Hypoxia‑induced mitochondrial translocation of DNM1L increases mitochondrial fission and triggers mPTP opening in HCC cells via activation of HK2

Retraction in: /10.3892/or.2021.8059

  • Authors:
    • Min Cai
    • Ping He
    • Dian‑Liang Fang
  • View Affiliations

  • Published online on: June 27, 2019     https://doi.org/10.3892/or.2019.7213
  • Pages: 1125-1132
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Disturbed mitochondrial dynamics are closely associated with the progression of different types of cancer including hepatocellular carcinoma (HCC). However, the manner in which mitochondrial dynamics are regulated in HCC remains largely unclear. In the present study, via immunofluorescence, real‑time PCR and western blot analysis, the effects of dynamin‑1‑like (DNM1L) on mitochondrial translocation and the mitochondrial permeability transition pore (mPTP) were investigated in HCC cells under hypoxic conditions, and the underlying molecular mechanisms were explored. Our data revealed that hypoxic treatment decreased the mitochondrial membrane potential, elevated generation of reactive oxygen species, and promoted mitochondrial fission in a DNM1L‑dependent manner. Instead of changing the levels of DNMlL, hypoxia increased the translocation of DNM1L to mitochondria, leading to excessive mitochondrial fission and decreased the viability of HCC cells. In addition to the effects on mitochondrial dynamics, DNM1L also regulated mPTP opening in HCC. IP analysis revealed that DNM1L interacted with the enzyme hexokinase 2 (HK2), and was involved in its phosphorylation, resulting in HK2 detachment from the mitochondria and consequently mPTP opening.

Introduction

Mitochondria, the main subcellular organelles for energy metabolism in eukaryotes, play extremely important roles in several physiological and pathological processes (1). Mitochondrial dysfunction mainly refers to an energy metabolism disorder caused by various factors including dissipation of the mitochondrial membrane, inhibition of the respiratory chain, reduction in enzyme activity, and mitochondrial DNA (mtDNA) damage, which can lead to a number of diseases including cancer (1). Mitochondrial functions are also regulated by mitochondrial dynamics (2,3). Under normal physiological conditions, mitochondria are highly dynamic and constantly undergo fusion and fission, but the equilibrium is disrupted under pathological conditions, when mitochondria fuse with each other into a long chain, ring, or mesh structure, or are split into smaller rod- or point-like structures. Increasing evidence has shown a strong association between cancer progression and unbalanced mitochondrial dynamics (2,3). Some studies have shown that disruption of the mitochondrial network strongly affects the apoptosis of cancer cells, including lung, bladder and colon cancer cells (2). Additionally, in lung cancer, clinical prognosis is significantly associated with abnormalities in proteins governing mitochondrial dynamics (4). In hepatocellular carcinoma (HCC), mitochondrial fission is frequently upregulated, significantly contributing to poor patient prognosis (5). Increased mitochondrial fission increases HCC cell survival, primarily by facilitating autophagy and inhibiting mitochondrial-dependent apoptosis, which are mediated via elevated production of reactive oxygen species (ROS) and subsequent degradation of p53 and activation of nuclear factor (NF)-κB (5). Moreover, mitochondrial fission activates calcium signaling in HCC cells, which in turn promotes mitochondrial fission in a positive feedback loop (6). However, the manner in which mitochondrial dynamics are regulated in HCC remains largely unclear.

