TOP

Research Article

Split Viewer

Mol. Cells 2023; 46(4): 219-230

Published online January 10, 2023

https://doi.org/10.14348/molcells.2023.2095

© The Korean Society for Molecular and Cellular Biology

RUNX1 Upregulation Causes Mitochondrial Dysfunction via Regulating the PI3K-Akt Pathway in iPSC from Patients with Down Syndrome

Yanna Liu1,4 , Yuehua Zhang1,4 , Zhaorui Ren1,2 , Fanyi Zeng1,2,3,* , and Jingbin Yan1,2,*

1Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai 200040, China, 2NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, China, 3Department of Histoembryology, Genetics & Development, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, 4These authors contributed equally to this work.

Correspondence to : yanjb@shchildren.com.cn (JY); fzeng@vip.163.com (FZ)

Received: June 6, 2022; Revised: September 30, 2022; Accepted: October 3, 2022

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.

Down syndrome (DS) is the most common autosomal aneuploidy caused by trisomy of chromosome 21. Previous studies demonstrated that DS affected mitochondrial functions, which may be associated with the abnormal development of the nervous system in patients with DS. Runt-related transcription factor 1 (RUNX1) is an encoding gene located on chromosome 21. It has been reported that RUNX1 may affect cell apoptosis via the mitochondrial pathway. The present study investigated whether RUNX1 plays a critical role in mitochondrial dysfunction in DS and explored the mechanism by which RUNX1 affects mitochondrial functions. Expression of RUNX1 was detected in induced pluripotent stem cells of patients with DS (DS-iPSCs) and normal iPSCs (N-iPSCs), and the mitochondrial functions were investigated in the current study. Subsequently, RUNX1 was overexpressed in N-iPSCs and inhibited in DS-iPSCs. The mitochondrial functions were investigated thoroughly, including reactive oxygen species levels, mitochondrial membrane potential, ATP content and lysosomal activity. Finally, RNA-sequencing was used to explore the global expression pattern. It was observed that the expression levels of RUNX1 in DS-iPSCs were significantly higher than those in normal controls. Impaired mitochondrial functions were observed in DS-iPSCs. Of note, overexpression of RUNX1 in N-iPSCs resulted in mitochondrial dysfunction, while inhibition of RUNX1 expression could improve the mitochondrial function in DS-iPSCs. Global gene expression analysis indicated that overexpression of RUNX1 may promote the induction of apoptosis in DS-iPSCs by activating the PI3K/Akt signaling pathway. The present findings indicate that abnormal expression of RUNX1 may play a critical role in mitochondrial dysfunction in DS-iPSCs.

Keywords Down syndrome, induced pluripotent stem cells, mitochondrial dysfunction, RUNX1

Down syndrome (DS) or trisomy 21 is one of the most common genetic diseases and is often accompanied by intellectual disability. It occurs with an incidence ~1 in 700-1,000 births (El Hajj et al., 2016). DS is a major cause of congenital malformations and mental retardation in humans. Patients with DS typically display associated dysfunctions and a range of cognitive deficits, with important differences in their phenotype and severe side effects in the nervous system (Vacca et al., 2019). Previous studies on the brains of patients with DS have revealed abnormalities in the structure and function of the central nervous system. Recent studies have shown that alterations in mitochondrial function may be associated with the occurrence of DS (Pecze and Szabo, 2021; Zamponi and Helguera, 2019).

Mitochondria play a central role in metabolism and energy conversion in eukaryotic cells, which generate ATP via oxidative phosphorylation (OXPHOS). These organelles provide energy for various cell-based activities (Ohnishi et al., 2018). Dysregulated mitochondria lead to the excessive production of reactive oxygen species (ROS), which are involved in the regulation of programmed cell death (Li et al., 2019; Wong, 2011). Impaired mitochondrial function leads to significant changes in the cell and affects neural cell proliferation, apoptosis, differentiation, regeneration, and other cellular activities (Beckervordersandforth, 2017; Johnson et al., 2021). It is known that mitochondrial dysfunction/oxidative stress is associated with DS. Mitochondrial dysfunction has been observed in the brain of early fetuses with DS and can manifest as abnormalities in OXPHOS complexes (Coskun and Busciglio, 2012; Salemi et al., 2018), accumulation of oxidative stress products and changes in mitochondrial biosynthesis. In addition, previous studies demonstrated that abnormal expression of genes was associated with mitochondrial function in DS (Qiu et al., 2017; Salemi et al., 2020). Mitochondrial dysfunction leads to impaired neuron proliferation, differentiation, and maturation. Previous studies have shown the presence of mitochondrial dysfunction in neurons, glial cells, and peripheral blood cells in patients with DS, and they were associated with mitochondrial fragmentation, impaired OXPHOS, and reduced ATP levels (Valenti et al., 2018; Zamponi and Helguera, 2019). Collectively, these studies indicated that mitochondrial dysfunction was strongly associated with abnormal development of the nervous system in DS. However, the key genes and molecular mechanisms that lead to mitochondrial dysfunction in patients with DS remain unknown.

DS is caused by an extra copy of the human chromosome 21 (Hsa21). Previous studies have proposed the gene dosage effect hypothesis, suggesting that increased expression of a specific set of dosage-sensitive genes on Hsa21 may eventually cause neurodevelopmental or neurocognitive disorders in DS (Delabar et al., 1993). A recent study indicated that trisomy 21 caused neuronal apoptosis, leading to a decrease in the neural population (Hirata et al., 2020). The DS critical region (DSCR), namely 21q22-21q23, is a genomic region on Hsa21 that is strongly linked to DS phenotypes (Pelleri et al., 2016). Aberrant expression of genes, notably transcription factors located in the DSCR, may be associated with mitochondrial dysfunction in DS (Flippo and Strack, 2017; Izzo et al., 2018).

Runt-related transcription factor 1 (RUNX1), which is encoded by the RUNX1 gene, is localized in chromosome 21q22.12. This transcription factor is an important transcriptional regulator found in the DSCR. It is known that RUNX1 is an essential master regulator, which is active during hematopoietic development. In recent years, an increasing number of studies have found that RUNX1 plays an essential role in the development of the central nervous system, the proliferation and differentiation of neural progenitor cells, neuronal differentiation and control of axon growth (Halevy et al., 2016; Yoshikawa et al., 2015; 2016). Gutti et al. (2018) demonstrated that Justicia adhatoda could upregulate the expression levels of RUNX1 and enhance mitochondrial ROS generation in human megakaryocytic cells. Excessive ROS production can damage mitochondrial proteins, which ultimately results in mitochondrial dysfunction (Zhang et al., 2019). Several studies have reported that microRNA (miR)-27b may repress the mitochondrial pathway of apoptosis by targeting RUNX1 (Li et al., 2017; Zhao et al., 2016). However, it is unknown whether RUNX1 plays a critical role in the mitochondrial dysfunction observed in DS. Therefore, the present study investigated the role of RUNX1 in the development of mitochondrial dysfunction in DS and its ability to affect mitochondrial functions.

Human pluripotent stem cell culture and treatments

Normal induced pluripotent stem cells (N-iPSCs) and induced pluripotent stem cells of patients with DS (DS-iPSCs) were purchased from the American Type Culture Collection (Cat. No. ATCC DYR0100 [normal, newborn, male] and Cat. No. ATCC DYP0730 [DS, newborn, male]; ATCC, USA). The clones were cultured in a feeder-free system. The cell culture dishes were coated with Cell Matrix Basement Membrane Gel (Cat. No. ACS-3035; ATCC) and cultured in Complete Pluripotent Stem Cell SFM XF/FF medium (Cat. No. ACS-3002; ATCC) in the presence of 10 µM inhibitor (Cat. No. Y0503; Sigma, USA). The culture medium was changed the day after cell resuscitation, and daily thereafter until the colonies reached 80% confluence. The cells were typically digested, sub-cultured at a 1:3 ratio using the Stem Cell Dissociation Reagent (Cat. No. ACS-3010; ATCC) and incubated overnight at 37°C in the presence of 5% CO2.

DS-iPSCs were then cultured in the medium supplemented with 20 µM LY294002 (Cat. No. S1105; Selleck, USA) for 1 h. Then the cells were collected, the expression level of Akt was identified by western blotting and the analysis of ROS level and cellular ATP content was also performed. The experiment was repeated in triplicate.

Plasmid vectors

The following vectors were purchased from AddGene (USA); RUNX1 expression vector (pCMV5-AML1B; plasmid Cat. No. 12426) and RUNX1 short hairpin RNA (shRNA) vector (pLKO.1 shRUNX1 puro; plasmid Cat. No. 45816).

Electroporation of iPSCs

Single clones were harvested from a 60-mm dish using Stem Cell Dissociation Reagent within 10 min and washed once with 2 ml phosphate-buffered saline (PBS). Subsequently, the cells were resuspended in R-buffer. N-iPSCs and DS-iPSCs were transfected with the Neon Transfection System (Cat. No. MPK1025; Invitrogen, USA) according to the manufacturer’s instructions. The conditions used were as follows: 1,240 V, 20 ms and 2-times pulses. A total volume of 10 µl containing 1 µg plasmid DNA was used. Following transfection, the samples were placed on a 24-well microplate, and subsequent analysis was performed 48 h after transfection.

Identification of the expression level of RUNX1 by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting (WB)

Total RNA was extracted from PBMCs and iPSCs using TRIzol® reagent. RT was performed using a reverse transcription kit (Cat. No. 18091050; Invitrogen). RT-qPCR was performed with TaqmanTM Real-Time PCR Assay. The primer sequences for RUNX1 and GAPDH were as follows: RUNX1, forward: 5’-TGGCACTCTGGTCACCGTCAT-3’ and reverse: 5’-GAAGCTCTTGCCTCTACCGC-3’, and GAPDH, forward: 5’-AGAGGGCTGTCGGCGCAGTA-3’ and reverse: 5’-GGCTGTGGTCTCGGTTGGGC-3’. The reactions were performed using an ABI7500 Real-Time PCR system (Thermo Fisher Scientific, USA). The transcript levels of RUNX1 were quantified using the 2-ΔΔCq method, with GAPDH as the reference gene used for normalization.

