Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2431 2023-07-06 19:28:13 |
2 format Meta information modification 2431 2023-07-07 04:10:05 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Kolahdouzmohammadi, M.; Kolahdouz-Mohammadi, R.; Tabatabaei, S.A.; Franco, B.; Totonchi, M. Autophagy in Cardiac Differentiation. Encyclopedia. Available online: https://encyclopedia.pub/entry/46538 (accessed on 01 July 2024).
Kolahdouzmohammadi M, Kolahdouz-Mohammadi R, Tabatabaei SA, Franco B, Totonchi M. Autophagy in Cardiac Differentiation. Encyclopedia. Available at: https://encyclopedia.pub/entry/46538. Accessed July 01, 2024.
Kolahdouzmohammadi, Mina, Roya Kolahdouz-Mohammadi, Seyed Abdolhossein Tabatabaei, Brunella Franco, Mehdi Totonchi. "Autophagy in Cardiac Differentiation" Encyclopedia, https://encyclopedia.pub/entry/46538 (accessed July 01, 2024).
Kolahdouzmohammadi, M., Kolahdouz-Mohammadi, R., Tabatabaei, S.A., Franco, B., & Totonchi, M. (2023, July 06). Autophagy in Cardiac Differentiation. In Encyclopedia. https://encyclopedia.pub/entry/46538
Kolahdouzmohammadi, Mina, et al. "Autophagy in Cardiac Differentiation." Encyclopedia. Web. 06 July, 2023.
Autophagy in Cardiac Differentiation
Edit

Autophagy is a critical biological process in which cytoplasmic components are sequestered in autophagosomes and degraded in lysosomes. This highly conserved pathway controls intracellular recycling and is required for cellular homeostasis, as well as the correct functioning of a variety of cellular differentiation programs, including cardiomyocyte differentiation. By decreasing oxidative stress and promoting energy balance, autophagy is triggered during differentiation to carry out essential cellular remodeling, such as protein turnover and lysosomal degradation of organelles. When it comes to controlling cardiac differentiation, the crosstalk between autophagy and other signaling networks such as fibroblast growth factor (FGF), Wnt, Notch, and bone morphogenetic proteins (BMPs) is essential, yet the interaction between autophagy and epigenetic controls remains poorly understood. Numerous studies have shown that modulating autophagy and precisely regulating it can improve cardiac differentiation, which can serve as a viable strategy for generating mature cardiac cells. 

autophagy cardiomyocyte differentiation

1. Crosstalk between Autophagy, Cardiac Differentiation, and Other Signaling Pathways

The crosstalk between autophagy and other signaling pathways plays a crucial role in the modulation of cardiac differentiation. Zhang et al. were the first to recognize the connection between autophagy and cardiac stem-cell differentiation, finding that fibroblast growth factor (FGF) regulates cardiac development through autophagy [1]. Their study revealed that the inhibition of FGF signaling led to an upsurge in the initiation of autophagy, thereby promoting the differentiation of cardiomyocytes. These findings strongly suggest that autophagy serves as a positive regulator of both cardiac progenitor cell differentiation and cardiomyocyte maturation.
FGF signaling exhibits a biphasic function in cardiogenesis. During the early stages, FGF signaling promotes mesoderm formation and commitment to the cardiac lineage. However, in later stages, FGF signaling inhibits premature differentiation to ensure proper cardiac development and maturation. The involvement of autophagy in mediating FGF signaling during the differentiation of heart progenitor cells highlights the significance of autophagy in cardiac development. The interplay between autophagy and FGF signaling provides novel insights into the regulatory mechanisms underlying cardiac differentiation.
Additionally, other signaling pathways, such as Wnt, Notch, and bone morphogenetic proteins (BMPs), are also involved in regulating heart development [2]. Wnt and Notch signaling pathways are important in heart development, and there is evidence of a crosstalk between the two [3]. The main components of the Wnt signaling pathway include dishevelled (Dvl), AXIN, glycogen synthase kinase-3 (GSK3)-β, and β-catenin. It has been shown that Wnt signaling plays a biphasic function in cardiogenesis. This pathway stimulates cardiomyocyte differentiation in the early stages of heart development but suppresses cardiogenesis later on [4]. Autophagy can be negatively regulated by Wnt signaling by promoting Dvl degradation through the autophagy–lysosome pathway, which, in turn, induces a concomitant decrease in the nuclear accumulation of β-catenin [5]. Furthermore, it has been demonstrated that autophagy induction resulted in a considerable drop in the gene expression of Axin2, Cyclin-D1, and c-Myc, as well as a decrease in the protein expression of β-catenin and Dvl2. Based on these findings, it was determined that autophagy has a negative regulatory effect on the Wnt signaling pathway [6].