Mitochondrial dynamics can be regulated by mitochondrial fusion proteins, such as mitofusin 1 (MFN1), mitofusin 2 (MFN2) and OPA1 mitochondrial dynamin like GTPase (OPA1), as well as mitochondrial fission proteins, such as dynamin-1-like (DNM1L), mitochondrial fission factor (MFF), fission 1 (FIS1) and mitochondrial dynamics protein 49/51 (MID49/51) (3). MFN1 and MFN2 together promote fusion of the outer mitochondrial membrane (OMM), while OPA1 regulates fusion of the inner mitochondrial membrane (IMM) (3). In mitochondrial fission, FIS1 protein is integrated into the OMM and acts as a receptor for DNM1L, which translocates from the cytosol to the OMM, and forms a ring that drives fission of the organelle (7). DNM1L is essential for the process of mitochondrial fission, whereas other fission proteins are replaceable as corresponding receptors required for DNM1L binding to the OMM (8). Therefore, aberrant expression of DNM1L causes severe disruptions in mitochondrial dynamics leading to pathologies, such as neurodegenerative, neoplastic, endocrine and cardiovascular diseases (8,9). Decreasing DNM1L in cardiomyocytes induces mitochondrial elongation, accumulation of damaged mitochondria and increased apoptosis (8). Furthermore, DNM1L knockdown has been shown to significantly enhance spontaneous apoptosis in vitro and in vivo in several cancer types (10,11). In HCC, DNM1L is aberrantly upregulated, and forced expression of DNM1L increases mitochondrial fission and promotes the survival of HCC cells (6,11). The expression of DNM1L may be regulated by both cytosolic Ca2+ signaling and p53/AR signaling (6). Cadmium levels have also been reported to regulate mitochondrial translocation of DNM1L (9). However, the manner in which the effect of DNM1L is regulated in HCC cells remains to be elucidated.

In this study, we investigated the effects of DNM1L on mitochondrial translocation and mitochondrial permeability transition pore (mPTP) opening in HCC cells under hypoxia, and explored the underlying molecular mechanisms and potential therapeutic applications. Our findings contribute to the understanding of the pathological roles of mitochondrial dynamics and provide strong evidence to support a novel strategy for HCC treatment targeting proteins of the mitochondrial fission machinery.

Materials and methods

Cell lines

The Huh7 cell line was obtained from the Shanghai Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in Dulbecco's modified Eagle's medium (DMEM) (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) supplemented with 10% fetal bovine serum (FBS) (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a humidified 5% CO2 incubator.

Immunoprecipitation and western blotting

Immunoprecipitation (IP) was carried out using the Protein G IP Kit (Roche, Basel, Switzerland) according to the manufacturer's instructions. SDS-PAGE was used for western blotting (WB) in order to separate proteins extracted from tissues or cells, and proteins were transferred onto polyvinylidene fluoride membranes. Membranes were incubated first with primary antibodies against DNM1L (dilution 1:1,000; cat. no. ab184247; Abcam), MID51 (dilution 1:100; cat. no. ab220672; Abcam), FIS1 (dilution 1:1,000; cat. no. ab71498; Abcam), mitochondrial fission factor (MFF) (dilution 1:1,000; cat. no. ab81127; Abcam), hypoxia inducible factor 1 subunit α (HIF1A) (dilution 1:1,000; cat. no. ab187524; Abcam, Cambridge, MA, USA), voltage-dependent anion channel (VDAC) (dilution 1:1,000; cat. no. 4661; Cell Signaling Technology, Danvers, MA, USA), adenine nucleotide translocator (ANT) (dilution 1:1,000; cat. no. 14671; Cell Signaling Technology), cyclophilin D (CypD) (dilution 1:1,000; cat. no. ab110324; Abcam), hexokinase 2 (HK2) (dilution 1:1,000; cat. no. 2867; Cell Signaling Technology), and GAPDH (dilution 1:10,000; cat. no. 60004-1-Ig; Proteintech, Chicago, IL, USA), followed by HRP-conjugated secondary antibody. Enhanced chemiluminescence was used for detection. WB results were quantified using ImageJ software (US National Institutes of Health, Bethesda, MD, USA; imagej.nih.gov/ij) following the WB quantification guidelines.

