka Y, Guhde G, Suter A, Eskelinen EL, Hartmann D, Lullmann‐Rauch R, Janssen PM, Blanz J, von Figura K, Saftig P. Accumulation of autophagic vacuoles and cardiomyopathy in LAMP‐2‐deficient mice. Nature 406: 902‐906, 2000.
 217.Traba J, Kwarteng‐Siaw M, Okoli TC, Li J, Huffstutler RD, Bray A, Waclawiw MA, Han K, Pelletier M, Sauve AA, Siegel RM, Sack MN. Fasting and refeeding differentially regulate NLRP3 inflammasome activation in human subjects. J Clin Invest 125: 4592‐4600, 2015.
 218.Trepanowski JF, Kroeger CM, Barnosky A, Klempel MC, Bhutani S, Hoddy KK, Gabel K, Freels S, Rigdon J, Rood J, Ravussin E, Varady KA. Effect of alternate‐day fasting on weight loss, weight maintenance, and cardioprotection among metabolically healthy obese adults: A randomized clinical trial. JAMA Intern Med 177: 930‐938, 2017.
 219.Tsai JY, Villegas‐Montoya C, Boland BB, Blasier Z, Egbejimi O, Gonzalez R, Kueht M, McElfresh TA, Brewer RA, Chandler MP, Bray MS, Young ME. Influence of dark phase restricted high fat feeding on myocardial adaptation in mice. J Mol Cell Cardiol 55: 147‐155, 2013.
 220.Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon E, Laposky A, Losee‐Olson S, Easton A, Jensen DR, Eckel RH, Takahashi JS, Bass J. Obesity and metabolic syndrome in circadian Clock mutant mice. Science 308: 1043‐1045, 2005.
 221.Varady KA, Bhutani S, Church EC, Klempel MC. Short‐term modified alternate‐day fasting: A novel dietary strategy for weight loss and cardioprotection in obese adults. Am J Clin Nutr 90: 1138‐1143, 2009.
 222.Varady KA, Bhutani S, Klempel MC, Kroeger CM, Trepanowski JF, Haus JM, Hoddy KK, Calvo Y. Alternate day fasting for weight loss in normal weight and overweight subjects: A randomized controlled trial. Nutr J 12: 146, 2013.
 223.Wang B, Nie J, Wu L, Hu Y, Wen Z, Dong L, Zou MH, Chen C, Wang DW. AMPKalpha2 protects against the development of heart failure by enhancing mitophagy via PINK1 phosphorylation. Circ Res 122: 712‐729, 2018.
 224.Wang C, Niederstrasser H, Douglas PM, Lin R, Jaramillo J, Li Y, Olswald NW, Zhou A, McMillan EA, Mendiratta S, Wang Z, Zhao T, Lin Z, Luo M, Huang G, Brekken RA, Posner BA, MacMillan JB, Gao J, White MA. Small‐molecule TFEB pathway agonists that ameliorate metabolic syndrome in mice and extend C. elegans lifespan. Nat Commun 8: 2270, 2017.
 225.Wang RH, Kim HS, Xiao C, Xu X, Gavrilova O, Deng CX. Hepatic Sirt1 deficiency in mice impairs mTorc2/Akt signaling and results in hyperglycemia, oxidative damage, and insulin resistance. J Clin Invest 121: 4477‐4490, 2011.
 226.Warren JS, Oka SI, Zablocki D, Sadoshima J. Metabolic reprogramming via PPARalpha signaling in cardiac hypertrophy and failure: From metabolomics to epigenetics. Am J Physiol Heart Circ Physiol 313: H584‐H596, 2017.
 227.Wei M, Brandhorst S, Shelehchi M, Mirzaei H, Cheng CW, Budniak J, Groshen S, Mack WJ, Guen E, Di Biase S, Cohen P, Morgan TE, Dorff T, Hong K, Michalsen A, Laviano A, Longo VD. Fasting‐mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease. Sci Trans Med 9(377): pii: eaai8700, 2017.
 228.Weir HJ, Yao P, Huynh FK, Escoubas CC, Goncalves RL, Burkewitz K, Laboy R, Hirschey MD, Mair WB. Dietary restriction and AMPK increase lifespan via mitochondrial network and peroxisome remodeling. Cell Metab 26: 884‐896 e885, 2017.
 229.Wing RR. Long‐term effects of a lifestyle intervention on weight and cardiovascular risk factors in individuals with type 2 diabetes mellitus: Four‐year results of the Look AHEAD trial. Arch Intern Med 170: 1566‐1575, 2010.
 230.Wohlgemuth SE, Julian D, Akin DE, Fried J, Toscano K, Leeuwenburgh C, Dunn WA, Jr. Autophagy in the heart and liver during normal aging and calorie restriction. Rejuvenation Res 10: 281‐292, 2007.
 231.Wolfson RL, Sabatini DM. The dawn of the age of amino acid sensors for the mTORC1 pathway. Cell Metab 26(2): 301‐309, 2017.
 232.Wolins NE, Quaynor BK, Skinner JR, Tzekov A, Croce MA, Gropler MC, Varma V, Yao‐Borengasser A, Rasouli N, Kern PA, Finck BN, Bickel PE. OXPAT/PAT‐1 is a PPAR‐induced lipid droplet protein that promotes fatty acid utilization. Diabetes 55: 3418‐3428, 2006.
 233.Woloszynek JC, Kovacs A, Ohlemiller KK, Roberts M, Sands MS. Metabolic adaptations to interrupted glycosaminoglycan recycling. J Biol Chem 284: 29684‐29691, 2009.
