scholarly journals Mitochondrial Dynamics in Stem Cells and Differentiation

2018 ◽  
Vol 19 (12) ◽  
pp. 3893 ◽  
Author(s):  
Bong Seo ◽  
Sang Yoon ◽  
Jeong Do

Mitochondria are highly dynamic organelles that continuously change their shape. Their main function is adenosine triphosphate (ATP) production; however, they are additionally involved in a variety of cellular phenomena, such as apoptosis, cell cycle, proliferation, differentiation, reprogramming, and aging. The change in mitochondrial morphology is closely related to the functionality of mitochondria. Normal mitochondrial dynamics are critical for cellular function, embryonic development, and tissue formation. Thus, defects in proteins involved in mitochondrial dynamics that control mitochondrial fusion and fission can affect cellular differentiation, proliferation, cellular reprogramming, and aging. Here, we review the processes and proteins involved in mitochondrial dynamics and their various associated cellular phenomena.

Author(s):  
Bong Jong Seo ◽  
Sang Hoon Yoon ◽  
Jeong Tae Do

Mitochondria are highly dynamic organelles that continuously change their shape. Their main function is ATP production; however, they are additionally involved in a variety of cellular phenomena, such as apoptosis, cell cycle, proliferation, differentiation, reprogramming, and aging. The change in mitochondrial morphology is closely related to the functionality of mitochondria. Normal mitochondrial dynamics are critical for cellular function, embryonic development, and tissue formation. Thus, defect in proteins involved in mitochondrial dynamics that control mitochondrial fusion and fission can affect cellular differentiation, proliferation, cellular reprogramming, and aging. Here we review the processes and proteins involved in mitochondrial dynamics and its various associated cellular phenomena.


2019 ◽  
Vol 2019 ◽  
pp. 1-12 ◽  
Author(s):  
Jarmon G. Lees ◽  
Anne M. Kong ◽  
Yi C. Chen ◽  
Priyadharshini Sivakumaran ◽  
Damián Hernández ◽  
...  

Human induced pluripotent stem cells (iPSCs) can be differentiated in vitro into bona fide cardiomyocytes for disease modelling and personalized medicine. Mitochondrial morphology and metabolism change dramatically as iPSCs differentiate into mesodermal cardiac lineages. Inhibiting mitochondrial fission has been shown to promote cardiac differentiation of iPSCs. However, the effect of hydrazone M1, a small molecule that promotes mitochondrial fusion, on cardiac mesodermal commitment of human iPSCs is unknown. Here, we demonstrate that treatment with M1 promoted mitochondrial fusion in human iPSCs. Treatment of iPSCs with M1 during embryoid body formation significantly increased the percentage of beating embryoid bodies and expression of cardiac-specific genes. The pro-fusion and pro-cardiogenic effects of M1 were not associated with changes in expression of the α and β subunits of adenosine triphosphate (ATP) synthase. Our findings demonstrate for the first time that hydrazone M1 is capable of promoting cardiac differentiation of human iPSCs, highlighting the important role of mitochondrial dynamics in cardiac mesoderm lineage specification and cardiac development. M1 and other mitochondrial fusion promoters emerge as promising molecular targets to generate lineages of the heart from human iPSCs for patient-specific regenerative medicine.


PLoS ONE ◽  
2020 ◽  
Vol 15 (12) ◽  
pp. e0231064
Author(s):  
Yoshitsugu Oikawa ◽  
Rumiko Izumi ◽  
Masashi Koide ◽  
Yoshihiro Hagiwara ◽  
Makoto Kanzaki ◽  
...  

Sporadic inclusion body myositis (sIBM) is the most common idiopathic inflammatory myopathy, and several reports have suggested that mitochondrial abnormalities are involved in its etiology. We recruited 9 sIBM patients and found significant histological changes and an elevation of growth differential factor 15 (GDF15), a marker of mitochondrial disease, strongly suggesting the involvement of mitochondrial dysfunction. Bioenergetic analysis of sIBM patient myoblasts revealed impaired mitochondrial function. Decreased ATP production, reduced mitochondrial size and reduced mitochondrial dynamics were also observed in sIBM myoblasts. Cell vulnerability to oxidative stress also suggested the existence of mitochondrial dysfunction. Mitochonic acid-5 (MA-5) increased the cellular ATP level, reduced mitochondrial ROS, and provided protection against sIBM myoblast death. MA-5 also improved the survival of sIBM skin fibroblasts as well as mitochondrial morphology and dynamics in these cells. The reduction in the gene expression levels of Opa1 and Drp1 was also reversed by MA-5, suggesting the modification of the fusion/fission process. These data suggest that MA-5 may provide an alternative therapeutic strategy for treating not only mitochondrial diseases but also sIBM.