Real-time PCR

Total RNA was extracted from cells using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. RNA was then reverse transcribed and the resulting cDNA was used as a template for quantitative PCR using a Takara RNA PCR kit and SYBR Premix Ex Taq (Takara Bio Inc., Kusatsu, Japan) in accordance with the manufacturer's instructions. GAPDH was used as an internal control. The following primers were used: Human HIF1A, sense, 5′ to 3′-GAACGTCGAAAAGAAAAGTCTCG and antisense, 5′ to 3′-CCTTATCAAGATGCGAACTCACA; human DNM1L, sense, 5′ to 3′-CTGCCTCAAATCGTCGTAGTG and antisense, 5′ to 3′-GAGGTCTCCGGGTGACAATTC; human MID51, sense, 5′ to 3′-CACGGCCATTGACTTTGTGC and antisense, 5′ to 3′-TCGTACATCCGCTTAACTGCC; human GAPDH, sense, 5′ to 3′-GAGTCAACGGATTTGGTCGT and antisense, 5′ to 3′-TTGATTTTGGAGGGATCTCG; human FIS1, sense, 5′ to 3′-GTCCAAGAGCACGCAGTTTG and antisense, 5′ to 3′-ATGCCTTTACGGATGTCATCATT; human MFF, sense, 5′ to 3′-ACTGAAGGCATTAGTCAGCGA and antisense, 5′ to 3′-TCCTGCTACAACAATCCTCTCC. The RT-qPCR protocol was: i) 95°C for 2 min; ii) 95°C for 5 sec, 60°C for 30 sec, 72°C for 30 sec, 40 cycles; iii) dissociation curve. All results were calculated using the 2−ΔΔCq method (12).

siRNA

The siRNA sequence (5′-aatatccatctctggcca-3′) for DNM1L was purchased from Sigma-Aldrich; Merck KGaA. A scrambled siRNA precursor was used as the negative control. siRNAs were transfected into cells using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. After 48 h, the transfection efficiency was assessed using qPCR and WB, and the cells were used for further experiments.

Overexpression of DNM1L

The coding sequences of DNM1L were constructed into p3×Flag-CMV7 (Sigma-Aldrich; Merck KGaA). Transfection of siRNA and overexpression plasmids was carried out using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. In the overexpression assays, p3×Flag-CMV7 vectors were used as a control.

Immunofluorescence staining (IMF)

Cells were seeded onto coverslips coated with poly-L-lysine, and fixed with 4% paraformaldehyde before staining. Cells were permeabilized with 0.5% Triton X-100 in phosphate-buffered saline (PBS) at room temperature for 5 min, and blocked with 1% bovine serum albumin. Then, cells were incubated with primary antibodies (anti-DNML1 and/or anti-HK2), secondary antibodies, and then stained with DAPI and photographed with a TCS SP5 imaging system (Leica Microsystems, Wetzlar, Germany). For mitochondrial morphology, cells were incubated with 100 nM MitoTracker Deep Red (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C for 10 min.

Mitochondrial transmembrane potential assays

Mitochondrial transmembrane potential was determined using JC-1 staining (Sigma-Aldrich; Merck KGaA) according to the manufacturer's instructions. Briefly, cells were incubated with JC-1 for 20 min at 37°C. JC-1 monomers were stained green and JC-1 aggregate was stained red. Cell images were obtained and analyzed by IMF.

ROS detection

2,7-Dichlorofluorescein diacetate (DCFH-DA; Sigma-Aldrich; Merck KGaA) was used to determine intracellular ROS levels according to the manufacturer's instructions. Briefly, cells were incubated with 20 µM DCFH-DA for 30 min at 37°C. After washing the cells with PBS three times, images were obtained and analyzed by IMF.

mPTP opening test

Briefly, treated cells were incubated with 2 µM calcein (Sigma-Aldrich; Merck KGaA) and 100 nM MitoTracker Deep Red for 30 min in the absence of light. Cells were subsequently exposed to hypoxia (1% O2, 5% CO2, 94% N2) for 24 h. After washing the cells with PBS three times, images were obtained and analyzed using IMF.