 234.Wood JG, Rogina B, Lavu S, Howitz K, Helfand SL, Tatar M, Sinclair D. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 430: 686‐689, 2004.
 235.Writing Group M, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER, III, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB, American Heart Association Statistics C, Stroke Statistics S. Heart Disease and Stroke Statistics‐2016 update: A report from the American Heart Association. Circulation 133: e38‐e360, 2016.
 236.Xiao Q, Yan P, Ma X, Liu H, Perez R, Zhu A, Gonzales E, Burchett JM, Schuler DR, Cirrito JR, Diwan A, Lee JM. Enhancing astrocytic lysosome biogenesis facilitates abeta clearance and attenuates amyloid plaque pathogenesis. J Neurosci 34: 9607‐9620, 2014.
 237.Xiao Q, Yan P, Ma X, Liu H, Perez R, Zhu A, Gonzales E, Tripoli DD, Czerniewski L, Ballabio A, Cirrito JR, Diwan A, Lee JM. Neuronal‐targeted TFEB accelerates lysosomal degradation of APP, reducing Aβ generation and amyloid plaque pathogenesis. J Neurosci 35: 12137‐12151, 2015.
 238.Xie K, Neff F, Markert A, Rozman J, Aguilar‐Pimentel JA, Amarie OV, Becker L, Brommage R, Garrett L, Henzel KS, Holter SM, Janik D, Lehmann I, Moreth K, Pearson BL, Racz I, Rathkolb B, Ryan DP, Schroder S, Treise I, Bekeredjian R, Busch DH, Graw J, Ehninger G, Klingenspor M, Klopstock T, Ollert M, Sandholzer M, Schmidt‐Weber C, Weiergraber M, Wolf E, Wurst W, Zimmer A, Gailus‐Durner V, Fuchs H, Hrabe de Angelis M, Ehninger D. Every‐other‐day feeding extends lifespan but fails to delay many symptoms of aging in mice. Nat Commun 8: 155, 2017.
 239.Yamamoto T, Tamaki K, Shirakawa K, Ito K, Yan X, Katsumata Y, Anzai A, Matsuhashi T, Endo J, Inaba T, Tsubota K, Sano M, Fukuda K, Shinmura K. Cardiac Sirt1 mediates the cardioprotective effect of caloric restriction by suppressing local complement system activation after ischemia‐reperfusion. Am J Physiol Heart Circ Physiol 310: H1003‐H1014, 2016.
 240.Yan L, Park JY, Dillinger JG, De Lorenzo MS, Yuan C, Lai L, Wang C, Ho D, Tian B, Stanley WC, Auwerx J, Vatner DE, Vatner SF. Common mechanisms for calorie restriction and adenylyl cyclase type 5 knockout models of longevity. Aging Cell 11: 1110‐1120, 2012.
 241.Yan L, Vatner DE, O'Connor JP, Ivessa A, Ge H, Chen W, Hirotani S, Ishikawa Y, Sadoshima J, Vatner SF. Type 5 adenylyl cyclase disruption increases longevity and protects against stress. Cell 130: 247‐258, 2007.
 242.Yang J, Sambandam N, Han X, Gross RW, Courtois M, Kovacs A, Febbraio M, Finck BN, Kelly DP. CD36 deficiency rescues lipotoxic cardiomyopathy. Circ Res 100: 1208‐1217, 2007.
 243.Yang KC, Ma X, Liu H, Murphy J, Barger PM, Mann DL, Diwan A. Tumor necrosis factor receptor‐associated factor 2 mediates mitochondrial autophagy. Circ Heart Fail 8: 175‐187, 2015.
 244.Yoshii SR, Kuma A, Akashi T, Hara T, Yamamoto A, Kurikawa Y, Itakura E, Tsukamoto S, Shitara H, Eishi Y, Mizushima N. Systemic analysis of Atg5‐null mice rescued from neonatal lethality by transgenic ATG5 expression in neurons. Dev Cell 39: 116‐130, 2016.
 245.Yoshino J, Baur JA, Imai SI. NAD(+) intermediates: The biology and therapeutic potential of NMN and NR. Cell Metab 27(3): 513‐528, 2017.
 246.Yousefi S, Perozzo R, Schmid I, Ziemiecki A, Schaffner T, Scapozza L, Brunner T, Simon HU. Calpain‐mediated cleavage of Atg5 switches autophagy to apoptosis. Nat Cell Biol 8: 1124‐1132, 2006.
 247.Yu L, McPhee CK, Zheng L, Mardones GA, Rong Y, Peng J, Mi N, Zhao Y, Liu Z, Wan F, Hailey DW, Oorschot V, Klumperman J, Baehrecke EH, Lenardo MJ. Termination of autophagy and reformation of lysosomes regulated by mTOR. Nature 465: 942‐946, 2010.
 248.Zhou J, Freeman TA, Ahmad F, Shang X, Mangano E, Gao E, Farber J, Wang Y, Ma XL, Woodgett J, Vagnozzi RJ, Lal H, Force T. GSK‐3alpha is a central regulator of age‐related pathologies in mice. J Clin Invest 123: 1821‐1832, 2013.
 249.Zoncu R, Bar‐Peled L, Efeyan A, Wang S, Sancak Y, Sabatini DM. mTORC1 senses lysosomal amino acids through an inside‐out mechanism that requires the vacuolar H(+)‐ATPase. Science 334: 678‐683, 2011.
 250.Zuo L, He F, Tinsley GM, Pannell BK, Ward E, Arciero PJ. Comparison of high‐protein, intermittent fasting low‐calorie diet and heart healthy diet for vascular health of the obese. Front Physiol 7: 350, 2016.