The iPSCs were scraped off in ice-cold PBS, collected into RIPA lysis buffer (Cat. No. sc-24948; Santa Cruz Biotechnology, USA) with a cell scraper, and incubated on ice for 30 min. The cell lysates were centrifuged at 14,000 × g for 15 min at 4°C, and the supernatants were then collected. The protein concentration was determined by BCA assay (Cat. No. P0010; Beyotime, China). Protein denaturation was performed at 98°C for 10 min. Lysed proteins (20 µg/lane) were separated by 10% SDS-PAGE and transferred onto nitrocellulose membranes, which were then blocked with 5% non-fat milk in TBS containing 0.05% Tween-20 (TBST) for 1 h at room temperature. Subsequently, the membranes were incubated at 4°C overnight with primary antibodies against RUNX1 (1:1,000, Cat. No. ab272456; Abcam) and GAPDH (1:5,000, Cat. No. 10494-1-AP; ProteinTech Group, USA). GAPDH was used as a loading control for normalization. Subsequently, the membranes were incubated with a secondary antibody (1:2,000, Cat. No. SA00001-2; ProteinTech Group) at room temperature for 1 h. The membranes were washed three times with TBST and visualized by chemiluminescence in an Amersham Imager 600 (GE Healthcare, USA).

Analysis of ROS levels using 2’-7’dichlorofluorescein diacetate (DCFH-DA)

ROS levels in iPSCs were measured using a ROS Assay Kit (Cat. No. S0033; Beyotime), following the manufacturer’s instructions. Briefly, iPSCs were cultured in 6-well culture plates pretreated with Matrigel, and subsequently, the cell culture medium was removed. The cells were then incubated with 10 µM DCFH-DA, which is a fluorescent probe used for ROS detection. Following subsequent culture at 37°C for 20 min in the dark, the iPSCs were washed three times with serum-free Dulbecco’s modified Eagle’s medium (DMEM), and the fluorescence emission of the samples was detected by fluorescence microscopy (Nikon Eclipse Ti; Nikon, Japan). The fluorescence intensity was analyzed using ImageJ software (ver. No. 1.6; National Institutes of Health, USA).

Lipid peroxidation measurement

BODIPY-C11 staining was performed according to the manufacturer’s protocol. The cells were seeded in a 35-mm-diameter dish. Culture medium was replaced with 2 ml medium containing 5 µM of BODIPY-C11 (Cat. No. D3861; Invitrogen) after 48 h and the culture was returned to the cell culture incubator for 30 min. Finally, the cells were fixed with 4% paraformaldehyde for 15 min and cell nuclei were stained with DAPI for 10 min. Fluorescence intensity was examined by fluorescence microscopy (Nikon Eclipse Ti), and images were analyzed with the ImageJ software.

Detection of mitochondrial membrane potential (MMP) with JC-1

Cells were seeded in culture flasks, and when they were 60%-70% confluent, the culture solution was aspirated. The cells were then washed once with PBS, and 2 ml cell culture medium containing 1 ml JC-1 solution (Cat. No. S2003S; Beyotime) was added for staining. The solution was thoroughly mixed in a cell culture incubator for 20 min at 37°C. Concomitantly, the JC-1 staining buffer (5×) was diluted five times in water (4 ml distilled water and 1 ml JC-1 staining buffer). The samples were placed on ice. Following incubation at 37°C, the supernatant was aspirated and washed with JC-1 buffer (1×) three times. Upon addition of 2 ml medium, the fluorescence emission of the samples was detected using a fluorescence microscope. The experiments were repeated three times.

Measurement of cellular ATP content

Cellular ATP content was determined with an ATP Assay Kit according to the manufacturer’s instructions (Cat. No. S0026; Beyotime). Briefly, the cells were lysed with ATP Detection Lysis Buffer 48 h after transfection, and 100 µl ATP detection working solution was added to the wells for 5 min at room temperature. A total of 20 µl sample or standard was added to each well, and the ATP concentration was measured using luminometry. A BCA Protein Assay Kit (Cat. No. P0010; Beyotime) was used to determine the protein concentrations of each sample, and the total ATP levels were evaluated as the ratio of cellular ATP level to protein concentration.

Lysosome detection using LysoTrackerTM Red

Following transfection of iPSCs with RUNX1-overexpressing plasmids for 48 h, the transfected cells cultured on 35-mm culture dishes were incubated with 100 nM lysosome probes (Cat. No. M7512; Invitrogen) at 37°C for 20 min. Following incubation, the cells were fixed with 4% paraformaldehyde for 15 min and 0.2% Triton X was used for permeabilization for 10 min. This was followed by DAPI staining (Cat. No. C1002; Beyotime) for 8 min to visualize the nuclei. Red fluorescence was determined by fluorescence microscopy, and image analysis was performed with ImageJ software.

Cell apoptosis assay

Cell apoptosis was evaluated with a Cell Apoptosis Kit (Cat. No. APOAF; Sigma), according to the manufacturer’s instructions. Briefly, iPSCs were collected 24 h following transfection of RUNX1-overexpressing plasmids. Subsequently, 500 µl binding buffer, 5 µl Annexin V-FITC conjugate and 10 µl propidium iodide solution were added successively into the cell suspension, and incubated at room temperature in the dark for 10 min. The fluorescence emission of the cells was immediately detected with a flow cytometer (BD AccuriTM C6 Flow Cytometer; BD Biosciences, USA).

RNA-sequencing (RNA-seq) and differential expression analysis

RNA-seq was conducted with the NovaSeq 6000 platform (Illumina, USA). An experimental group transfected with a RUNX1 expression vector and a control group transfected with a GFP expression vector were used. Each group had three replicates.

Following total RNA extraction using TRIzol® reagent (Cat. No. 15596018; Invitrogen), the quantification, qualification, library preparation, and subsequent RNA-seq of the above two groups were performed by Novogene (China). The details of RNA-seq have been previously described (Jia et al., 2020). Raw data were cleaned by removing reads with adaptors and excluding low quality (>50%) or a high proportion of unknown bases (>10%). Subsequently, the data were assembled using Trinity methodology (Grabherr et al., 2011).

For each sequenced library, the read counts were adjusted using the edgeR program package through one scaling normalized factor prior to differential gene expression analysis. Differential expression analysis of two conditions was performed using the edgeR R package. P values were adjusted using the Benjamin and Hochberg method. Corrected P = 0.05 (Q-value) and absolute fold-change = 2 was set as the thresholds for significant differential expression. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was conducted, and the corrected P value cutoff was set at 0.05.

Differential expression genes and apoptotic biomarkers were confirmed by RT-qPCR. The primers are listed in Supplementary Table S1. To detect activation of the PI3K-Akt signaling pathway, the protein expression and phosphorylation levels of Akt, S6, and 4E-binding protein (4E-BP1) were detected by western blotting as previously described. The antibodies used are listed in Supplementary Table S2.

Impaired mitochondrial function is observed in DS-iPSCs

Our previous study demonstrated that the expression levels of almost all the genes related to mitochondrial function were downregulated in DS-iPSCs, suggesting that mitochondria were dysfunctional in these cells (Qiu et al., 2017). To further demonstrate the impairment of mitochondrial function in DS-iPSCs, specific changes in mitochondrial MMP, ROS and ATP content were investigated to provide further evidence of impaired mitochondrial function. As shown in Figs. 1A and 1B, a, a high green fluorescence intensity was noted in the DS-iPSCs. The quantification of green fluorescence intensity was analyzed by ImageJ (Alafnan et al., 2021), and the results suggested that the contents of ROS were increased 2.24-fold in DS-iPSCs (0.076 ± 0.004 vs 0.034 ± 0.003; P < 0.0001). The ATP contents in DS-iPSCs were 1.68-fold lower than those in N-iPSCs (3.95 ± 0.39 vs 6.64 ± 0.17; P = 0.0029) (Fig. 1C), indicating that energy production by mitochondria was impaired in DS. JC-1 staining was also used to determine whether the MMP was impaired in DS-iPSCs. The results indicated that higher green fluorescence signals and weaker red fluorescence signals were present in DS-iPSCs, resulting in an attenuated ratio of red/green fluorescence intensity (3.12 ± 0.38 vs 7.05 ± 0.38; P < 0.0001), indicating that there was loss of MMP in DS-iPSCs (Figs. 1D and 1E). The aforementioned results indicated that the loss of mitochondrial function in DS-iPSCs could lead to damage of MMP, increased ROS levels and reduced ATP synthesis.

Overexpression of RUNX1 affects mitochondrial function in iPSCs

Previous studies have shown that RUNX1 is involved in the pathogenesis of DS (Bourquin et al., 2006; Edwards et al., 2009; Patel et al., 2011). Furthermore, overexpression of RUNX1 was detected in DS-iPSCs and peripheral blood of children with DS (Supplementary Fig. S1), leading to the speculation that RUNX1 may be a key gene regulating mitochondrial function in DS. To determine whether RUNX1 affects mitochondrial function, RUNX1 was overexpressed in N-iPSCs (Supplementary Fig. S2). The results indicated that mitochondrial dysfunction was present in N-iPSCs following RUNX1 overexpression, which was similar to that in DS-iPSCs. JC-1 staining indicated that the green fluorescence intensity was considerably enhanced in RUNX1-overexpressing N-iPSCs compared with that observed in N-iPSCs expressing normal levels of RUNX1 (0.090 ± 0.017 vs 0.037 ± 0.0039; P = 0.035). These data suggested that RUNX1 promoted the production of ROS in iPSCs (Figs. 2A and 2B).

Accumulation of ROS in mitochondria causes abnormalities in mitochondrial structure and function, including ATP synthesis obstruction, reduced mitochondrial MMP, and impaired respiratory chain function. Mitochondrial respiratory chain deficiencies block OXPHOS and inhibit ROS production, which aggravates mitochondrial dysfunction and creates a vicious cycle of mitochondrial damage (Bhatti et al., 2017; Hu and Liu, 2011). Thus, mitochondrial function was evaluated in iPSCs overexpressing RUNX1. The results indicated that overexpression of RUNX1 led to a 3.17-fold decrease in ATP content compared with that of control cells (4.75 ± 0.36 vs 2.11 ± 0.26; P = 0.0043) (Fig. 2C). JC-1 indicated that overexpression of RUNX1 increased the intensity of green fluorescence in RUNX1-overexpressing N-iPSCs, while the reduction in the ratio of red to green fluorescence (2.92 ± 0.38 vs 1.44 ± 0.50; P = 0.015) indicated a loss of MMP (Figs. 2D and 2E). These data suggested that RUNX1 overexpression resulted in the accumulation of ROS levels, which ultimately caused mitochondrial dysfunction in DS-iPSCs.

High levels of mitochondrial ROS can induce oxidative damage in mitochondria, which impairs the function of the mitochondrial respiratory chain, leading to the accumulation of intracellular ROS. The membrane ROS levels of iPSCs were determined using BODIPYTM C11, and the data confirmed that RUNX1 overexpression increased mitochondrial and cell membrane ROS levels compared with those of control cells (0.10 ± 0.0034 vs 0.077 ± 0.0015; P < 0.0001) (Figs. 2F and 2G). Mitochondrial dysfunction was previously reported to promote ROS production and impair lysosome function in macrophages cultured under hyperglycemic conditions (Yuan et al., 2019). The present study experimentally confirmed that a high ROS content was present in DS-iPSCs with high expression of RUNX1. The inhibition of lysosomal activity was measured using LysoTrackerTM Red probe, and the data revealed that the number of active lysosomes was reduced in iPSCs overexpressing RUNX1 (0.75 ± 0.054 vs 0.43 ± 0.048; P = 0.0014) (Figs. 2H and 2I). Therefore, it was concluded that overexpression of RUNX1 may disrupt the normal function of mitochondria, thereby affecting the cell biological function of DS-iPSCs.