Mitochondrial isolation

Mitochondrial proteins were isolated from tumor cells using Mitochondria Isolation Kit (MP-007; Invent Biotechnologies, Plymouth, MN, USA) according to the manufacturer's instructions. Briefly, the cell lysate was centrifuged at 20,000 × g for 1 min and the precipitate was suspended briefly. The floccule was centrifuged for 1 min at 1,500 × g after overhanging and then the supernatant was mixed by vortexing with 400 µl buffer. After centrifugation at 20,000 × g for 10 min, the supernatant was removed completely and the pellet was suspended in 200 µl buffer. After a further 30 min of centrifugation at 20,000 × g, the supernatant was discarded and the pellet comprised isolated mitochondria.

Cell viability assay

Cell viability was quantified with a Cell Counting Kit-8 (CCK-8) (Dojindo, Tokyo, Japan), according to the manufacturer's instructions. The cells were plated at a density of 3,000 cells per well in 96-well plates after transfection with siRNA in a 10 cm dish for 48 h. CCK-8 assays were assessed at 12, 24 and 48 h after transfection by measuring the absorbance at 450 nm.

Statistical analysis

All data are presented with error bars denoting standard deviation. All statistical data are based on experiments conducted in triplicate. Statistical analysis was performed with GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA). Differences between two groups were examined by Student's two-tailed t-test; two-way analysis of variance was used when comparing more than two groups. For comparisons among multiple groups, one-way analysis of variance (ANOVA) followed by Scheffe post hoc test was used. Correlations between two groups were analyzed using nonparametric Spearman's r-test. Two-sided P-values <0.05 were considered statistically significant.

Results

Hypoxia induces mitochondrial translocation of DNM1L in HCC cells

Mitochondrial function is affected by hypoxic exposure in different types of cells. To determine the effect of hypoxia on HCC cells, we measured two indicators of mitochondrial function, mitochondrial membrane potential and ROS generation, in hypoxia-exposed Huh7 cells. JC-1 fluorescence, which was used to determine the mitochondrial membrane potential, was significantly decreased by hypoxia, whereas ROS generation was significantly elevated (Fig. 1A and B). Meanwhile, a significant increase in mitochondrial fragments was observed in hypoxia-exposed Huh7 cells (Fig. 1C), suggesting that mitochondrial dynamic equilibrium of fusion and fission in Huh7 cells is disrupted by hypoxia. Next we determined whether regulators of mitochondrial fission, such as DNM1L, MID51, FIS1 and MFF, are affected in Huh7 cells under hypoxia. HIF1A was used as positive control under hypoxia. No significant changes were observed for any of these regulators at the mRNA or protein levels (Fig. 1D and E). Since the effect of DNMlL depends on their translocation into mitochondria, we also determined their levels in mitochondrial and cytoplasmic fractions. Among these proteins, only DNM1L was decreased in the cytoplasm and increased in mitochondria under hypoxic conditions (Fig. 1F). In addition, confocal images verified that hypoxia increased the recruitment of DNM1L into the mitochondria (Fig. 1G). Taken together, these data indicate that hypoxia induces mitochondrial translocation of DNM1L in HCC cells.

DNM1L translocation leads to excessive mitochondrial fission and decreased viability of HCC cells

Our confocal images revealed that hypoxia triggered mitochondrial fission and aggravated mitochondrial fragmentation (Fig. 1). To determine whether DNM1L is critical for this process, DNM1L was knocked down in Huh7 cells. Knockdown efficiency was determined by qPCR and WB analysis (Fig. 2A). DNM1L knockdown significantly elevated the mitochondrial membrane potential and significantly reduced ROS levels in both hypoxia-exposed and unexposed Huh7 cells, and significantly alleviated hypoxia-induced elevation of ROS generation and reduction of mitochondrial membrane potential (Fig. 2B and C). The alleviation was also observed with treatment of Huh7 cells with the DNM1L translocation inhibitor Mdivi (Fig. 2B and C). These data demonstrated the important role of DNM1L in hypoxia-induced excessive mitochondrial fission and the subsequent mitochondrial dysfunction. Since mitochondrial dynamics have been reported to regulate apoptosis and proliferation of HCC cells, we also determined the effects of DNM1L knockdown on the viability of Huh7 cells under hypoxia. DNM1L knockdown decreased the viability of Huh7 cells without hypoxic treatment, whereas hypoxia reversed the effects of DNM1L knockdown on viability, indicating that DNM1L alleviated the suppressive effects of hypoxia on tumor cell viability, and the oncogenic effects of DNM1L depends on hypoxic condition (Fig. 2D).