 

 

 

 

Teaching Material

 

 

K. Mani, A. Javaheri, A. Diwan. Lysosomes Mediate Benefits of Intermittent Fasting in Cardiometabolic Disease: The Janitor Is the Undercover Boss. Compr Physiol 8: 2018, 1639-1667.

Didactic Synopsis

Major Teaching Points:

  • Lysosomes play a critical role in cardiac myocyte homeostasis and preserving normal cardiac structure and function.
  • Lysosomes regulate substrate supply to maintain cardiac energetics and maintain organelle quality by degrading damaged organelles via autophagy, to promote cardiac myocyte homeostasis in both fed and fasted states.
  • Lysosomal nutrient sensing (LYNUS) complex plays a critical role in transcriptional control of autophagy in cardiac myocytes.
  • Intermittent fasting stimulates the lysosome machinery to precondition the myocardium, and attenuate effects of injurious stimuli as well as attenuate development of adverse ventricular remodeling in response.
  • Intermittent fasting benefits cardiometabolic parameters in healthy humans, as well as those suffering from diabetes or other risk factors for development of cardiac diseases.

Didactic Legends

The figures—in a freely downloadable PowerPoint format—can be found on the Images tab along with the formal legends published in the article. The following legends to the same figures are written to be useful for teaching.

Figure 1 Teaching points: The heart is omnivorous in its substrate preference and can generate energy with breakdown of a diverse set of nutrients. In physiologic states, fatty acid oxidation and glucose oxidation are the first and second most preferred substrates. Under fasting stress, ketone bodies and amino acids are increasing utilized. Under pathologic stress such as with ischemia or ischemia-reperfusion injury or in disease states, this metabolic flexibility is reduced and the heart increasingly relies on glucose (and ketone bodies) as the primary source for energy, except in the diabetic myocardium wherein fatty acid and ketone metabolism are upregulated at the expense of glucose utilization.