Inhibition of RUNX1 expression can improve the mitochondrial function of DS- iPSCs

To further determine whether RUNX1 overexpression could affect mitochondrial function, DS-iPSCs were transfected with RUNX1 shRNA vector, and the mitochondrial function was measured. The results indicated that the green fluorescence intensity was markedly decreased when the expression levels of RUNX1 were inhibited in DS-iPSCs (0.068 ± 0.015 vs 0.029 ± 0.0020; P = 0.017) compared with that exhibited by normal controls, indicating that the increase in ROS content in DS-iPSCs was closely associated with high expression levels of RUNX1 (Figs. 3A and 3B). In addition, ATP content was only slightly increased in the DS-iPSC + shRUNX1 group compared with that in control cells (5.19 ± 0.58 vs 6.86 ± 0.52; P = 0.10) (Fig. 3C). Furthermore, overexpression of RUNX1 also increased the green fluorescence intensity in the cells and the reduction in the ratio of red to green fluorescence (2.26 ± 0.18 vs 2.82 ± 0.11; P = 0.020) (Figs. 3D and 3E), which indicated that the MMP of DS-iPSCs was significantly recovered when the expression levels of RUNX1 were inhibited.

Overexpression of RUNX1 in iPSCs leads to abnormalities in the PI3K-Akt signaling pathway

RNA-seq analysis indicated that 51 differentially expressed genes were identified between the experimental and control groups using the edgeR R package (Fig. 4A). This included 39 upregulated and 12 downregulated genes (Fig. 4B). KEGG analysis revealed a set of 39 upregulated genes, and the results indicated that PI3K-Akt was identified as the most significant signaling pathway, in which five genes (IGF2, ITGA11, ITGB3, CREB5, and Bim) were involved (Fig. 4C). The RT-qPCR results confirmed that the expression levels of these genes were markedly increased in RUNX1-overexpressing N-iPSCs (Fig. 4D).

A previous study has shown that the activation of mTOR signaling via the PI3K/Akt signaling pathway is involved in the regulation of mitochondrial ROS production in PC-12 cells, which can ultimately affect cell apoptosis (Li et al., 2020). In the present study, PI3K/Akt inhibitor LY294002 was used to suppress the expression of Akt in DS-iPSCs. The results showed that the expression level of Akt in DS-iPSCs was significantly decreased, accompanied by a significant increase in ATP content and a decrease in ROS level (Supplementary Fig. S3).

Akt is a major downstream effector molecule of the PI3K/Akt signaling pathway, and increased phosphorylation implies activation of the PI3K signaling pathway (Hart and Vogt, 2011; McCubrey et al., 2012). The present results revealed a significant increase in the phosphorylation of Akt, S6 and 4EBP1 in N-iPSCs + RUNX1 cells (Fig. 5A). These results indicated that upregulation of RUNX1 in iPSCs activated the PI3K-Akt signaling pathway by increasing the phosphorylation of Akt, S6 and 4EBP1.

The effects of RUNX1 overexpression on the induction of apoptosis of iPSCs were investigated by flow cytometry. A significant increase in apoptosis was observed in RUNX1-overexpressing N-iPSCs (19.00 ± 1.79 vs 32.46 ± 1.54; P = 0.0047) (Figs. 5B and 5C). Furthermore, the results of expression analysis of apoptotic biomarkers (Bax, Bcl-2, and caspase 3) further supported the hypothesis that overexpression of RUNX1 may promote apoptosis in DS-iPSCs by activating the PI3K/Akt signaling pathway (Fig. 5D).

The most apparent clinical features of DS are neurological complications, including early onset of Alzheimer’s disease (AD) and mental retardation. The central nervous system requires high quantities of energy. Redox homeostasis and adequate ATP levels are essential for normal neurodevelopment (Arrázola et al., 2019; Kann and Kovács, 2007). Mitochondria supply virtually all eukaryotic cells with energy through ATP production by OXPHOS. Accordingly, maintenance of the mitochondrial function is fundamentally important to sustain cellular health. Previous studies reported that mitochondrial abnormalities, including aberrant mitochondrial morphology and function, were detected in both patients with DS and in DS cells (Aburawi and Souid, 2012; Shah et al., 2019). In the present study, mitochondrial abnormalities were confirmed in DS-iPSCs. Therefore, it is reasonable to speculate that the aberrant development of the nervous system in DS is closely associated with mitochondrial dysfunction, as iPSCs have the pluripotency to differentiate into various nerve cell types. However, the mechanism leading to mitochondrial dysfunction in DS is still not clear, and the key genes responsible for this process have not been identified thus far.

RUNX1 is an important transcriptional regulator located in DSCR, which is closely associated with the development and regeneration of the nervous system. A recent study indicated that large-scale hypermethylation of RUNX1 (~28 kb) was the strongest signal within the DS differentially methylated regions in peripheral blood (Laufer et al., 2021), which was also confirmed by the current study. In the present study, overexpression of RUNX1 was observed in both DS-iPSCs and peripheral blood samples of children with DS. It was therefore speculated that RUNX1 may be a crucial regulatory gene involved in the pathogenesis of DS. In a recent study, RUNX1 mutations were detected in a patient diagnosed with Pearson syndrome, which was caused by deletions in mitochondrial DNA (Nishimura et al., 2021).

The present study investigated whether RUNX1 affects the neurological development of DS by regulating mitochondrial function. The results revealed that overexpression of RUNX1 induced impaired mitochondrial functions, characterized by excessive generation of ROS, decreased MMP and reduced overall energy metabolism. It was also observed that RUNX1 could reduce the number of active lysosomes and cause lysosomal dysfunction. Furthermore, the data confirmed that impaired mitochondrial functions, including excessive generation of ROS, and decreased MMP and ATP content, could be reversed when the expression levels of RUNX1 were inhibited in DS-iPSCs. All these results strongly suggested that the abnormal expression levels of RUNX1 may be a critical factor (although not the sole factor) in the occurrence of mitochondrial dysfunction in DS-iPSCs, and indicated that overexpression of RUNX1 led to neurological abnormalities by affecting mitochondrial function in DS.

It is well known that the neuropathology of AD is an important clinical feature of DS, which is almost always present in patients with DS >40 years of age. Perluigi et al. (2014) demonstrated that the PI3K/Akt signaling pathway displayed increased activity in patients with DS and those with DS/AD. Notably, the PI3K/Akt signaling pathway plays a critical role in the regulation of cell proliferation, death, and metastasis (Lim et al., 2015), RUNX1 overexpression increased the phosphorylation levels of PI3K, Akt, and mTOR proteins (Liu et al., 2020).

Akt is a well-known pro-survival kinase and activated by phosphorylation at Ser 473 via the PI3K signaling pathway. Activated Akt plays a critical role in inhibiting neuronal death through further activation or inhibition of its downstream target proteins (Ye et al., 2015). In the present study, it was further found that overexpression of RUNX1 may activate the PI3K/Akt signaling pathway via increasing the phosphorylation of Akt, S6 and 4EBP1 in iPSCs. Certain upregulated genes in the PI3K-Akt signaling pathway, including Bim, IGF2, ITGA11, ITGB3, and CREB5, were considered to be closely involved in cell proliferation, apoptosis and tumor progression (Pardo et al., 2019; Shin et al., 2017; Wu et al., 2015; Yu et al., 2018). These genes were identified when RUNX1 was overexpressed in iPSCs. The present study also demonstrated that cell apoptosis was significantly increased in RUNX1-overexpressing iPSCs. Therefore, it was speculated that overexpression of RUNX1 in DS-iPSCs increased cell apoptosis by activating the PI3K-Akt signaling pathway.

Previous reports showed that Akt plays a central role in apoptosis inhibition through its regulatory effects on various downstream targets such as anti-apoptotic Bcl-2 (Yu et al., 2016). Normal mitochondrial function depends on the integrity of the mitochondrial membrane, which is strictly regulated by the Bcl-2 family of proteins (D'Orsi et al., 2017). The balance of cell death and survival signals in the Bcl-2 protein family determines the cell fate. As a pro-apoptotic member of the Bcl-2 family, Bim induces the release of cytochrome c from the mitochondria and activates the mitochondrial apoptotic pathway, which is implicated in the regulation of neuronal apoptosis (Concannon et al., 2010; Putcha et al., 2001). Bax is a pro-apoptotic protein, which can release cytochrome c from the mitochondria and induce changes in mitochondrial permeability, ultimately resulting in severe morphological changes in mitochondrial structure. Bcl-2 is an anti-apoptotic protein that regulates apoptosis by controlling the permeability of the mitochondrial membrane (Lv et al., 2018; Valiyari et al., 2017). As a result, the Bcl-2/Bax ratio can affect mitochondrial membrane permeability and is closely associated with the mitochondrial-mediated apoptotic pathway. In the present study, RUNX1 overexpression led to a significant increase in Bax expression and a significant decrease in Bcl-2 expression, thereby reducing the ratio of Bcl-2/Bax. Thus, abnormal expression of genes that belong to the Bcl-2 family, which was mediated by RUNX1 overexpression, may be an important factor for the increased apoptosis observed in DS-iPSCs. Based on the aforementioned results, it was concluded that RUNX1 could induce apoptosis by activating the mitochondrial apoptotic pathway in DS-iPSCs.

In conclusion, the present study demonstrated overexpression of RUNX1 in DS-iPSCs, and found that RUNX1 could regulate mitochondrial function and mitochondria-mediated apoptosis via the modulation of the PI3K-Akt signaling pathway in iPSCs. However, further investigation is required to assess whether RUNX1 promotes the neural differentiation of DS-iPSCs by regulating mitochondrial function.

This research was supported by the National Key Research and Development Program of China (2019YFA0801402) and the National Natural Science Foundation of China (81971421, 81471485), Shanghai key clinical specialty project (shslczdzk05705), Innovative Research Team of High-Level Local Universities in Shanghai (SHSMU-ZDCX20212200).

Y.L., J.Y., and F.Z. conceived and performed experiments, wrote the manuscript, and secured funding. Y.L. and Y.Z. performed experiments. J.Y., Z.R., and F.Z. provided expertise and feedback. All authors have read and agreed to the published version of the manuscript.