DNM1L also regulates mPTP opening

To note, compared to Mdivi treatment, DNM1L knockdown conferred stronger effects on mitochondrial function (Fig. 2B and C), suggesting that DNM1L regulates mitochondrial function in ways other than mitochondrial fission. Since DNM1L has been reported to regulate calcium signaling in HCC (6), and calcium signaling is closely associated with mPTP opening (13), we speculated that DNM1L may also be able to regulate mPTP opening in HCC cells. We analyzed the mPTP opening levels in calcein-CoCl2 tests, and found that DNM1L knockdown significantly inhibited mPTP opening in hypoxia-exposed Huh7 cells (Fig. 3A). Further, we overexpressed DNM1L in Huh7 cells (Fig. 3B) and treated these cells with CsA (an mPTP opening antagonist). CsA treatment alleviated the effects of DNM1L on mitochondrial membrane potential, ROS generation and cell viability (Fig. 3C-E).

DNM1L increases mitochondrial detachment of hexokinase 2 (HK2) to regulate mPTP opening

To further investigate the regulatory mechanism of DNM1L on mPTP opening, the expression and mitochondrial translocation of several mPTP structural proteins, including VDAC, ANT, CypD and HK2 were assessed. WB analysis showed that the total levels of these proteins were not altered after DNM1L knockdown in hypoxic Huh7 cells (Fig. 4A). Meanwhile, as control, no changes in mitochondrial translocation were observed for mitochondrial proteins CypD, VDAC, and ANT, whereas mitochondrial HK2 was increased after DNM1L knockdown (Fig. 4A), indicating that instead of regulating the expression of mPTP-related proteins, DNM1L regulates mitochondrial detachment of HK2 in hypoxia-exposed Huh7 cells. This effect of DNM1L on HK2 was demonstrated in our confocal images (Fig. 4B). To determine how DNM1L regulates HK2 translocation, we performed IP analysis and found a slight interaction between DNM1L and HK2 in Huh7 cells (Fig. 4C). Of note, hypoxia enhanced their binding affinity (Fig. 4C). Moreover, hypoxia increased the phosphorylation of HK2, which was attenuated by DNM1L knockdown (Fig. 4C). These data indicate that DNM1L interacts with HK2 and mediates the activation of HK2, which induces mitochondrial detachment, thus regulating the mPTP (Fig. 4D).