Notch signaling, a pivotal signaling pathway in cardiac differentiation, demonstrates intricate dynamics and complexity throughout the process of cardiac development. Initially, it is believed that Notch signaling suppresses cardiac differentiation to maintain a pool of undifferentiated progenitor cells. However, different studies show that Notch1 is required for cardiac progenitor cells to differentiate into cardiomyocytes by positively regulating the expression of cardiac transcription factors in mouse embryos and embryonic stem (ES) cells [7][8][9]. In the context of cardiac development, autophagy has been investigated in relation to the Wnt and Notch signaling pathways [10][11]. Zhuqing Jia et al. studied the involvement of autophagy in the cardiac development of P19CL6 cells, a well-established in vitro cardiomyocyte differentiation system [12]. Their findings demonstrated that autophagy is initiated during the early stages of cardiomyocyte differentiation and remains active throughout the final stages. The researchers also observed that β-catenin and Notch intracellular domain (NICD), which are effectors of the Wnt and Notch pathways, can form a complex with LC3 and P62, leading to their elimination through autophagy. Interestingly, β-catenin not only inhibits the formation of autophagosomes but also directly reduces the expression of the autophagy adaptor p62 [11].
Moreover, epigenetic factors, such as histone deacetylases (HDACs), have also been shown to play a role in regulating autophagy during cardiac differentiation [13][14][15][16][17]. Specifically, studies have demonstrated that HDACs control the differentiation of P19 embryonic cancer cells into cardiac lineage cells [18]. Despite the advances in understanding the roles of autophagy and epigenetic regulation in cardiac differentiation, the exact mechanisms underlying their interaction in this process remain incompletely understood. Further investigations are required to elucidate the intricate relationship between autophagy, Wnt signaling, and cardiac differentiation, which will provide valuable insights into the development and potential therapeutic strategies for cardiac-related conditions.
The mammalian target of rapamycin (mTOR) is a crucial serine/threonine kinase involved in cellular survival and growth. It operates through two distinct complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), which are predominantly made up of Raptor and Rictor as their core components, respectively. Reports from different laboratories have demonstrated that mTOR complex 1 acts as a master regulator of autophagic processes since inhibition of mTORC1 was shown to be required to start autophagy. Subsequent studies have also demonstrated that mTORC1 directly regulates the subsequent phases, such as autophagosomes formation, maturation, and autophagy termination [19]. Raptor/mTORC1 and Rictor/mTORC2 play distinct and significant roles in the differentiation of cardiomyocytes derived from mouse embryonic stem cells (mESCs). Numerous molecular elements and phosphorylation processes mediate the intricate interaction between these two signaling pathways [20].
AKT, also known as protein kinase B, is a key protein and acts as a bridge between the two complexes. AKT can be phosphorylated by mTORC2 or 3-phosphoinositide-dependent kinase 1 (PDK1), thereby regulating downstream signaling [21]. Specifically, Raptor knockdown reduced the phosphorylation of Rictor at Thr1135 by p70S6K, leading to mTORC2 activation [20].
Raptor/mTORC1 primarily regulates metabolism, cell growth, and protein synthesis in response to nutrient availability, and growth factors [22]. It controls the phosphorylation of S6 kinase 1 (S6K1) and eukaryotic translation initiation factor 4E-binding protein (eIF4E), thereby promoting translation and protein synthesis [23]. In the context of cardiomyocyte differentiation, Raptor knockdown enhances the process by activating Rictor/mTORC2 signaling, which, in turn, facilitates the differentiation of mESCs into cardiomyocytes. This is accompanied by elevated expression levels of brachyury (a mesoderm protein), Nkx2.5 (a cardiac progenitor cell protein), and α-actinin (a cardiomyocyte marker) in Raptor knockdown cells [20].