Fig. 1. Impaired mitochondrial function in DS-iPSCs. (A) ROS levels in iPSCs were measured using a ROS assay kit. DS-iPSCs and N-iPSCs were observed under bright light and fluorescence microscopy, respectively. (B) Fluorescence intensity of DS-iPSCs and N-iPSCs was analyzed using ImageJ. ***P < 0.001 versus control samples. (C) ATP content in DS-iPSCs and N-iPSCs. **P < 0.01 versus control samples. (D) The mitochondrial membrane potential was measured with the JC-1 dye. DS-iPSCs and N-iPSCs were observed using different wavelengths under a fluorescence microscope (514 nm for green fluorescence and 585 nm for red fluorescence). (E) The ratio of red fluorescence to green fluorescence intensity was analyzed using ImageJ software. ***P < 0.001 versus control samples. DS, Down syndrome; DS-iPSCs, induced pluripotent stem cells of patients with DS; ROS, reactive oxygen species; N-iPSCs, normal induced pluripotent stem cells.
Fig. 2. Overexpression of RUNX1 affects mitochondrial function in iPSCs. (A) The ROS levels of N-iPSCs transfected with RUNX1 expression vector (N-iPSCs + RUNX1) or control vector (N-iPSCs + Ctrl) were observed under bright light or fluorescence microscopy, respectively. (B) The fluorescence intensity of the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups was analyzed using ImageJ. *P < 0.05 versus control group. (C) Measurement of ATP content in the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups. **P < 0.01 versus control group. (D) The mitochondrial membrane potential was measured with the JC-1 dye, and the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups were imaged by fluorescence microscopy. (E) The ratio of red fluorescence to green fluorescence intensity was analyzed using ImageJ. *P < 0.05 versus control group. (F) The membrane ROS levels of iPSCs were measured using BODIPY-C11. The cell nuclei were stained with DAPI. The N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups were observed using different wavelengths under a fluorescence microscope (581 nm for red fluorescence and 340 nm for blue fluorescence). (G) The fluorescence intensity of the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups was analyzed using ImageJ. ***P < 0.001 versus control group. (H) The lysosomes of the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups were detected with LysoTrackerTM Red. The cells were imaged using different wavelengths (577 nm for red fluorescence and 340 nm for blue fluorescence) under a fluorescence microscope. (I) The relative fluorescence intensity of the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups was analyzed using ImageJ. **P < 0.01 versus control group. RUNX1, Runt-related transcription factor 1; iPSCs, induced pluripotent stem cells; ROS, reactive oxygen species; N-iPSCs, normal induced pluripotent stem cells; Ctrl, control.
Fig. 3. Inhibition of RUNX1 expression can improve the mitochondrial function of DS- iPSCs. (A) ROS levels were detected in DS-iPSCs transfected with RUNX1 shRNA vector (DS-iPSCs + shRUNX1) or control vector (DS-iPSCs + Ctrl). The results were imaged with bright light or fluorescence microscopy, respectively. (B) The fluorescence intensity of the DS-iPSCs + shRUNX1 and DS-iPSCs + Ctrl groups was analyzed using ImageJ. *P < 0.05 versus control group. (C) ATP content in the DS-iPSCs + shRUNX1 and DS-iPSCs + Ctrl groups. (D) The mitochondrial membrane potential was measured with the JC-1 dye, and the DS-iPSCs + shRUNX1 and DS-iPSCs + Ctrl groups were imaged by fluorescence microscopy. (E) The ratio of red fluorescence to green fluorescence intensity was analyzed using ImageJ software. *P < 0.05 versus control group. RUNX1, Runt-related transcription factor 1; DS, Down syndrome; DS-iPSCs; induced pluripotent stem cells of patients with DS; ROS, reactive oxygen species; shRNA, short hairpin RNA; Ctrl, control.
Fig. 4. Overexpression of RUNX1 in iPSCs leads to abnormalities in the PI3K-Akt signaling pathway. (A) Heatmap based on significantly differentially expressed genes between the N-iPSCs + RUNX1 (n = 3) and N-iPSCs + Ctrl groups (n = 3). (B) Volcano plot of 51 differentially expressed genes. The red dots indicate upregulated gene expression (log2 fold-change > 2), whereas the blue dots indicate downregulated gene expression (log2 fold-change ≤ 2), and the grey dots correspond to non-significant differences. The total number of downregulated genes was 12, while the total number of upregulated genes was 39. (C) Results of Kyoto Encyclopedia of Genes and Genomes pathway analysis. The scatter plot indicates the pathway category on the left-hand side of the plot. The red dots indicate significant pathways. The most impacted upregulated pathway was the PI3K-Akt signaling pathway. (D) The relative expression levels of certain differentially expressed genes involved in the PI3K-Akt signaling pathway were evaluated by RT-qPCR. N-iPSCs + Ctrl and N-iPSCs + RUNX1 were represented with black and grey colors, respectively. *P < 0.05, **P < 0.01 versus control group. RUNX1, Runt-related transcription factor; iPSCs; induced pluripotent stem cells; N-iPSCs, normal induced pluripotent stem cells; Ctrl, control; RT-qPCR, reverse transcription-quantitative polymerase chain reaction.
Fig. 5. Effect of overexpression of RUNX1 on the PI3K-Akt signaling pathway and cell apoptosis. (A) The expression levels and phosphorylation of Akt, S6 and 4E-binding protein were identified using western blotting in the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups. (B) Fluorescence activated cell sorting analysis was used to assess cell apoptosis, and the data were expressed as the percentage of annexin-V FITC-positive cells, including viable apoptotic cells (PE-positive cells, Q2) and non-viable apoptotic cells (PE-negative cells, Q3). (C) Apoptotic rate in the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups. **P < 0.01 versus control group. (D) The relative expression levels of certain apoptotic biomarkers were assessed by RT-qPCR. N-iPSCs + Ctrl and N-iPSCs + RUNX1 were represented with black and grey colors, respectively. *P < 0.05, **P < 0.01 versus control group. RUNX1, Runt-related transcription factor 1; N-iPSCs, normal induced pluripotent stem cells; Ctrl, control; PE, phycoerythrin; Q, quadrant; RT-qPCR, reverse transcription-quantitative polymerase chain reaction.
  1. Aburawi E.H. and Souid A.K. (2012). Lymphocyte respiration in children with Trisomy 21. BMC Pediatr. 12, 193.
    Pubmed KoreaMed CrossRef
  2. Alafnan A., Hussain T., Rizvi S.M.D., Moin A., and Alamri A. (2021). Prostate apoptotic induction and NFκB suppression by dammarolic acid: mechanistic insight into onco-therapeutic action of an aglycone asiaticoside. Curr. Issues Mol. Biol. 43, 932-940.
    Pubmed KoreaMed CrossRef
  3. Arrázola M.S., Andraini T., Szelechowski M., Mouledous L., Arnauné-Pelloquin L., Davezac N., Belenguer P., Rampon C., and Miquel M.C. (2019). Mitochondria in developmental and adult neurogenesis. Neurotox. Res. 36, 257-267.
    Pubmed CrossRef
  4. Beckervordersandforth R. (2017). Mitochondrial metabolism-mediated regulation of adult neurogenesis. Brain Plast. 3, 73-87.
    Pubmed KoreaMed CrossRef
  5. Bhatti J.S., Bhatti G.K., and Reddy P.H. (2017). Mitochondrial dysfunction and oxidative stress in metabolic disorders - a step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 1863, 1066-1077.
    Pubmed KoreaMed CrossRef
  6. Bourquin J.P., Subramanian A., Langebrake C., Reinhardt D., Bernard O., Ballerini P., Baruchel A., Cavé H., Dastugue N., and Hasle H., et al. (2006). Identification of distinct molecular phenotypes in acute megakaryoblastic leukemia by gene expression profiling. Proc. Natl. Acad. Sci. U. S. A. 103, 3339-3344.
    Pubmed KoreaMed CrossRef
  7. Concannon C.G., Tuffy L.P., Weisová P., Bonner H.P., Dávila D., Bonner C., Devocelle M.C., Strasser A., Ward M.W., and Prehn J.H. (2010). AMP kinase-mediated activation of the BH3-only protein Bim couples energy depletion to stress-induced apoptosis. J. Cell Biol. 189, 83-94.
    Pubmed KoreaMed CrossRef
  8. Coskun P.E. and Busciglio J. (2012). Oxidative stress and mitochondrial dysfunction in Down's syndrome: relevance to aging and dementia. Curr. Gerontol. Geriatr. Res. 2012, 383170.
    Pubmed KoreaMed CrossRef
  9. D'Orsi B., Mateyka J., and Prehn J.H.M. (2017). Control of mitochondrial physiology and cell death by the Bcl-2 family proteins Bax and Bok. Neurochem. Int. 109, 162-170.
    Pubmed CrossRef
  10. Delabar J.M., Theophile D., Rahmani Z., Chettouh Z., Blouin J.L., Prieur M., Noel B., and Sinet P.M. (1993). Molecular mapping of twenty-four features of Down syndrome on chromosome 21. Eur. J. Hum. Genet. 1, 114-124.
    Pubmed CrossRef
  11. Edwards H., Xie C., LaFiura K.M., Dombkowski A.A., Buck S.A., Boerner J.L., Taub J.W., Matherly L.H., and Ge Y. (2009). RUNX1 regulates phosphoinositide 3-kinase/AKT pathway: role in chemotherapy sensitivity in acute megakaryocytic leukemia. Blood 114, 2744-2752.
    Pubmed KoreaMed CrossRef
  12. El Hajj N., Dittrich M., Böck J., Kraus T.F., Nanda I., Müller T., Seidmann L., Tralau T., Galetzka D., and Schneider E., et al. (2016). Epigenetic dysregulation in the developing Down syndrome cortex. Epigenetics 11, 563-578.
    Pubmed KoreaMed CrossRef
  13. Flippo K.H. and Strack S. (2017). Mitochondrial dynamics in neuronal injury, development and plasticity. J. Cell Sci. 130, 671-681.
    Pubmed KoreaMed CrossRef
  14. Grabherr M.G., Haas B.J., Yassour M., Levin J.Z., Thompson D.A., Amit I., Adiconis X., Fan L., Raychowdhury R., and Zeng Q., et al. (2011). Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat. Biotechnol. 29, 644-652.
    Pubmed KoreaMed CrossRef
  15. Gutti U., Komati J.K., Kotipalli A., Saladi R.G.V., and Gutti R.K. (2018). Justicia adhatoda induces megakaryocyte differentiation through mitochondrial ROS generation. Phytomedicine 43, 135-139.
    Pubmed CrossRef
  16. Halevy T., Biancotti J.C., Yanuka O., Golan-Lev T., and Benvenisty N. (2016). Molecular characterization of Down syndrome embryonic stem cells reveals a role for RUNX1 in neural differentiation. Stem Cell Reports 7, 777-786.
    Pubmed KoreaMed CrossRef
  17. Hart J.R. and Vogt P.K. (2011). Phosphorylation of AKT: a mutational analysis. Oncotarget 2, 467-476.
    Pubmed KoreaMed CrossRef