Discussion

Mitochondrial fusion and fission dynamically remodel the morphology of mitochondria within cells, and dysregulation of the fusion/fission process has been linked to the tumorigenesis of different types of cancers including hepatocellular carcinoma (HCC) (14). Recently, it was demonstrated that mitochondrial fission is frequently upregulated in HCC tissues mainly because of elevated expression of dynamin-1-like (DNM1L), which also was found to contribute to the poor prognosis of HCC patients (5,6,11). However, the manner in which DNM1L and mitochondrial dynamics regulate HCC cells remains largely unclear. In the present study, we found that hypoxia could induce the dissociation of DNM1L from mitochondria, and the resulting excessive mitochondrial fission promoted the viability of HCC cells. It is well established that hypoxia can induce mitochondrial fragmentation and mitophagy (15). In response to hypoxic stress, DNM1L is recruited by the outer mitochondrial membrane (OMM) protein FUNDC1 to drive mitochondrial fission (15). Hypoxia also induces HIF-1α activation which in turn activates DNM1L S616 phosphorylation (16). Hypoxia-induced DNM1L recruitment is often observed in disorders of the neuronal and vascular systems (17,18). Recently, the effects of hypoxia on mitochondrial fission were also found to promote migration, invasion, and tube formation in breast cancer, glioblastoma, and endothelial progenitor cells (1921). Meanwhile, DNM1L-dependent mitochondrial fission was shown to regulate hypoxia-induced migration of breast cancer cells, and to increase their sensitivity to chemotherapeutic agents (20). However, although DNM1L-mediated mitochondrial fission was found to promote proliferation and inhibit apoptosis in most previous studies (10,11), some reports also suggested that increased mitochondrial fission could be positively related with cell death in HCC, and the effect of hypoxia-induced DNM1L translocation may reduce tumor cell viability (21). Here, our results revealed that, in contrast to the effect in non-hypoxic conditions, DNM1L knockdown increased the viability of HCC cells in hypoxic conditions and alleviated the suppressive effects of hypoxia on tumor cell viability. These data indicated that DNM1L promotes cell viability in normal conditions, but hypoxia reverses this effects.

In addition to the effects on mitochondrial dynamics, our data showed that DNM1L also regulated the mitochondrial permeability transition pore (mPTP) opening in HCC cells. Excessive opening of mPTP produces osmotic dysregulation of the inner mitochondrial membrane (IMM), loss of mitochondrial cytochrome c and NADPH, uncoupling of oxidative phosphorylation, and cell death (22). There are many factors that regulate mPTP opening, including elevated mitochondrial ROS, excessive mitochondrial calcium levels, mitochondrial DNA damage, and cytoplasmic signal transduction (13). The liberation of HK2 from mitochondria has also been demonstrated to regulate mPTP opening (23). HK2 binds to the OMM via association with VDAC, which interacts with ANT, forming the mPTP (24). HK2 detachment from mitochondria induces a conformational change of the molecular complex, leading to mitochondrial depolarization and cell death (25). A recent study in neonatal cardiac myocytes revealed a reciprocal interaction between DNM1L and HK2 binding to mitochondria, and demonstrated that DNM1L knockdown prevented the loss of HK2 as well as mPTP opening (26). It has been suggested that the opposite translocation of DNM1L and HK2 may reflect a shared but competing binding region on the OMM, thus regulating mPTP opening (27). These effects of DNM1L on mPTP opening have been demonstrated in ischemia and hypoxia-exposed myocytes (27). In HCC cells, DNM1L-mediated mitochondrial fission has also been reported to increase the cytosolic calcium concentration and promote calcium oscillation (6), which might induce excessive mPTP opening (13). Here, we provided direct evidence for a connection between DNM1L and mPTP opening in HCC. Both DNM1L depletion and Mdivi treatment were able to improve mPTP opening. However, the opening of mPTP channels following hypoxia was not fully restored after DNM1L deletion or inhibition. A similar phenomenon was reported in dilated cardiomyopathy where mPTP channels remained open in DNM1L-deleted myocardial mitochondria (28), indicating that critical factors other than DNM1L are involved in regulating mPTP opening in hypoxia-exposed HCC cells. This possibility requires further investigation.

In conclusion, hypoxia induced mitochondrial translocation of DNM1L in HCC cells, leading to excessive mitochondrial fission and decreased viability of HCC cells. In hypoxia-exposed HCC cells, DNM1L also regulated mPTP opening via increasing mitochondrial detachment of HK2.

Acknowledgements

Not applicable.

Funding

This study was supported by the Science and Technology Research Project of Chongqing Education (grant no: KJ1600215) and the Preliminary National Natural Science Foundation of China at Chongqing Medical University (grant no: NSFYY201519).

Availability of data and materials

Not applicable.

Authors' contributions

MC, PH and DLF conceived and designed the study. DLF prepared the manuscript. MC and PH performed the experiments. All authors participated in the discussion and data interpretation. All the authors read and approved the manuscript and agree to be accountable for all the aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ward DM and Cloonan SM: Mitochondrial iron in human health and disease. Ann Rev Physiol. 10:453–482. 2018.