On the other hand, Rictor/mTORC2 exerts its impact on cytoskeletal organization, cell survival, and cell polarization. It has been implicated in the regulation of cellular processes such as cell adhesion, migration, and differentiation [24]. Rictor knockout in cardiac-specific cells leads to cardiac dysfunction and impaired adaptation to pressure overload. In the context of mESCs differentiation into cardiomyocytes, Rictor knockdown suppresses cardiogenesis, indicating the critical role of Rictor/mTORC2 in this process [20].
Upstream of mTOR, various signals, including insulin signaling through phosphatidylinositol-3-kinase (PI3K) and AKT, and energy/stress response signaling via AMP-activated protein kinase (AMPK), converge to regulate the activity of the tuberous sclerosis complexes (TSC1-TSC2), which act as GTPase-activating proteins for Rheb. When Rheb is in its GTP-bound state, it directly activates mTOR. The convergence of these signals ensures precise control over mTOR activation and downstream cellular processes [23].
The regulation of cardiomyocyte differentiation can be better understood by gaining valuable insights from the complex relationship between these signaling pathways, which may also have implications for potential therapeutic applications using induced pluripotent stem cells.
Aside from the main autophagy pathway, other types of autophagy, such as mitophagy, play a crucial role in cardiac differentiation. Mitophagy, a form of autophagy, is a vital process in cardiac differentiation. It helps in the removal of dysfunctional mitochondria and facilitates metabolic remodeling, which is crucial for successful differentiation. Regulated mitophagy plays a critical role in maintaining the cell’s glycolytic status and is believed to contribute to the metabolic remodeling necessary for successful differentiation [25][26][27][28].
A specific process of mitophagy is initiated by the PINK1/Parkin system, where Parkin, an E3 ubiquitin ligase, is recruited to dysfunctional mitochondria. Parkin ubiquitinates outer mitochondrial proteins, labeling them for proteasome degradation [29]. In mice, the deletion of Parkin during early postnatal stages impedes cardiomyocyte maturation, affects mitochondrial biogenesis, and disrupts fatty acid oxidation, highlighting the importance of PINK1/Parkin-mediated mitophagy in cardiac development [25]. These findings highlight the important role of PINK1/Parkin-mediated mitophagy in promoting proper cardiac development.
The regulation of mitophagy in cardiac differentiation is influenced by nutrient-sensing pathways, such as Akt/mTOR, which is upregulated in high-nutrient states, and AMPK, which is upregulated during fasting. Inhibiting mTOR signaling increases mitophagy, providing cardioprotection after myocardial infarction. Conversely, AMPKα2 phosphorylates PINK1, enhancing mitophagy and protecting against pressure overload in the heart. AMPK also influences the opening of the mitochondrial permeability transition pore (mPTP), which regulates mitochondrial depolarization and subsequent mitophagy [30][31]. Increasing Parkin-mediated mitophagy through enhanced fatty acid oxidation, achieved by deleting acetyl coenzyme A carboxylase 2 (ACC2), prevents myocardial dysfunction resulting from a high-fat diet [32]. These interconnected mechanisms underscore the significance of metabolic shifts, mitochondrial quality control, and mitophagy in maintaining proper cardiac function.

2. How Does Manipulation of Autophagy Improve Cardiac Differentiation?

Recent advances in stem-cell research have utilized embryoid body suspension and monolayer attachment methods to increase differentiation efficiency, but further improvements are required to optimize this process [33][34][35]. Adding growth factors such as BMP2, BMP4, FGFs, activin A, b-fibroblast growth factor (bFGF), FGF2, FGF10, and Wnt family member 3A (Wnt3a) during cardiac differentiation can improve induction efficiency, but the high cost of these reagents limits their practicality [6][36][37]. Thus, the use of small chemical cocktails that target the autophagy pathway is a promising alternative. However, in order to boost differentiation efficacy even further, the induction strategy that improves differentiation efficiency, as well as the detailed mechanisms, must be defined.