  18. Hirata K., Nambara T., Kawatani K., Nawa N., Yoshimatsu H., Kusakabe H., Banno K., Nishimura K., Ohtaka M., and Nakanishi M., et al. (2020). 4-Phenylbutyrate ameliorates apoptotic neural cell death in Down syndrome by reducing protein aggregates. Sci. Rep. 10, 14047.
    Pubmed KoreaMed CrossRef
  19. Hu F. and Liu F. (2011). Mitochondrial stress: a bridge between mitochondrial dysfunction and metabolic diseases? Cell. Signal. 23, 1528-1533.
    Pubmed KoreaMed CrossRef
  20. Izzo A., Mollo N., Nitti M., Paladino S., Calì G., Genesio R., Bonfiglio F., Cicatiello R., Barbato M., and Sarnataro V., et al. (2018). Mitochondrial dysfunction in down syndrome: molecular mechanisms and therapeutic targets. Mol. Med. 24, 2.
    Pubmed KoreaMed CrossRef
  21. Jia L., Jiang Y., Li X., and Chen Z. (2020). Purβ promotes hepatic glucose production by increasing Adcy6 transcription. Mol. Metab. 31, 85-97.
    Pubmed KoreaMed CrossRef
  22. Johnson J., Mercado-Ayon E., Mercado-Ayon Y., Dong Y.N., Halawani S., Ngaba L., and Lynch D.R. (2021). Mitochondrial dysfunction in the development and progression of neurodegenerative diseases. Arch. Biochem. Biophys. 702, 108698.
    Pubmed CrossRef
  23. Kann O. and Kovács R. (2007). Mitochondria and neuronal activity. Am. J. Physiol. Cell Physiol. 292, C641-C657.
    Pubmed CrossRef
  24. Laufer B.I., Hwang H., Jianu J.M., Mordaunt C.E., Korf I.F., Hertz-Picciotto I., and LaSalle J.M. (2021). Low-pass whole genome bisulfite sequencing of neonatal dried blood spots identifies a role for RUNX1 in Down syndrome DNA methylation profiles. Hum. Mol. Genet. 29, 3465-3476.
    Pubmed KoreaMed CrossRef
  25. Li H., Liu J., Wang Y., Fu Z., Hüttemann M., Monks T.J., Chen A.F., and Wang J.M. (2017). MiR-27b augments bone marrow progenitor cell survival via suppressing the mitochondrial apoptotic pathway in Type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 313, E391-E401.
    Pubmed KoreaMed CrossRef
  26. Li X., Sun Y., Jin Q., Song D., and Diao Y. (2019). Kappa opioid receptor agonists improve postoperative cognitive dysfunction in rats via the JAK2/STAT3 signaling pathway. Int. J. Mol. Med. 44, 1866-1876.
    Pubmed KoreaMed CrossRef
  27. Li Z., Jiang T., Lu Q., Xu K., He J., Xie L., Chen Z., Zheng Z., Ye L., and Xu K., et al. (2020). Berberine attenuated the cytotoxicity induced by t-BHP via inhibiting oxidative stress and mitochondria dysfunction in PC-12 cells. Cell. Mol. Neurobiol. 40, 587-602.
    Pubmed CrossRef
  28. Lim H.J., Crowe P., and Yang J.L. (2015). Current clinical regulation of PI3K/PTEN/Akt/mTOR signalling in treatment of human cancer. J. Cancer Res. Clin. Oncol. 141, 671-689.
    Pubmed CrossRef
  29. Liu S., Zhang Y., Huang C., and Lin S. (2020). miR-215-5p is an anticancer gene in multiple myeloma by targeting RUNX1 and deactivating the PI3K/AKT/mTOR pathway. J. Cell. Biochem. 121, 1475-1490.
    Pubmed CrossRef
  30. Lv J., Liang Y., Tu Y., Chen J., and Xie Y. (2018). Hypoxic preconditioning reduces propofol-induced neuroapoptosis via regulation of Bcl-2 and Bax and downregulation of activated caspase-3 in the hippocampus of neonatal rats. Neurol. Res. 40, 767-773.
    Pubmed CrossRef
  31. McCubrey J.A., Steelman L.S., Chappell W.H., Abrams S.L., Montalto G., Cervello M., Nicoletti F., Fagone P., Malaponte G., and Mazzarino M.C., et al. (2012). Mutations and deregulation of Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascades which alter therapy response. Oncotarget 3, 954-987.
    Pubmed KoreaMed CrossRef
  32. Nishimura A., Hirabayashi S., Hasegawa D., Yoshida K., Shiraishi Y., Ashiarai M., Hosoya Y., Fujiwara T., Harigae H., and Miyano S., et al. (2021). Acquisition of monosomy 7 and a RUNX1 mutation in Pearson syndrome. Pediatr. Blood Cancer 68, e28799.
    Pubmed CrossRef
  33. Ohnishi T., Ohnishi S.T., and Salerno J.C. (2018). Five decades of research on mitochondrial NADH-quinone oxidoreductase (complex I). Biol. Chem. 399, 1249-1264.
    Pubmed CrossRef
  34. Pardo M., Cheng Y., Sitbon Y.H., Lowell J.A., Grieco S.F., Worthen R.J., Desse S., and Barreda-Diaz A. (2019). Insulin growth factor 2 (IGF2) as an emergent target in psychiatric and neurological disorders. Review. Neurosci. Res. 149, 1-13.
    Pubmed CrossRef
  35. Patel A., Rees S.D., Kelly M.A., Bain S.C., Barnett A.H., Thalitaya D., and Prasher V.P. (2011). Association of variants within APOE, SORL1, RUNX1, BACE1 and ALDH18A1 with dementia in Alzheimer's disease in subjects with Down syndrome. Neurosci. Lett. 487, 144-148.
    Pubmed CrossRef
  36. Pecze L. and Szabo C. (2021). Meta-analysis of gene expression patterns in Down syndrome highlights significant alterations in mitochondrial and bioenergetic pathways. Mitochondrion 57, 163-172.
    Pubmed CrossRef
  37. Pelleri M.C., Cicchini E., Locatelli C., Vitale L., Caracausi M., Piovesan A., Rocca A., Po-letti G., Seri M., and Strippoli P., et al. (2016). Systematic reanalysis of partial trisomy 21 cases with or without Down syndrome suggests a small region on 21q22.13 as critical to the phenotype. Hum. Mol. Genet. 25, 2525-2538.
    Pubmed KoreaMed CrossRef
  38. Perluigi M., Pupo G., Tramutola A., Cini C., Coccia R., Barone E., Head E., Butterfield D.A., and Di Domenico F. (2014). Neuropathological role of PI3K/Akt/mTOR axis in Down syndrome brain. Biochim. Biophys. Acta 1842, 1144-1153.
    Pubmed KoreaMed CrossRef
  39. Putcha G.V., Moulder K.L., Golden J.P., Bouillet P., Adams J.A., Strasser A., and Johnson E.M. (2001). Induction of BIM, a proapoptotic BH3-only BCL-2 family member, is critical for neuronal apoptosis. Neuron 29, 615-628.
    Pubmed CrossRef
  40. Qiu J.J., Liu Y.N., Ren Z.R., and Yan J.B. (2017). Dysfunctions of mitochondria in close association with strong perturbation of long noncoding RNAs expression in down syndrome. Int. J. Biochem. Cell Biol. 92, 115-120.
    Pubmed CrossRef
  41. Salemi M., Giambirtone M., Barone C., Salluzzo M.G., Russo R., Giudice M.L., Cutuli S., Ridolfo F., and Romano C. (2018). Mitochondrial mRNA expression in fibroblasts of Down syndrome subjects. Hum. Cell 31, 179-181.
    Pubmed CrossRef
  42. Salemi M., Ridolfo F., Salluzzo M.G., Cannarrella R., Giambirtone M., Caniglia S., Tirolo C., Ferri R., and Romano C. (2020). Humanin gene expression in fibroblast of Down syndrome subjects. Int. J. Med. Sci. 17, 320-324.
    Pubmed KoreaMed CrossRef
  43. Shah S.I., Paine J.G., Perez C., and Ullah G. (2019). Mitochondrial fragmentation and network architecture in degenerative diseases. PLoS One 14, e0223014.
    Pubmed KoreaMed CrossRef
  44. Shin J., Shim H.G., Hwang T., Kim H., Kang S.H., Dho Y.S., Park S.H., Kim S.J., and Park C.K. (2017). Restoration of miR-29b exerts anti-cancer effects on glioblastoma. Cancer Cell Int. 17, 104.
    Pubmed KoreaMed CrossRef
  45. Vacca R.A., Bawari S., Valenti D., Tewari D., Nabavi S.F., Shirooie S., Sah A.N., Volpicella M., Braidy N., and Nabavi S.M. (2019). Down syndrome: neurobiological alterations and therapeutic targets. Neurosci. Biobehav. Rev. 98, 234-255.
    Pubmed CrossRef
  46. Valenti D., Braidy N., De Rasmo D., Signorile A., Rossi L., Atanasov A.G., Volpicella M., Henrion-Caude A., Nabavi S.M., and Vacca R.A. (2018). Mitochondria as pharmacological targets in Down syndrome. Free Radic. Biol. Med. 114, 69-83.
    Pubmed CrossRef
  47. Valiyari S., Salami M., Mahdian R., Shokrgozar M.A., Oloomi M., Mohammadi Farsani A., and Bouzari S. (2017). sIL-24 peptide, a human interleukin-24 isoform, induces mitochondrial-mediated apoptosis in human cancer cells. Cancer Chemother. Pharmacol. 80, 451-459.
    Pubmed CrossRef
  48. Wong R.S. (2011). Apoptosis in cancer: from pathogenesis to treatment. J. Exp. Clin. Cancer Res. 30, 87.
    Pubmed KoreaMed CrossRef
  49. Wu J., Zheng C., Wang X., Yun S., Zhao Y., Liu L., Lu Y., Ye Y., Zhu X., and Zhang C., et al. (2015). MicroRNA-30 family members regulate calcium/calcineurin signaling in podocytes. J. Clin. Invest. 125, 4091-4106.
    Pubmed KoreaMed CrossRef
  50. Ye Z., Xia P., Cheng Z.G., and Guo Q. (2015). Neuroprotection induced by sevoflurane-delayed post-conditioning is attributable to increased phosphorylation of mitochondrial GSK-3β through the PI3K/Akt survival pathway. J. Neurol. Sci. 348, 216-225.
    Pubmed CrossRef
  51. Yoshikawa M., Hirabayashi M., Ito R., Ozaki S., Aizawa S., Masuda T., Senzaki K., and Shiga T. (2015). Contribution of the Runx1 transcription factor to axonal pathfinding and muscle innervation by hypoglossal motoneurons. Dev. Neurobiol. 75, 1295-1314.
    Pubmed CrossRef
  52. Yoshikawa M., Masuda T., Kobayashi A., Senzaki K., Ozaki S., Aizawa S., and Shiga T. (2016). Runx1 contributes to the functional switching of bone morphogenetic protein 4 (BMP4) from neurite outgrowth promoting to suppressing in dorsal root ganglion. Mol. Cell. Neurosci. 72, 114-122.
    Pubmed CrossRef
  53. Yu G., Yin C., Jiang L., Zheng Z., Wang Z., Wang C., Zhou H., Jiang X., Liu Q., and Meng F. (2016). Amyloid precursor protein cooperates with c-KIT mutation/overexpression to regulate cell apoptosis in AML1-ETO-positive leukemia via the PI3K/AKT signaling pathway. Oncol. Rep. 36, 1626-1632.
    Pubmed CrossRef
  54. Yu M., Du G., Xu Q., Huang Z., Huang X., Qin Y., Han L., Fan Y., Zhang Y., and Han X., et al. (2018). Integrated analysis of DNA methylome and transcriptome identified CREB5 as a novel risk gene contributing to recurrent pregnancy loss. EBioMedicine 35, 334-344.
    Pubmed KoreaMed CrossRef
  55. Yuan Y., Chen Y., Peng T., Li L., Zhu W., Liu F., Liu S., An X., Luo R., and Cheng J., et al. (2019). Mitochondrial ROS-induced lysosomal dysfunction impairs autophagic flux and contributes to M1 macrophage polarization in a diabetic condition. Clin. Sci. (Lond.) 133, 1759-1777.
    Pubmed CrossRef
  56. Zamponi E. and Helguera P.R. (2019). The shape of mitochondrial dysfunction in Down syndrome. Dev. Neurobiol. 79, 613-621.
    Pubmed CrossRef
  57. Zhang W., Hu X., Shen Q., and Xing D. (2019). Mitochondria-specific drug release and reactive oxygen species burst induced by polyprodrug nanoreactors can enhance chemotherapy. Nat. Commun. 10, 1704.
    Pubmed KoreaMed CrossRef
  58. Zhao X., Song X., Bai X., Fei N., Huang Y., Zhao Z., Du Q., Zhang H., Zhang L., and Tong D. (2016). miR-27b attenuates apoptosis induced by transmissible gastroenteritis virus (TGEV) infection via targeting runt-related transcription factor 1 (RUNX1). PeerJ 4, e1635.
    Pubmed KoreaMed CrossRef