2 

Chen H and Chan DC: Mitochondrial dynamics in regulating the unique phenotypes of cancer and stem cells. Cell Metab. 26:39–48. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Youle RJ and van der Bliek AM: Mitochondrial fission, fusion, and stress. Science. 337:1062–1065. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Rehman J, Zhang HJ, Toth PT, Zhang Y, Marsboom G, Hong Z, Salgia R, Husain AN, Wietholt C and Archer SL: Inhibition of mitochondrial fission prevents cell cycle progression in lung cancer. FASEB J. 26:2175–2186. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Huang Q, Zhan L, Cao H, Li J, Lyu Y, Guo X, Zhang J, Ji L, Ren T, An J, et al: Increased mitochondrial fission promotes autophagy and hepatocellular carcinoma cell survival through the ROS-modulated coordinated regulation of the NFKB and TP53 pathways. Autophagy. 12:999–1014. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Huang Q, Cao H, Zhan L, Sun X, Wang G, Li J, Guo X, Ren T, Wang Z, Lyu Y, et al: Mitochondrial fission forms a positive feedback loop with cytosolic calcium signaling pathway to promote autophagy in hepatocellular carcinoma cells. Cancer Lett. 403:108–118. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Cerveny KL, Tamura Y, Zhang Z, Jensen RE and Sesaki H: Regulation of mitochondrial fusion and division. Trends Cell Biol. 17:563–569. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Hu C, Huang Y and Li L: Drp1-Dependent mitochondrial fission plays critical roles in physiological and pathological progresses in mammals. Int J Mol Sci. 18:2017. View Article : Google Scholar

9 

Kasahara A and Scorrano L: Mitochondria: From cell death executioners to regulators of cell differentiation. Trends Cell Biol. 24:761–770. 2014. View Article : Google Scholar

10 

Kashatus JA, Nascimento A, Myers LJ, Sher A, Byrne FL, Hoehn KL, Counter CM and Kashatus DF: Erk2 phosphorylation of Drp1 promotes mitochondrial fission and MAPK-driven tumor growth. Mol Cell. 57:537–551. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Zhan L, Cao H, Wang G, Lyu Y, Sun X, An J, Wu Z, Huang Q, Liu B and Xing J: Drp1-mediated mitochondrial fission promotes cell proliferation through crosstalk of p53 and NF-kappaB pathways in hepatocellular carcinoma. Oncotarget. 7:65001–65011. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Bernardi P, Rasola A, Forte M and Lippe G: The mitochondrial permeability transition pore: Channel formation by F-ATP synthase, integration in signal transduction, and role in pathophysiology. Physiol Rev. 95:1111–1155. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Zong WX, Rabinowitz JD and White E: Mitochondria and cancer. Mol Cell. 61:667–676. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Wu W, Lin C, Wu K, Jiang L, Wang X, Li W, Zhuang H, Zhang X, Chen H, Li S, et al: FUNDC1 regulates mitochondrial dynamics at the ER-mitochondrial contact site under hypoxic conditions. EMBO J. 35:1368–1384. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Zhang D, Liu Y, Tang Y, Wang X, Li Z, Li R, Ti Z, Gao W, Bai J and Lv Y: Increased mitochondrial fission is critical for hypoxia-induced pancreatic beta cell death. PLoS One. 13:e01972662018. View Article : Google Scholar : PubMed/NCBI