Methods that have been developed to manipulate the autophagy pathway to promote cardiac differentiation are discussed below. Rapamycin is an inhibitor of mTOR that has been found to promote cardiomyocyte differentiation in a stage-dependent manner. It works similarly to Ku0063794, another mTOR kinase inhibitor [6]. While rapamycin inhibits mTORC1 activity, its effect on mTORC2 is limited [38]. Nevertheless, rapamycin has been found to induce autophagy, upregulate Fgf8 and Nodal expression, alleviate stressors during cardiac differentiation, reduce hPSC apoptosis, promote BMP signaling, and suppress the activation of Wnt/β-catenin and Notch signaling during cardiac induction [33][39][40][41][42]. The efficiency of cardiac differentiation can be affected by different rapamycin concentrations and the presence of CHIR99021, a small molecule inhibitor that targets the GSK-3β enzyme. In addition to reducing hPSC apoptosis and boosting hESC survival, rapamycin has been shown to have other benefits in cardiac differentiation. It has been found to reduce medium usage by around 50% and overcome variability in differentiation efficiency among different hESC lines and repetitions, resulting in a greater efficiency, ranging from 87% to 99.06% [33].
Moreover, combining rapamycin and ascorbic acid has also been suggested as an effective regimen for cardiac induction. This combination has been found to increase collagen IV synthesis as well as a number of molecular signals triggered by collagen, such as the extracellular signal-regulated kinase (ERK), c- JNKs, and STAT1/3 pathways, which promote cardiac differentiation through various molecular signaling pathways [43]. These findings provide insight into the diverse and complex mechanisms underlying the regulation of cellular differentiation and suggest the potential therapeutic applications of rapamycin in promoting cardiac regeneration.
AMPK is an energy-sensing protein kinase involved in the regulation of autophagy. In addition to its direct role in regulating autophagy, AMPK can also influence other cellular processes, such as mitochondrial function, posttranslational acetylation, cardiomyocyte metabolism, mitochondrial autophagy, endoplasmic reticulum stress, and apoptosis. As AMPK is involved in the control of various cellular processes, it can influence the health and survival of cardiomyocytes [44][45]. AMPK activation increases fatty acid oxidation in human induced pluripotent stem cell (hiPSC)-cardiomyocytes (CMs) by upregulating the expression of carnitine palmitoyl transferase (CPT)-1, fatty acid transport protein (FATP), and fatty acid-binding protein (FABP) [44][46]. Moreover, AMPK has a distinct regulatory impact on numerous areas of mitochondrial biology and homeostasis, such as controlling the number of mitochondria via stimulating mitochondrial synthesis, regulating the architecture of the mitochondrial network, and controlling the quality of mitochondria via autophagy and mitosis [47].
Understanding the significance of AMPK in cardiomyocyte differentiation, Ye et al. employed 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) to induce AMPK activation in hiPSC-CMs [48]. AICAR dramatically raised the oxygen consumption rate (OCR) related to baseline respiration, adenosine 5′-triphosphate (ATP) generation, maximum respiration, and reserve capacity, indicating that cardiomyocytes were in a more mature metabolic state. Furthermore, AMPK activation enhances mitochondrial fusion in hiPSC-CMs [48]. In times of cellular stress, mitochondrial respiratory reserve capacity is employed to sustain an increase in energy demand, assisting in the maintenance of cell and organ functions, cell repair, or active drug detoxification [49]. AICAR treatment boosted respiratory reserve capacity in hiPSC-CMs, implying that the treated cells may perform better in the face of increasing energy demand. In addition, AICAR-treated hiPSC-CMs exhibited a larger cell perimeter, a lower circularity index, and longer sarcomeres than the control group, making them more similar to adult cardiomyocytes [48]. AICAR also increased the expression of cardiac maturation-related genes. Although the AICAR activation of AMPK promotes the morphological and metabolic maturity of hiPSC-CMs, the effect of AMPK on the electrophysiological maturation of hiPSC-CMs remains unknown [48].

3. Other Forms of Autophagy and Cardiac Differentiation

Mitophagy, the selective removal of damaged or depolarized mitochondria, plays a crucial role in maintaining mitochondrial health as a quality control mechanism [50]. During stem cell differentiation, changes in metabolism occur to increase oxidative phosphorylation and mitochondrial ATP production [51]. This metabolic shift has been shown to drive cell differentiation, implying a positive feedback loop between differentiation and metabolic rewiring [52].