2012 ◽  
Vol 11 (3) ◽  
pp. 353-367 ◽  
Author(s):  
John P. Gerstenberger ◽  
Patricia Occhipinti ◽  
Amy S. Gladfelter

ABSTRACT In the multinucleate filamentous fungus Ashbya gossypii , nuclei divide asynchronously in a common cytoplasm. We hypothesize that the division cycle machinery has a limited zone of influence in the cytoplasm to promote nuclear autonomy. Mitochondria in cultured mammalian cells undergo cell cycle-specific changes in morphology and membrane potential and therefore can serve as a reporter of the cell cycle state of the cytoplasm. To evaluate if the cell cycle state of nuclei in A. gossypii can influence the adjacent cytoplasm, we tested whether local mitochondrial morphology and membrane potential in A. gossypii are associated with the division state of a nearby nucleus. We found that mitochondria exhibit substantial heterogeneity in both morphology and membrane potential within a single multinucleated cell. Notably, differences in mitochondrial morphology or potential are not associated with a specific nuclear division state. Heterokaryon mutants with a mixture of nuclei with deletions of and wild type for the mitochondrial fusion/fission genes DNM1 and FZO1 exhibit altered mitochondrial morphology and severe growth and sporulation defects. This dominant effect suggests that the gene products may be required locally near their expression site rather than diffusing widely in the cell. Our results demonstrate that mitochondrial dynamics are essential in these large syncytial cells, yet morphology and membrane potential are independent of nuclear cycle state.


2018 ◽  
Vol 218 (2) ◽  
pp. 559-579 ◽  
Author(s):  
Eric M. Sawyer ◽  
Pallavi R. Joshi ◽  
Victoria Jorgensen ◽  
Julius Yunus ◽  
Luke E. Berchowitz ◽  
...  

Cellular differentiation involves remodeling cellular architecture to transform one cell type to another. By investigating mitochondrial dynamics during meiotic differentiation in budding yeast, we sought to understand how organelle morphogenesis is developmentally controlled in a system where regulators of differentiation and organelle architecture are known, but the interface between them remains unexplored. We analyzed the regulation of mitochondrial detachment from the cell cortex, a known meiotic alteration to mitochondrial morphology. We found that mitochondrial detachment is enabled by the programmed destruction of the mitochondria–endoplasmic reticulum–cortex anchor (MECA), an organelle tether that bridges mitochondria and the plasma membrane. MECA regulation is governed by a meiotic transcription factor, Ndt80, which promotes the activation of a conserved kinase, Ime2. We further present evidence for Ime2-dependent phosphorylation and degradation of MECA in a temporally controlled manner. Our study defines a key mechanism that coordinates mitochondrial morphogenesis with the landmark events of meiosis and demonstrates that cells can developmentally regulate tethering to induce organelle remodeling.


2020 ◽  
Vol 27 (12) ◽  
pp. 3273-3288
Author(s):  
Hye In Cho ◽  
Min Seong Kim ◽  
Jina Lee ◽  
Byong Chul Yoo ◽  
Kyung Hee Kim ◽  
...  

AbstractBrpf-histone acetyltransferase (HAT) complexes have important roles in embryonic development and regulating differentiation in ESCs. Among Brpf family, Brpf3 is a scaffold protein of Myst2 histone acetyltransferase complex that plays crucial roles in gene regulation, DNA replication, development as well as maintaining pluripotency in embryonic stem cells (ESCs). However, its biological functions in ESCs are not elucidated. In this study, we find out that Brpf3 protein level is critical for Myst2 stability and E3 ligase Huwe1 functions as a novel negative regulator of Myst2 via ubiquitin-mediated degradation. Importantly, Brpf3 plays an antagonistic role in Huwe1-mediated degradation of Myst2, suggesting that protein–protein interaction between Brpf3 and Myst2 is required for retaining Myst2 stability. Further, Brpf3 overexpression causes the aberrant upregulation of Myst2 protein levels which in turn induces the dysregulated cell-cycle progression and also delay of early embryonic development processes such as embryoid-body formation and lineage commitment of mouse ESCs. The Brpf3 overexpression-induced phenotypes can be reverted by Huwe1 overexpression. Together, these results may provide novel insights into understanding the functions of Brpf3 in proper differentiation as well as cell-cycle progression of ESCs via regulation of Myst2 stability by obstructing Huwe1-mediated ubiquitination. In addition, we suggest that this is a useful report which sheds light on the function of an unknown gene in ESC field.