Article

Research Article

Mol. Cells 2023; 46(4): 219-230

Published online April 30, 2023 https://doi.org/10.14348/molcells.2023.2095

Copyright © The Korean Society for Molecular and Cellular Biology.

RUNX1 Upregulation Causes Mitochondrial Dysfunction via Regulating the PI3K-Akt Pathway in iPSC from Patients with Down Syndrome

Yanna Liu1,4 , Yuehua Zhang1,4 , Zhaorui Ren1,2 , Fanyi Zeng1,2,3,* , and Jingbin Yan1,2,*

1Shanghai Children’s Hospital, Shanghai Institute of Medical Genetics, Shanghai Jiao Tong University School of Medicine, Shanghai 200040, China, 2NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200040, China, 3Department of Histoembryology, Genetics & Development, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, 4These authors contributed equally to this work.

Correspondence to:yanjb@shchildren.com.cn (JY); fzeng@vip.163.com (FZ)

Received: June 6, 2022; Revised: September 30, 2022; Accepted: October 3, 2022

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/.

Abstract

Down syndrome (DS) is the most common autosomal aneuploidy caused by trisomy of chromosome 21. Previous studies demonstrated that DS affected mitochondrial functions, which may be associated with the abnormal development of the nervous system in patients with DS. Runt-related transcription factor 1 (RUNX1) is an encoding gene located on chromosome 21. It has been reported that RUNX1 may affect cell apoptosis via the mitochondrial pathway. The present study investigated whether RUNX1 plays a critical role in mitochondrial dysfunction in DS and explored the mechanism by which RUNX1 affects mitochondrial functions. Expression of RUNX1 was detected in induced pluripotent stem cells of patients with DS (DS-iPSCs) and normal iPSCs (N-iPSCs), and the mitochondrial functions were investigated in the current study. Subsequently, RUNX1 was overexpressed in N-iPSCs and inhibited in DS-iPSCs. The mitochondrial functions were investigated thoroughly, including reactive oxygen species levels, mitochondrial membrane potential, ATP content and lysosomal activity. Finally, RNA-sequencing was used to explore the global expression pattern. It was observed that the expression levels of RUNX1 in DS-iPSCs were significantly higher than those in normal controls. Impaired mitochondrial functions were observed in DS-iPSCs. Of note, overexpression of RUNX1 in N-iPSCs resulted in mitochondrial dysfunction, while inhibition of RUNX1 expression could improve the mitochondrial function in DS-iPSCs. Global gene expression analysis indicated that overexpression of RUNX1 may promote the induction of apoptosis in DS-iPSCs by activating the PI3K/Akt signaling pathway. The present findings indicate that abnormal expression of RUNX1 may play a critical role in mitochondrial dysfunction in DS-iPSCs.

Keywords: Down syndrome, induced pluripotent stem cells, mitochondrial dysfunction, RUNX1

INTRODUCTION

Down syndrome (DS) or trisomy 21 is one of the most common genetic diseases and is often accompanied by intellectual disability. It occurs with an incidence ~1 in 700-1,000 births (El Hajj et al., 2016). DS is a major cause of congenital malformations and mental retardation in humans. Patients with DS typically display associated dysfunctions and a range of cognitive deficits, with important differences in their phenotype and severe side effects in the nervous system (Vacca et al., 2019). Previous studies on the brains of patients with DS have revealed abnormalities in the structure and function of the central nervous system. Recent studies have shown that alterations in mitochondrial function may be associated with the occurrence of DS (Pecze and Szabo, 2021; Zamponi and Helguera, 2019).

Mitochondria play a central role in metabolism and energy conversion in eukaryotic cells, which generate ATP via oxidative phosphorylation (OXPHOS). These organelles provide energy for various cell-based activities (Ohnishi et al., 2018). Dysregulated mitochondria lead to the excessive production of reactive oxygen species (ROS), which are involved in the regulation of programmed cell death (Li et al., 2019; Wong, 2011). Impaired mitochondrial function leads to significant changes in the cell and affects neural cell proliferation, apoptosis, differentiation, regeneration, and other cellular activities (Beckervordersandforth, 2017; Johnson et al., 2021). It is known that mitochondrial dysfunction/oxidative stress is associated with DS. Mitochondrial dysfunction has been observed in the brain of early fetuses with DS and can manifest as abnormalities in OXPHOS complexes (Coskun and Busciglio, 2012; Salemi et al., 2018), accumulation of oxidative stress products and changes in mitochondrial biosynthesis. In addition, previous studies demonstrated that abnormal expression of genes was associated with mitochondrial function in DS (Qiu et al., 2017; Salemi et al., 2020). Mitochondrial dysfunction leads to impaired neuron proliferation, differentiation, and maturation. Previous studies have shown the presence of mitochondrial dysfunction in neurons, glial cells, and peripheral blood cells in patients with DS, and they were associated with mitochondrial fragmentation, impaired OXPHOS, and reduced ATP levels (Valenti et al., 2018; Zamponi and Helguera, 2019). Collectively, these studies indicated that mitochondrial dysfunction was strongly associated with abnormal development of the nervous system in DS. However, the key genes and molecular mechanisms that lead to mitochondrial dysfunction in patients with DS remain unknown.

DS is caused by an extra copy of the human chromosome 21 (Hsa21). Previous studies have proposed the gene dosage effect hypothesis, suggesting that increased expression of a specific set of dosage-sensitive genes on Hsa21 may eventually cause neurodevelopmental or neurocognitive disorders in DS (Delabar et al., 1993). A recent study indicated that trisomy 21 caused neuronal apoptosis, leading to a decrease in the neural population (Hirata et al., 2020). The DS critical region (DSCR), namely 21q22-21q23, is a genomic region on Hsa21 that is strongly linked to DS phenotypes (Pelleri et al., 2016). Aberrant expression of genes, notably transcription factors located in the DSCR, may be associated with mitochondrial dysfunction in DS (Flippo and Strack, 2017; Izzo et al., 2018).

Runt-related transcription factor 1 (RUNX1), which is encoded by the RUNX1 gene, is localized in chromosome 21q22.12. This transcription factor is an important transcriptional regulator found in the DSCR. It is known that RUNX1 is an essential master regulator, which is active during hematopoietic development. In recent years, an increasing number of studies have found that RUNX1 plays an essential role in the development of the central nervous system, the proliferation and differentiation of neural progenitor cells, neuronal differentiation and control of axon growth (Halevy et al., 2016; Yoshikawa et al., 2015; 2016). Gutti et al. (2018) demonstrated that Justicia adhatoda could upregulate the expression levels of RUNX1 and enhance mitochondrial ROS generation in human megakaryocytic cells. Excessive ROS production can damage mitochondrial proteins, which ultimately results in mitochondrial dysfunction (Zhang et al., 2019). Several studies have reported that microRNA (miR)-27b may repress the mitochondrial pathway of apoptosis by targeting RUNX1 (Li et al., 2017; Zhao et al., 2016). However, it is unknown whether RUNX1 plays a critical role in the mitochondrial dysfunction observed in DS. Therefore, the present study investigated the role of RUNX1 in the development of mitochondrial dysfunction in DS and its ability to affect mitochondrial functions.

MATERIALS AND METHODS

Human pluripotent stem cell culture and treatments

Normal induced pluripotent stem cells (N-iPSCs) and induced pluripotent stem cells of patients with DS (DS-iPSCs) were purchased from the American Type Culture Collection (Cat. No. ATCC DYR0100 [normal, newborn, male] and Cat. No. ATCC DYP0730 [DS, newborn, male]; ATCC, USA). The clones were cultured in a feeder-free system. The cell culture dishes were coated with Cell Matrix Basement Membrane Gel (Cat. No. ACS-3035; ATCC) and cultured in Complete Pluripotent Stem Cell SFM XF/FF medium (Cat. No. ACS-3002; ATCC) in the presence of 10 µM inhibitor (Cat. No. Y0503; Sigma, USA). The culture medium was changed the day after cell resuscitation, and daily thereafter until the colonies reached 80% confluence. The cells were typically digested, sub-cultured at a 1:3 ratio using the Stem Cell Dissociation Reagent (Cat. No. ACS-3010; ATCC) and incubated overnight at 37°C in the presence of 5% CO2.