Figure 2 Teaching points: Autophagy, a lysosomal degradative process for breakdown of intracellular constituents, happens via three distinct pathways: macroautophagy, microautophagy, and chaperone-mediated autophagy. In macroautophagy, formation of double-membrane bound autophagosomes happens constitutively (at basal levels) or is triggered in response to stress. A coordinated system of conjugating enzymatic systems (orchestrated by ATG proteins) causes nucleation of a preautophagosomal membrane followed by extension and closure to enclose cargo targeted for degradation. The cargo is typically identified based upon presence of ubiquitin tags on proteins which are brought into the autophagosomes via an interaction with adaptor proteins such as p62 that interact with the LC3 family of proteins. The LC3 proteins are lipidated and embedded on both autophagosome membranes to permit this interaction. Subsequently, a mature autophagosome rapidly fuses with a lysosome to form a autolysosome wherein the cargo is degraded. Lysosomes support the LYNUS complex on cytosolic face wherein mTOR binds and responds to nutrient cues to regulate activity of the TFEB family of transcription factors, which transcriptionally regulate lysosome biogenesis as well as induction of autophagy. Chaperone-mediated autophagy facilitates uptake of proteins bearing the “KFERQ” motif through interactions with a lysosomal membrane protein LAMP2A into the lysosome for degradation. Microautophagy involves direct lysosomal invagination to take up cytosolic cargo for degradation.

Figure 3 Teaching points: Autophagy and lysosome biogenesis program are highly regulated processes that are controlled by various environmental and genetic cues. Various mechanisms activate autophagy in response to organelle damage to orchestrate selective autophagy, as summarized herein. Autophagy is also induced as the backup degradative program with impairment of the ubiquitin-proteasome machinery that degrades proteins. Metabolic cues directly control autophagy-lysosome pathway activation and transcription. Various transcriptional pathways regulates autophagy in response to fasting, with a complex interplay of transcriptional activators (FOXO, TFEB, CREB, and CRTC2) and repressors (MAX and FXR) to control lipolysis programs (via PPARα) to breakdown lipids for energy generation. Circadian cues are likely to regulate the autophagy-lysosome machinery although the specific signaling pathways remain to be worked out. Aging, by and large, has negative effects on autophagy-lysosome system, and this phenomenon likely contributes to aging-related diseases with loss of the protective effects.

Figure 4 Teaching points: The effects of the fasting strategy on cardiac pathophysiology are likely to be regulated differently based upon regulation of the LYNUS complex. Disorders predisposed to by high calorie diet, diabetes, aging are characterized by sustained mTOR activation which is postulated to inhibit the autophagy-lysosome machinery. Loss of circadian cycling as well lysosomal disorders are also expected to manifest similarly. Intermittent fasting, exercise, intermittent rapamycin, and time-restricted feeding based upon circadian cues is likely to induced mTOR cycling permitting rhythmic activation of the autophagy-lysosome pathway to mimic its endogenous regulation by the circadian machinery, thereby transducing benefits in cardiometabolic disease. In contrast, sustained activation of the autophagy-lysosome pathway is likely to have negative consequences on organismal health. These paradigms need to be rigorously tested in future studies.

Figure 5 Teaching point: Studies suggest that the circadian clock (which cycles with the day-night cycle) may regulate activation of the autophagy-lysosome pathway. We speculate that this regulation is likely to be transduced by activity of the mTOR pathway and its effects on the regulation of the autophagy-lysosome machinery by the TFEB/TFE3 family of transcription factors. This paradigm needs to be rigorously evaluated in experimental models.

Figure 6 Teaching points: Lysosomes regulate various cellular processes that have a beneficial or detrimental role in cellular survival and homeostasis. The autophagy pathway is by and large cytoprotective and sustains survival under stress and injury. In specific instances, excessive or dysregulated autophagy has been implicated in driving cell death. Lysosomes, also regulate nutrient sensing, are postulated to regulate the circadian rhythms, and control proteastasis with clear beneficial effects to the organism. Lysosomal degradation of invading organisms (xenophagy) and phagocytosed dead cells (by efferocytosis) regulates the innate immune response. Lysosome function is likely to be key regulator of gene expression in these states. Lysosomes control endocytosis and exocytosis which are essential processes for maintaining cellular homeostasis. It is speculated that lysosomes participate in sarcomere remodeling in cardiac myocytes, experimental proof for which is forthcoming. Lysosomal membrane permeabilization can trigger cell death if the damaged lysosomes are not removed by specialized autophagy (termed lysophagy).

 

 

 

 


Related Articles:

Intracellular Digestion: Lysosomes and Cellular Injury
Circadian Rhythms
Cardiac Metabolism in Perspective
Mitophagy as a Protective Mechanism against Myocardial Stress

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Kartik Mani, Ali Javaheri, Abhinav Diwan. Lysosomes Mediate Benefits of Intermittent Fasting in Cardiometabolic Disease: The Janitor Is the Undercover Boss. Compr Physiol 2018, 8: 1639-1667. doi: 10.1002/cphy.c180005