17 

Qu Y, Shi J, Tang Y, Zhao F, Li S, Meng J, Tang J, Lin X, Peng X and Mu D: MLKL inhibition attenuates hypoxia-ischemia induced neuronal damage in developing brain. Exp Neurol. 279:223–231. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Han Q, Li G, Ip MS, Zhang Y, Zhen Z, Mak JC and Zhang N: Haemin attenuates intermittent hypoxia-induced cardiac injury via inhibiting mitochondrial fission. J Cell Mol Med. 22:2717–2726. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Kim DY, Jung SY, Kim YJ, Kang S, Park JH, Ji ST, Jang WB, Lamichane S, Lamichane BD, Chae YC, et al: Hypoxia-dependent mitochondrial fission regulates endothelial progenitor cell migration, invasion, and tube formation. Korean J Physiol Pharmacol. 22:203–213. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Han XJ, Yang ZJ, Jiang LP, Wei YF, Liao MF, Qian Y, Li Y, Huang X, Wang JB, Xin HB and Wan YY: Mitochondrial dynamics regulates hypoxia-induced migration and antineoplastic activity of cisplatin in breast cancer cells. Int J Oncol. 46:691–700. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Wan YY, Zhang JF, Yang ZJ, Jiang LP, Wei YF, Lai QN, Wang JB, Xin HB and Han XJ: Involvement of Drp1 in hypoxia-induced migration of human glioblastoma U251 cells. Oncol Rep. 32:619–626. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Perez MJ and Quintanilla RA: Development or disease: Duality of the mitochondrial permeability transition pore. Dev Biol. 426:1–7. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Zhou H, Zhang Y, Hu S, Shi C, Zhu P, Ma Q, Jin Q, Cao F, Tian F and Chen Y: Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis. J Pineal Res. 63:2017. View Article : Google Scholar :

24 

Chatterjee A, Mambo E and Sidransky D: Mitochondrial DNA mutations in human cancer. Oncogene. 25:4663–4674. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Nederlof R, Eerbeek O, Hollmann MW, Southworth R and Zuurbier CJ: Targeting hexokinase II to mitochondria to modulate energy metabolism and reduce ischaemia-reperfusion injury in heart. Br J Pharmacol. 171:2067–2079. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Yang YL, Li J, Liu K, Zhang L, Liu Q, Liu B and Qi LW: Ginsenoside Rg5 increases cardiomyocyte resistance to ischemic injury through regulation of mitochondrial hexokinase-II and dynamin-related protein 1. Cell Death Dis. 8:e26252017. View Article : Google Scholar : PubMed/NCBI

27 

Andrienko TN, Pasdois P, Pereira GC, Ovens MJ and Halestrap AP: The role of succinate and ROS in reperfusion injury-A critical appraisal. J Mol Cell Cardiol. 110:1–14. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Song M, Mihara K, Chen Y, Scorrano L and Dorn GW II: Mitochondrial fission and fusion factors reciprocally orchestrate mitophagic culling in mouse hearts and cultured fibroblasts. Cell Metab. 21:273–286. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2019
Volume 42 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cai M, He P and Fang DL: Hypoxia‑induced mitochondrial translocation of DNM1L increases mitochondrial fission and triggers mPTP opening in HCC cells via activation of HK2 Retraction in /10.3892/or.2021.8059. Oncol Rep 42: 1125-1132, 2019
APA
Cai, M., He, P., & Fang, D. (2019). Hypoxia‑induced mitochondrial translocation of DNM1L increases mitochondrial fission and triggers mPTP opening in HCC cells via activation of HK2 Retraction in /10.3892/or.2021.8059. Oncology Reports, 42, 1125-1132. https://doi.org/10.3892/or.2019.7213
MLA
Cai, M., He, P., Fang, D."Hypoxia‑induced mitochondrial translocation of DNM1L increases mitochondrial fission and triggers mPTP opening in HCC cells via activation of HK2 Retraction in /10.3892/or.2021.8059". Oncology Reports 42.3 (2019): 1125-1132.
Chicago
Cai, M., He, P., Fang, D."Hypoxia‑induced mitochondrial translocation of DNM1L increases mitochondrial fission and triggers mPTP opening in HCC cells via activation of HK2 Retraction in /10.3892/or.2021.8059". Oncology Reports 42, no. 3 (2019): 1125-1132. https://doi.org/10.3892/or.2019.7213