DS-iPSCs were then cultured in the medium supplemented with 20 µM LY294002 (Cat. No. S1105; Selleck, USA) for 1 h. Then the cells were collected, the expression level of Akt was identified by western blotting and the analysis of ROS level and cellular ATP content was also performed. The experiment was repeated in triplicate.

Plasmid vectors

The following vectors were purchased from AddGene (USA); RUNX1 expression vector (pCMV5-AML1B; plasmid Cat. No. 12426) and RUNX1 short hairpin RNA (shRNA) vector (pLKO.1 shRUNX1 puro; plasmid Cat. No. 45816).

Electroporation of iPSCs

Single clones were harvested from a 60-mm dish using Stem Cell Dissociation Reagent within 10 min and washed once with 2 ml phosphate-buffered saline (PBS). Subsequently, the cells were resuspended in R-buffer. N-iPSCs and DS-iPSCs were transfected with the Neon Transfection System (Cat. No. MPK1025; Invitrogen, USA) according to the manufacturer’s instructions. The conditions used were as follows: 1,240 V, 20 ms and 2-times pulses. A total volume of 10 µl containing 1 µg plasmid DNA was used. Following transfection, the samples were placed on a 24-well microplate, and subsequent analysis was performed 48 h after transfection.

Identification of the expression level of RUNX1 by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting (WB)

Total RNA was extracted from PBMCs and iPSCs using TRIzol® reagent. RT was performed using a reverse transcription kit (Cat. No. 18091050; Invitrogen). RT-qPCR was performed with TaqmanTM Real-Time PCR Assay. The primer sequences for RUNX1 and GAPDH were as follows: RUNX1, forward: 5’-TGGCACTCTGGTCACCGTCAT-3’ and reverse: 5’-GAAGCTCTTGCCTCTACCGC-3’, and GAPDH, forward: 5’-AGAGGGCTGTCGGCGCAGTA-3’ and reverse: 5’-GGCTGTGGTCTCGGTTGGGC-3’. The reactions were performed using an ABI7500 Real-Time PCR system (Thermo Fisher Scientific, USA). The transcript levels of RUNX1 were quantified using the 2-ΔΔCq method, with GAPDH as the reference gene used for normalization.

The iPSCs were scraped off in ice-cold PBS, collected into RIPA lysis buffer (Cat. No. sc-24948; Santa Cruz Biotechnology, USA) with a cell scraper, and incubated on ice for 30 min. The cell lysates were centrifuged at 14,000 × g for 15 min at 4°C, and the supernatants were then collected. The protein concentration was determined by BCA assay (Cat. No. P0010; Beyotime, China). Protein denaturation was performed at 98°C for 10 min. Lysed proteins (20 µg/lane) were separated by 10% SDS-PAGE and transferred onto nitrocellulose membranes, which were then blocked with 5% non-fat milk in TBS containing 0.05% Tween-20 (TBST) for 1 h at room temperature. Subsequently, the membranes were incubated at 4°C overnight with primary antibodies against RUNX1 (1:1,000, Cat. No. ab272456; Abcam) and GAPDH (1:5,000, Cat. No. 10494-1-AP; ProteinTech Group, USA). GAPDH was used as a loading control for normalization. Subsequently, the membranes were incubated with a secondary antibody (1:2,000, Cat. No. SA00001-2; ProteinTech Group) at room temperature for 1 h. The membranes were washed three times with TBST and visualized by chemiluminescence in an Amersham Imager 600 (GE Healthcare, USA).

Analysis of ROS levels using 2’-7’dichlorofluorescein diacetate (DCFH-DA)

ROS levels in iPSCs were measured using a ROS Assay Kit (Cat. No. S0033; Beyotime), following the manufacturer’s instructions. Briefly, iPSCs were cultured in 6-well culture plates pretreated with Matrigel, and subsequently, the cell culture medium was removed. The cells were then incubated with 10 µM DCFH-DA, which is a fluorescent probe used for ROS detection. Following subsequent culture at 37°C for 20 min in the dark, the iPSCs were washed three times with serum-free Dulbecco’s modified Eagle’s medium (DMEM), and the fluorescence emission of the samples was detected by fluorescence microscopy (Nikon Eclipse Ti; Nikon, Japan). The fluorescence intensity was analyzed using ImageJ software (ver. No. 1.6; National Institutes of Health, USA).

Lipid peroxidation measurement

BODIPY-C11 staining was performed according to the manufacturer’s protocol. The cells were seeded in a 35-mm-diameter dish. Culture medium was replaced with 2 ml medium containing 5 µM of BODIPY-C11 (Cat. No. D3861; Invitrogen) after 48 h and the culture was returned to the cell culture incubator for 30 min. Finally, the cells were fixed with 4% paraformaldehyde for 15 min and cell nuclei were stained with DAPI for 10 min. Fluorescence intensity was examined by fluorescence microscopy (Nikon Eclipse Ti), and images were analyzed with the ImageJ software.

Detection of mitochondrial membrane potential (MMP) with JC-1

Cells were seeded in culture flasks, and when they were 60%-70% confluent, the culture solution was aspirated. The cells were then washed once with PBS, and 2 ml cell culture medium containing 1 ml JC-1 solution (Cat. No. S2003S; Beyotime) was added for staining. The solution was thoroughly mixed in a cell culture incubator for 20 min at 37°C. Concomitantly, the JC-1 staining buffer (5×) was diluted five times in water (4 ml distilled water and 1 ml JC-1 staining buffer). The samples were placed on ice. Following incubation at 37°C, the supernatant was aspirated and washed with JC-1 buffer (1×) three times. Upon addition of 2 ml medium, the fluorescence emission of the samples was detected using a fluorescence microscope. The experiments were repeated three times.

Measurement of cellular ATP content

Cellular ATP content was determined with an ATP Assay Kit according to the manufacturer’s instructions (Cat. No. S0026; Beyotime). Briefly, the cells were lysed with ATP Detection Lysis Buffer 48 h after transfection, and 100 µl ATP detection working solution was added to the wells for 5 min at room temperature. A total of 20 µl sample or standard was added to each well, and the ATP concentration was measured using luminometry. A BCA Protein Assay Kit (Cat. No. P0010; Beyotime) was used to determine the protein concentrations of each sample, and the total ATP levels were evaluated as the ratio of cellular ATP level to protein concentration.

Lysosome detection using LysoTrackerTM Red

Following transfection of iPSCs with RUNX1-overexpressing plasmids for 48 h, the transfected cells cultured on 35-mm culture dishes were incubated with 100 nM lysosome probes (Cat. No. M7512; Invitrogen) at 37°C for 20 min. Following incubation, the cells were fixed with 4% paraformaldehyde for 15 min and 0.2% Triton X was used for permeabilization for 10 min. This was followed by DAPI staining (Cat. No. C1002; Beyotime) for 8 min to visualize the nuclei. Red fluorescence was determined by fluorescence microscopy, and image analysis was performed with ImageJ software.

Cell apoptosis assay

Cell apoptosis was evaluated with a Cell Apoptosis Kit (Cat. No. APOAF; Sigma), according to the manufacturer’s instructions. Briefly, iPSCs were collected 24 h following transfection of RUNX1-overexpressing plasmids. Subsequently, 500 µl binding buffer, 5 µl Annexin V-FITC conjugate and 10 µl propidium iodide solution were added successively into the cell suspension, and incubated at room temperature in the dark for 10 min. The fluorescence emission of the cells was immediately detected with a flow cytometer (BD AccuriTM C6 Flow Cytometer; BD Biosciences, USA).

RNA-sequencing (RNA-seq) and differential expression analysis

RNA-seq was conducted with the NovaSeq 6000 platform (Illumina, USA). An experimental group transfected with a RUNX1 expression vector and a control group transfected with a GFP expression vector were used. Each group had three replicates.

Following total RNA extraction using TRIzol® reagent (Cat. No. 15596018; Invitrogen), the quantification, qualification, library preparation, and subsequent RNA-seq of the above two groups were performed by Novogene (China). The details of RNA-seq have been previously described (Jia et al., 2020). Raw data were cleaned by removing reads with adaptors and excluding low quality (>50%) or a high proportion of unknown bases (>10%). Subsequently, the data were assembled using Trinity methodology (Grabherr et al., 2011).

For each sequenced library, the read counts were adjusted using the edgeR program package through one scaling normalized factor prior to differential gene expression analysis. Differential expression analysis of two conditions was performed using the edgeR R package. P values were adjusted using the Benjamin and Hochberg method. Corrected P = 0.05 (Q-value) and absolute fold-change = 2 was set as the thresholds for significant differential expression. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was conducted, and the corrected P value cutoff was set at 0.05.

Differential expression genes and apoptotic biomarkers were confirmed by RT-qPCR. The primers are listed in Supplementary Table S1. To detect activation of the PI3K-Akt signaling pathway, the protein expression and phosphorylation levels of Akt, S6, and 4E-binding protein (4E-BP1) were detected by western blotting as previously described. The antibodies used are listed in Supplementary Table S2.

RESULTS

Impaired mitochondrial function is observed in DS-iPSCs

Our previous study demonstrated that the expression levels of almost all the genes related to mitochondrial function were downregulated in DS-iPSCs, suggesting that mitochondria were dysfunctional in these cells (Qiu et al., 2017). To further demonstrate the impairment of mitochondrial function in DS-iPSCs, specific changes in mitochondrial MMP, ROS and ATP content were investigated to provide further evidence of impaired mitochondrial function. As shown in Figs. 1A and 1B, a, a high green fluorescence intensity was noted in the DS-iPSCs. The quantification of green fluorescence intensity was analyzed by ImageJ (Alafnan et al., 2021), and the results suggested that the contents of ROS were increased 2.24-fold in DS-iPSCs (0.076 ± 0.004 vs 0.034 ± 0.003; P < 0.0001). The ATP contents in DS-iPSCs were 1.68-fold lower than those in N-iPSCs (3.95 ± 0.39 vs 6.64 ± 0.17; P = 0.0029) (Fig. 1C), indicating that energy production by mitochondria was impaired in DS. JC-1 staining was also used to determine whether the MMP was impaired in DS-iPSCs. The results indicated that higher green fluorescence signals and weaker red fluorescence signals were present in DS-iPSCs, resulting in an attenuated ratio of red/green fluorescence intensity (3.12 ± 0.38 vs 7.05 ± 0.38; P < 0.0001), indicating that there was loss of MMP in DS-iPSCs (Figs. 1D and 1E). The aforementioned results indicated that the loss of mitochondrial function in DS-iPSCs could lead to damage of MMP, increased ROS levels and reduced ATP synthesis.