The involvement of mitochondrial biogenesis and the β-catenin pathway in the differentiation of endothelial progenitor cells is supported by scientific evidence. An increase in mitochondrial biogenesis, associated with the upregulation of PGC-1α, has been observed during differentiation towards the endothelial progenitor cell stage [53][54]. PGAM5, a mitochondrial phosphatase released during mitophagy, activates the β-catenin pathway by dephosphorylating β-catenin, allowing it to translocate to the nucleus and transcribe Wnt/β-catenin pathway genes and promoting mitophagy and mitochondrial biogenesis [55][56]. This interplay between PGAM5 and β-catenin is crucial for the differentiation of hiPSCs into endothelial cells [53].
While the downstream impact of mitophagy on iPSC differentiation has been studied, the upstream initiators of mitophagy during cell differentiation remain unknown. Krantz et al. investigated the role of mitophagy in iPSC differentiation and the initiation of compensatory mitochondrial biogenesis via the PGAM5 pathway but did not identify the upstream initiators of mitophagy [53]. In the review article authored by Garbern in 2021, a comprehensive examination of the role of mitophagy in cardiomyocyte differentiation is presented [30].

References

  1. Zhang, J.; Liu, J.; Liu, L.; McKeehan, W.L.; Wang, F. The fibroblast growth factor signaling axis controls cardiac stem cell differentiation through regulating autophagy. Autophagy 2012, 8, 690–691.
  2. Foley, A.; Mercola, M. Heart induction: Embryology to cardiomyocyte regeneration. Trends Cardiovasc. Med. 2004, 14, 121–125.
  3. Li, B.; Jia, Z.; Wang, T.; Wang, W.; Zhang, C.; Chen, P.; Ma, K.; Zhou, C. Interaction of Wnt/β-catenin and notch signaling in the early stage of cardiac differentiation of P19CL6 cells. J. Cell. Biochem. 2012, 113, 629–639.
  4. Ueno, S.; Weidinger, G.; Osugi, T.; Kohn, A.D.; Golob, J.L.; Pabon, L.; Reinecke, H.; Moon, R.T.; Murry, C.E. Biphasic role for Wnt/β-catenin signaling in cardiac specification in zebrafish and embryonic stem cells. Proc. Natl. Acad. Sci. USA 2007, 104, 9685–9690.
  5. Cao, N.; Liu, Z.; Chen, Z.; Wang, J.; Chen, T.; Zhao, X.; Ma, Y.; Qin, L.; Kang, J.; Wei, B.; et al. Ascorbic acid enhances the cardiac differentiation of induced pluripotent stem cells through promoting the proliferation of cardiac progenitor cells. Cell Res. 2012, 22, 219–236.
  6. Lu, Q.; Liu, Y.; Wang, Y.; Yang, Z.; Li, T.; Tian, Y.; Chen, P.; Ma, K.; Jia, Z.; Zhou, C. Rapamycin efficiently promotes cardiac differentiation of mouse embryonic stem cells. Biosci. Rep. 2017, 37, BSR20160552.
  7. Kwon, C.; Qian, L.; Cheng, P.; Nigam, V.; Arnold, J.; Srivastava, D. A regulatory pathway involving Notch1/β-catenin/Isl1 determines cardiac progenitor cell fate. Nat. Cell Biol. 2009, 11, 951–957.
  8. Kasahara, A.; Cipolat, S.; Chen, Y.; Dorn, G.W.; Scorrano, L. Mitochondrial fusion directs cardiomyocyte differentiation via calcineurin and Notch signaling. Science 2013, 342, 734–737.
  9. Saravanakumar, M.; Devaraj, H. Notch signalling in cardiovasculogenesis: Insight into their role in early cardiovascular development. Mol. Biol. Rep. 2013, 40, 3537–3547.
  10. Nàger, M.; Sallán, M.C.; Visa, A.; Pushparaj, C.; Santacana, M.; Macià, A.; Yeramian, A.; Canti, C.; Herreros, J. Inhibition of WNT-CTNNB1 signaling upregulates SQSTM1 and sensitizes glioblastoma cells to autophagy blockers. Autophagy 2018, 14, 619–636.
  11. Petherick, K.J.; Williams, A.C.; Lane, J.D.; Ordóñez-Morán, P.; Huelsken, J.; Collard, T.J.; Smartt, H.J.M.; Batson, J.; Malik, K.; Paraskeva, C.; et al. Autolysosomal β-catenin degradation regulates Wnt-autophagy-p62 crosstalk. EMBO J. 2013, 32, 1903–1916.