2017 ◽  
Vol 2017 ◽  
pp. 1-11 ◽  
Author(s):  
Javier Prieto ◽  
Josema Torres

Somatic cells can be reprogrammed into a pluripotent cellular state similar to that of embryonic stem cells. Given the significant physiological differences between the somatic and pluripotent cells, cell reprogramming is associated with a profound reorganization of the somatic phenotype at all levels. The remodeling of mitochondrial morphology is one of these dramatic changes that somatic cells have to undertake during cell reprogramming. Somatic cells transform their tubular and interconnected mitochondrial network to the fragmented and isolated organelles found in pluripotent stem cells early during cell reprogramming. Accordingly, mitochondrial fission, the process whereby the mitochondria divide, plays an important role in the cell reprogramming process. Here, we present an overview of the importance of mitochondrial fission in both cell reprogramming and cellular transformation.


2020 ◽  
Author(s):  
Yoshitsugu Oikawa ◽  
Rumiko Izumi ◽  
Masashi Koide ◽  
Yoshihiro Hagiwara ◽  
Makoto Kanzaki ◽  
...  

AbstractSporadic inclusion body myositis (sIBM) is the most common idiopathic inflammatory myopathy, and several reports have suggested that mitochondrial abnormalities are involved in its etiology.We recruited 9 sIBM patients and found significant histological changes and an elevation of growth differential factor 15 (GDF15), a marker of mitochondrial disease, strongly suggesting the involvement of mitochondrial dysfunction. Bioenergetic analysis of sIBM patient myoblasts revealed impaired mitochondrial function.Decreased ATP production, reduced mitochondrial size and reduced mitochondrial dynamics were also observed in sIBM myoblasts. Cell vulnerability to oxidative stress also suggested the existence of mitochondrial dysfunction.Mitochonic acid-5 (MA-5) increased the cellular ATP level, reduced mitochondrial ROS, and provided protection against sIBM myoblast death.MA-5 also improved the survival of sIBM skin fibroblasts as well as mitochondrial morphology and dynamics in these cells. The reduction in the gene expression levels of Opa1 and Drp1 was also reversed by MA-5, suggesting the modification of the fusion/fission process. These data suggest that MA-5 may provide an alternative therapeutic strategy for treating not only mitochondrial diseases but also sIBM.


2013 ◽  
Vol 305 (12) ◽  
pp. H1736-H1751 ◽  
Author(s):  
Jin O-Uchi ◽  
Bong Sook Jhun ◽  
Stephen Hurst ◽  
Sara Bisetto ◽  
Polina Gross ◽  
...  

Ca+ influx to mitochondria is an important trigger for both mitochondrial dynamics and ATP generation in various cell types, including cardiac cells. Mitochondrial Ca2+ influx is mainly mediated by the mitochondrial Ca2+ uniporter (MCU). Growing evidence also indicates that mitochondrial Ca2+ influx mechanisms are regulated not solely by MCU but also by multiple channels/transporters. We have previously reported that skeletal muscle-type ryanodine receptor (RyR) type 1 (RyR1), which expressed at the mitochondrial inner membrane, serves as an additional Ca2+ uptake pathway in cardiomyocytes. However, it is still unclear which mitochondrial Ca2+ influx mechanism is the dominant regulator of mitochondrial morphology/dynamics and energetics in cardiomyocytes. To investigate the role of mitochondrial RyR1 in the regulation of mitochondrial morphology/function in cardiac cells, RyR1 was transiently or stably overexpressed in cardiac H9c2 myoblasts. We found that overexpressed RyR1 was partially localized in mitochondria as observed using both immunoblots of mitochondrial fractionation and confocal microscopy, whereas RyR2, the main RyR isoform in the cardiac sarcoplasmic reticulum, did not show any expression at mitochondria. Interestingly, overexpression of RyR1 but not MCU or RyR2 resulted in mitochondrial fragmentation. These fragmented mitochondria showed bigger and sustained mitochondrial Ca2+ transients compared with basal tubular mitochondria. In addition, RyR1-overexpressing cells had a higher mitochondrial ATP concentration under basal conditions and showed more ATP production in response to cytosolic Ca2+ elevation compared with nontransfected cells as observed by a matrix-targeted ATP biosensor. These results indicate that RyR1 possesses a mitochondrial targeting/retention signal and modulates mitochondrial morphology and Ca2+-induced ATP production in cardiac H9c2 myoblasts.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1703-1703 ◽  
Author(s):  
Shanshan Pei ◽  
Mohammad Minhajuddin ◽  
Biniam Adane ◽  
Brett M Stevens ◽  
Nabilah Khan ◽  
...  