  12. Jia, Z.; Wang, J.; Wang, W.; Tian, Y.; XiangWei, W.; Chen, P.; Ma, K.; Zhou, C. Autophagy eliminates cytoplasmic β-catenin and NICD to promote the cardiac differentiation of P19CL6 cells. Cell Signal. 2014, 26, 2299–2305.
  13. Lin, S.Y.; Li, T.Y.; Liu, Q.; Zhang, C.; Li, X.; Chen, Y.; Zhang, S.-M.; Lian, G.; Liu, Q.; Ruan, K.; et al. GSK3-TIP60-ULK1 signaling pathway links growth factor deprivation to autophagy. Science 2012, 336, 477–481.
  14. Li, W.; Liu, H.; Liu, P.; Yin, D.; Zhang, S.; Zhao, J. Sphingosylphosphorylcholine promotes the differentiation of resident Sca-1 positive cardiac stem cells to cardiomyocytes through lipid raft/JNK/STAT3 and β-catenin signaling pathways. BBA Mol. Cell Res. 2016, 1863, 1579–1588.
  15. Rojanasakul, Y. Linking JNK-STAT3-Akt signaling axis to EZH2 phosphorylation: A novel pathway of carcinogenesis. Cell Cycle 2013, 12, 202.
  16. Moslehi, M.; Yazdanparast, R. SK-N-MC cell death occurs by distinct molecular mechanisms in response to hydrogen peroxide and superoxide anions: Involvements of JAK2-STAT3, JNK, and p38 MAP kinases pathways. Cell Biochem. Biophys. 2013, 66, 817–829.
  17. Nakamura, T.; Sano, M.; Songyang, Z.; Schneider, M.D. A Wnt-and β-catenin-dependent pathway for mammalian cardiac myogenesis. Proc. Natl. Acad. Sci. USA 2003, 100, 5834–5839.
  18. Karamboulas, C.; Swedani, A.; Ward, C.; Al-Madhoun, A.S.; Wilton, S.; Boisvenue, S.; Ridgeway, A.G.; Skerjanc, I.S. HDAC activity regulates entry of mesoderm cells into the cardiac muscle lineage. J. Cell Sci. 2006, 119, 4305–4314.
  19. Dossou, A.S.; Basu, A. The emerging roles of mTORC1 in macromanaging autophagy. Cancers 2019, 11, 1422.
  20. Zheng, B.; Wang, J.; Tang, L.; Shi, J.; Zhu, D. mTORC1 and mTORC2 play different roles in regulating cardiomyocyte differentiation from embryonic stem cells. Int. J. Dev. Biol. 2017, 61, 65–72.
  21. Xie, J.; Proud, C.G. Crosstalk between mTOR complexes. Nat. Cell Biol. 2013, 15, 1263–1265.
  22. Goodman, C.A. The role of mTORC1 in regulating protein synthesis and skeletal muscle mass in response to various mechanical stimuli. Rev. Physiol. Biochem. Pharmacol. 2014, 166, 43–95.
  23. Burnett, P.E.; Barrow, R.K.; Cohen, N.A.; Snyder, S.H.; Sabatini, D.M. RAFT1 phosphorylation of the translational regulators p70 S6 kinase and 4E-BP1. Proc. Natl. Acad. Sci. USA 1998, 95, 1432–1437.
  24. Sarbassov, D.D.; Ali, S.M.; Kim, D.-H.; Guertin, D.A.; Latek, R.R.; Erdjument-Bromage, H.; Tempst, P.; Sabatini, D.M. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr. Biol. 2004, 14, 1296–1302.
  25. Gong, G.; Song, M.; Csordas, G.; Kelly, D.P.; Matkovich, S.J.; Dorn, G.W. Parkin-mediated mitophagy directs perinatal cardiac metabolic maturation in mice. Science 2015, 350, aad2459.
  26. Lampert, M.A.; Orogo, A.M.; Najor, R.H.; Hammerling, B.C.; Leon, L.J.; Wang, B.J.; Kim, T.; Sussman, M.A.; Gustafsson, Å. B BNIP3L/NIX and FUNDC1-mediated mitophagy is required for mitochondrial network remodeling during cardiac progenitor cell differentiation. Autophagy 2019, 15, 1182–1198.
  27. Rodger, C.E.; McWilliams, T.G.; Ganley, I.G. Mammalian mitophagy–from in vitro molecules to in vivo models. FEBS J. 2018, 285, 1185–1202.
  28. Montava-Garriga, L.; Ganley, I.G. Outstanding questions in mitophagy: What we do and do not know. J. Mol. Biol. 2020, 432, 206–230.
  29. Nguyen, T.N.; Padman, B.S.; Lazarou, M. Deciphering the molecular signals of PINK1/Parkin mitophagy. Trends Cell Biol. 2016, 26, 733–744.
  30. Garbern, J.C.; Lee, R.T. Mitochondria and metabolic transitions in cardiomyocytes: Lessons from development for stem cell-derived cardiomyocytes. Stem Cell Res. Ther. 2021, 12, 177.
  31. Ishihara, T.; Ban-Ishihara, R.; Maeda, M.; Maeda, Y.; Ichimura, A.; Kyogoku, S.; Aoki, H.; Katada, S.; Nakada, K.; Nomura, M.; et al. Dynamics of mitochondrial DNA nucleoids regulated by mitochondrial fission is essential for maintenance of homogeneously active mitochondria during neonatal heart development. Mol. Cell Biol. 2015, 35, 211–223.
  32. Spaan, J.A.; Piek, J.J.; Hoffman, J.I.E.; Siebes, M. Physiological basis of clinically used coronary hemodynamic indices. Circulation 2006, 113, 446–455.
  33. Qiu, X.X.; Liu, Y.; Zhang, Y.F.; Guan, Y.N.; Jia, Q.Q.; Wang, C.; Liang, H.; Li, Y.-Q.; Yang, H.-T.; Qin, Y.-W.; et al. Rapamycin and CHIR 99021 Coordinate Robust Cardiomyocyte Differentiation from Human Pluripotent Stem Cells Via Reducing p53-Dependent Apoptosis. J. Am. Heart Assoc. 2017, 6, e005295.
  34. Shi, X.; Li, W.; Liu, H.; Yin, D.; Zhao, J. β-Cyclodextrin induces the differentiation of resident cardiac stem cells to cardiomyocytes through autophagy. BBA-Mol. Cell Res. 2017, 1864, 1425–1434.
  35. Hatani, T.; Miki, K.; Yoshida, Y. Induction of human induced pluripotent stem cells to cardiomyocytes using embryoid bodies. In Experimental Models of Cardiovascular Diseases; Ishikawa, K., Ed.; Springer: Berlin/Heidelberg, Germany, 2018; Volume 1816, pp. 79–92.
  36. Tran, T.H.; Wang, X.; Browne, C.; Zhang, Y.; Schinke, M.; Izumo, S.; Burcin, M. Wnt3a-induced mesoderm formation and cardiomyogenesis in human embryonic stem cells. Stem Cells 2009, 27, 1869–1878.
  37. Takei, S.; Ichikawa, H.; Johkura, K.; Mogi, A.; No, H.; Yoshie, S.; Tomotsune, D.; Sasaki, K. Bone morphogenetic protein-4 promotes induction of cardiomyocytes from human embryonic stem cells in serum-based embryoid body development. Am. J. Physiol. Circ. Physiol. 2009, 296, H1793–H1803.
  38. Laplante, M.; Sabatini, D.M. mTOR signaling in growth control and disease. Cell 2012, 149, 274–293.
  39. Jiang, M.; Liu, T.; Zhang, J.; Gao, S.; Tao, B.; Cao, R.; Qiu, Y.; Liu, J.; Li, Y.; Wang, Y.; et al. Rapamycin promotes cardiomyocyte differentiation of human induced pluripotent stem cells in a stage-dependent manner. Stem Cells Dev. 2020, 29, 1229–1239.
  40. Liu, Y.; Li, P.; Liu, K.; He, Q.; Han, S.; Sun, X.; Li, T.; Shen, L. Timely inhibition of Notch signaling by DAPT promotes cardiac differentiation of murine pluripotent stem cells. PLoS ONE 2014, 9, e109588.