Abstract Mitochondrial dynamics describes the ability of mitochondria to switch between fission and fusion-active states to change morphology. Although it has long been thought that mitochondrial dynamics is downstream of various growth/differentiation signals and metabolic cues, recent studies have shown that mitochondrial morphology can also actively regulate cellular metabolism and the cell fate of normal stem cells including hematopoietic stem cells. In the present studies, we examined the role of mitochondrial dynamics in the biology of human leukemia stem cells (LSCs). We observe that LSCs are highly sensitive to the perturbation of mitochondrial morphology and that defined signal transduction pathways are involved in this process. We therefore propose that maintenance of the LSC state is critically linked to mitochondrial dynamics. Our data show that LSC enriched populations derived from primary human acute myeloid leukemia (AML) specimens have hyper-active mitochondrial fission regulators as evidenced by higher levels of mitochondrial fission 1 (FIS1) and activating phosphorylation of dynamin-related protein 1 (DRP1). LSCs also contain an increased number of smaller and globular-shaped mitochondria, indicating that they reside in a fission-active state relative to non-LSCs. Inhibition of mitochondrial fission through genetic knock-down of FIS1 (FIS1-KD) induces mitochondrial fusion and dramatically diminishes both colony-forming ability and serial engraftment potential of primary AML cells, suggesting a fission-active state of mitochondrial morphology is critical for LSC function. To dissect the mechanism by which inhibition of mitochondrial fission impairs the stem and progenitor potential of LSCs, we performed detailed analyses of molecular events following FIS1-KD. We determined that FIS1-KD in AML cells simultaneously induces activating phosphorylation of AMPK and inhibitory phosphorylation of GSK3. Consistent with AMPK activation, FIS1-depleted AML cells had elevated oxidative phosphorylation (OXPHOS) activity, increased cellular ATP, and dramatic changes of various metabolomic intermediates, indicating a clear shift to active global energy metabolism. With regard to GSK3 inhibition, we found that the FIS1-KD-induced gene expression signature strongly phenocopied the gene signature obtained using a pan-GSK3 inhibitor or shRNA-mediated knock down of GSK3. Further, FIS1-KD in AML cells also strongly induced hematopoietic differentiation as evidenced by increased surface expression of hematopoietic lineage markers, global upregulation of hematopoietic differentiation genes, and collapse of the HOXA9 transcriptional program. Importantly, both shRNA-mediated AMPK inhibition and overexpression of a constitutively active GSK3 allele can rescue the FIS1-KD-induced hematopoietic differentiation phenotype, suggesting FIS1-KD-induced differentiation is dependent on both AMPK activation and GSK3 inhibition. To investigate if FIS1-KD-induced hematopoietic differentiation depends on an altered state of mitochondrial morphology, we also studied other regulators of mitochondrial dynamics including fission player DRP1 and fusion players mitofusin 2 (MFN2) and Optic Atrophy 1 (OPA1). We showed that shRNA-mediated KD of fusion players MFN2 and OPA1 can also rescue FIS1-KD induced hematopoietic differentiation, suggesting the balance between the activity of fission and fusion regulators is critical in determining the fate of LSCs. In addition, we showed that shRNA-mediated KD of the fission player DRP1 could also lead to AMPK activation and GSK3 inhibition, in strong agreement with FIS1-KD. Finally, we showed that combined treatment of AMPK activator and GSK3 inhibitors can severely kill AML cells, suggesting a potential therapeutic strategy. Taken together, we propose that mitochondrial dynamics plays a previously unrealized yet important role in sustaining LSCs of AML. Further, inhibition of mitochondrial fission players can activate AMPK signaling, force active energy metabolism, collapse GSK3-mediated transcription programs, and induce hematopoietic differentiation, all of which lead to loss of stem and progenitor potential of AML cells. Thus the current study reveals a novel dependence of LSCs on mitochondrial dynamics, and provides novel insights towards improved therapeutic regimens. Disclosures Pollyea: Celgene: Other: advisory board, Research Funding; Ariad: Other: advisory board; Glycomimetics: Other: DSMB member; Pfizer: Other: advisory board, Research Funding; Alexion: Other: advisory board.


Sign in / Sign up

Export Citation Format

Share Document