  41. Zhang, J.; Liu, J.; Huang, Y.; Chang, J.Y.; Liu, L.; McKeehan, W.L.; Martin, J.F.; Wang, F. FRS2α-mediated FGF signals suppress premature differentiation of cardiac stem cells through regulating autophagy activity. Circ. Res. 2012, 110, e29–e39.
  42. Abu-Issa, R.; Smyth, G.; Smoak, I.; Yamamura, K.I.; Meyers, E.N. Fgf8 is required for pharyngeal arch and cardiovascular development in the mouse. Development 2002, 129, 4613–4625.
  43. Leitinger, B. Transmembrane collagen receptors. Annu. Rev. Cell Dev. Biol. 2011, 27, 265–290.
  44. Sarikhani, M.; Garbern, J.C.; Ma, S.; Sereda, R.; Conde, J.; Krähenbühl, G.; Escalante, G.O.; Ahmed, A.; Buenrostro, J.D.; Lee, R.T. Sustained activation of AMPK enhances differentiation of human iPSC-derived cardiomyocytes via sirtuin activation. Stem Cell Rep. 2020, 15, 498–514.
  45. Lin, S.-C.; Hardie, D.G. AMPK: Sensing glucose as well as cellular energy status. Cell Metab. 2018, 27, 299–313.
  46. Yang, X.; Rodriguez, M.L.; Leonard, A.; Sun, L.; Fischer, K.A.; Wang, Y.; Ritterhoff, J.; Zhao, L.; Kolwicz, S.C., Jr.; Pabon, L.; et al. Fatty acids enhance the maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cell Rep. 2019, 13, 657–668.
  47. Herzig, S.; Shaw, R.J. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. 2018, 19, 121–135.
  48. Ye, L.; Zhang, X.; Zhou, Q.; Tan, B.; Xu, H.; Yi, Q.; Yan, L.; Xie, M.; Zhang, Y.; Tian, J.; et al. Activation of AMPK promotes maturation of cardiomyocytes derived from human induced pluripotent stem cells. Front. Cell Dev. Biol. 2021, 9, 644667.
  49. Hill, B.G.; Dranka, B.P.; Zou, L.; Chatham, J.C.; Darley-Usmar, V.M. Importance of the bioenergetic reserve capacity in response to cardiomyocyte stress induced by 4-hydroxynonenal. Biochem. J. 2009, 424, 99–107.
  50. Ng, M.Y.W.; Wai, T.; Simonsen, A. Quality control of the mitochondrion. Dev. Cell 2021, 56, 881–905.
  51. Mathieu, J.; Ruohola-Baker, H. Metabolic remodeling during the loss and acquisition of pluripotency. Development 2017, 144, 541–551.
  52. Chung, S.; Arrell, D.K.; Faustino, R.S.; Terzic, A.; Dzeja, P.P. Glycolytic network restructuring integral to the energetics of embryonic stem cell cardiac differentiation. J. Mol. Cell. Cardiol. 2010, 48, 725–734.
  53. Krantz, S.; Kim, Y.-M.; Srivastava, S.; Leasure, J.W.; Toth, P.T.; Marsboom, G.; Rehman, J. Mitophagy mediates metabolic reprogramming of induced pluripotent stem cells undergoing endothelial differentiation. J. Biol. Chem. 2021, 297, 101410.
  54. Gureev, A.P.; Shaforostova, E.A.; Popov, V.N. Regulation of mitochondrial biogenesis as a way for active longevity: Interaction between the Nrf2 and PGC-1α signaling pathways. Front. Genet. 2019, 10, 435.
  55. Jedrzejas, M.J. Structure, function, and evolution of phosphoglycerate mutases: Comparison with fructose-2, 6-bisphosphatase, acid phosphatase, and alkaline phosphatase. Prog. Biophys. Mol. 2000, 73, 263–287.
  56. Bernkopf, D.B.; Jalal, K.; Brückner, M.; Knaup, K.X.; Gentzel, M.; Schambony, A.; Behrens, J. Pgam5 released from damaged mitochondria induces mitochondrial biogenesis via Wnt signaling. J. Cell Biol. 2018, 217, 1383–1394.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 194
Revisions: 2 times (View History)
Update Date: 07 Jul 2023
1000/1000
Video Production Service