Overexpression of RUNX1 affects mitochondrial function in iPSCs

Previous studies have shown that RUNX1 is involved in the pathogenesis of DS (Bourquin et al., 2006; Edwards et al., 2009; Patel et al., 2011). Furthermore, overexpression of RUNX1 was detected in DS-iPSCs and peripheral blood of children with DS (Supplementary Fig. S1), leading to the speculation that RUNX1 may be a key gene regulating mitochondrial function in DS. To determine whether RUNX1 affects mitochondrial function, RUNX1 was overexpressed in N-iPSCs (Supplementary Fig. S2). The results indicated that mitochondrial dysfunction was present in N-iPSCs following RUNX1 overexpression, which was similar to that in DS-iPSCs. JC-1 staining indicated that the green fluorescence intensity was considerably enhanced in RUNX1-overexpressing N-iPSCs compared with that observed in N-iPSCs expressing normal levels of RUNX1 (0.090 ± 0.017 vs 0.037 ± 0.0039; P = 0.035). These data suggested that RUNX1 promoted the production of ROS in iPSCs (Figs. 2A and 2B).

Accumulation of ROS in mitochondria causes abnormalities in mitochondrial structure and function, including ATP synthesis obstruction, reduced mitochondrial MMP, and impaired respiratory chain function. Mitochondrial respiratory chain deficiencies block OXPHOS and inhibit ROS production, which aggravates mitochondrial dysfunction and creates a vicious cycle of mitochondrial damage (Bhatti et al., 2017; Hu and Liu, 2011). Thus, mitochondrial function was evaluated in iPSCs overexpressing RUNX1. The results indicated that overexpression of RUNX1 led to a 3.17-fold decrease in ATP content compared with that of control cells (4.75 ± 0.36 vs 2.11 ± 0.26; P = 0.0043) (Fig. 2C). JC-1 indicated that overexpression of RUNX1 increased the intensity of green fluorescence in RUNX1-overexpressing N-iPSCs, while the reduction in the ratio of red to green fluorescence (2.92 ± 0.38 vs 1.44 ± 0.50; P = 0.015) indicated a loss of MMP (Figs. 2D and 2E). These data suggested that RUNX1 overexpression resulted in the accumulation of ROS levels, which ultimately caused mitochondrial dysfunction in DS-iPSCs.

High levels of mitochondrial ROS can induce oxidative damage in mitochondria, which impairs the function of the mitochondrial respiratory chain, leading to the accumulation of intracellular ROS. The membrane ROS levels of iPSCs were determined using BODIPYTM C11, and the data confirmed that RUNX1 overexpression increased mitochondrial and cell membrane ROS levels compared with those of control cells (0.10 ± 0.0034 vs 0.077 ± 0.0015; P < 0.0001) (Figs. 2F and 2G). Mitochondrial dysfunction was previously reported to promote ROS production and impair lysosome function in macrophages cultured under hyperglycemic conditions (Yuan et al., 2019). The present study experimentally confirmed that a high ROS content was present in DS-iPSCs with high expression of RUNX1. The inhibition of lysosomal activity was measured using LysoTrackerTM Red probe, and the data revealed that the number of active lysosomes was reduced in iPSCs overexpressing RUNX1 (0.75 ± 0.054 vs 0.43 ± 0.048; P = 0.0014) (Figs. 2H and 2I). Therefore, it was concluded that overexpression of RUNX1 may disrupt the normal function of mitochondria, thereby affecting the cell biological function of DS-iPSCs.

Inhibition of RUNX1 expression can improve the mitochondrial function of DS- iPSCs

To further determine whether RUNX1 overexpression could affect mitochondrial function, DS-iPSCs were transfected with RUNX1 shRNA vector, and the mitochondrial function was measured. The results indicated that the green fluorescence intensity was markedly decreased when the expression levels of RUNX1 were inhibited in DS-iPSCs (0.068 ± 0.015 vs 0.029 ± 0.0020; P = 0.017) compared with that exhibited by normal controls, indicating that the increase in ROS content in DS-iPSCs was closely associated with high expression levels of RUNX1 (Figs. 3A and 3B). In addition, ATP content was only slightly increased in the DS-iPSC + shRUNX1 group compared with that in control cells (5.19 ± 0.58 vs 6.86 ± 0.52; P = 0.10) (Fig. 3C). Furthermore, overexpression of RUNX1 also increased the green fluorescence intensity in the cells and the reduction in the ratio of red to green fluorescence (2.26 ± 0.18 vs 2.82 ± 0.11; P = 0.020) (Figs. 3D and 3E), which indicated that the MMP of DS-iPSCs was significantly recovered when the expression levels of RUNX1 were inhibited.

Overexpression of RUNX1 in iPSCs leads to abnormalities in the PI3K-Akt signaling pathway

RNA-seq analysis indicated that 51 differentially expressed genes were identified between the experimental and control groups using the edgeR R package (Fig. 4A). This included 39 upregulated and 12 downregulated genes (Fig. 4B). KEGG analysis revealed a set of 39 upregulated genes, and the results indicated that PI3K-Akt was identified as the most significant signaling pathway, in which five genes (IGF2, ITGA11, ITGB3, CREB5, and Bim) were involved (Fig. 4C). The RT-qPCR results confirmed that the expression levels of these genes were markedly increased in RUNX1-overexpressing N-iPSCs (Fig. 4D).

A previous study has shown that the activation of mTOR signaling via the PI3K/Akt signaling pathway is involved in the regulation of mitochondrial ROS production in PC-12 cells, which can ultimately affect cell apoptosis (Li et al., 2020). In the present study, PI3K/Akt inhibitor LY294002 was used to suppress the expression of Akt in DS-iPSCs. The results showed that the expression level of Akt in DS-iPSCs was significantly decreased, accompanied by a significant increase in ATP content and a decrease in ROS level (Supplementary Fig. S3).

Akt is a major downstream effector molecule of the PI3K/Akt signaling pathway, and increased phosphorylation implies activation of the PI3K signaling pathway (Hart and Vogt, 2011; McCubrey et al., 2012). The present results revealed a significant increase in the phosphorylation of Akt, S6 and 4EBP1 in N-iPSCs + RUNX1 cells (Fig. 5A). These results indicated that upregulation of RUNX1 in iPSCs activated the PI3K-Akt signaling pathway by increasing the phosphorylation of Akt, S6 and 4EBP1.

The effects of RUNX1 overexpression on the induction of apoptosis of iPSCs were investigated by flow cytometry. A significant increase in apoptosis was observed in RUNX1-overexpressing N-iPSCs (19.00 ± 1.79 vs 32.46 ± 1.54; P = 0.0047) (Figs. 5B and 5C). Furthermore, the results of expression analysis of apoptotic biomarkers (Bax, Bcl-2, and caspase 3) further supported the hypothesis that overexpression of RUNX1 may promote apoptosis in DS-iPSCs by activating the PI3K/Akt signaling pathway (Fig. 5D).

DISCUSSION

The most apparent clinical features of DS are neurological complications, including early onset of Alzheimer’s disease (AD) and mental retardation. The central nervous system requires high quantities of energy. Redox homeostasis and adequate ATP levels are essential for normal neurodevelopment (Arrázola et al., 2019; Kann and Kovács, 2007). Mitochondria supply virtually all eukaryotic cells with energy through ATP production by OXPHOS. Accordingly, maintenance of the mitochondrial function is fundamentally important to sustain cellular health. Previous studies reported that mitochondrial abnormalities, including aberrant mitochondrial morphology and function, were detected in both patients with DS and in DS cells (Aburawi and Souid, 2012; Shah et al., 2019). In the present study, mitochondrial abnormalities were confirmed in DS-iPSCs. Therefore, it is reasonable to speculate that the aberrant development of the nervous system in DS is closely associated with mitochondrial dysfunction, as iPSCs have the pluripotency to differentiate into various nerve cell types. However, the mechanism leading to mitochondrial dysfunction in DS is still not clear, and the key genes responsible for this process have not been identified thus far.

RUNX1 is an important transcriptional regulator located in DSCR, which is closely associated with the development and regeneration of the nervous system. A recent study indicated that large-scale hypermethylation of RUNX1 (~28 kb) was the strongest signal within the DS differentially methylated regions in peripheral blood (Laufer et al., 2021), which was also confirmed by the current study. In the present study, overexpression of RUNX1 was observed in both DS-iPSCs and peripheral blood samples of children with DS. It was therefore speculated that RUNX1 may be a crucial regulatory gene involved in the pathogenesis of DS. In a recent study, RUNX1 mutations were detected in a patient diagnosed with Pearson syndrome, which was caused by deletions in mitochondrial DNA (Nishimura et al., 2021).

The present study investigated whether RUNX1 affects the neurological development of DS by regulating mitochondrial function. The results revealed that overexpression of RUNX1 induced impaired mitochondrial functions, characterized by excessive generation of ROS, decreased MMP and reduced overall energy metabolism. It was also observed that RUNX1 could reduce the number of active lysosomes and cause lysosomal dysfunction. Furthermore, the data confirmed that impaired mitochondrial functions, including excessive generation of ROS, and decreased MMP and ATP content, could be reversed when the expression levels of RUNX1 were inhibited in DS-iPSCs. All these results strongly suggested that the abnormal expression levels of RUNX1 may be a critical factor (although not the sole factor) in the occurrence of mitochondrial dysfunction in DS-iPSCs, and indicated that overexpression of RUNX1 led to neurological abnormalities by affecting mitochondrial function in DS.

It is well known that the neuropathology of AD is an important clinical feature of DS, which is almost always present in patients with DS >40 years of age. Perluigi et al. (2014) demonstrated that the PI3K/Akt signaling pathway displayed increased activity in patients with DS and those with DS/AD. Notably, the PI3K/Akt signaling pathway plays a critical role in the regulation of cell proliferation, death, and metastasis (Lim et al., 2015), RUNX1 overexpression increased the phosphorylation levels of PI3K, Akt, and mTOR proteins (Liu et al., 2020).

Akt is a well-known pro-survival kinase and activated by phosphorylation at Ser 473 via the PI3K signaling pathway. Activated Akt plays a critical role in inhibiting neuronal death through further activation or inhibition of its downstream target proteins (Ye et al., 2015). In the present study, it was further found that overexpression of RUNX1 may activate the PI3K/Akt signaling pathway via increasing the phosphorylation of Akt, S6 and 4EBP1 in iPSCs. Certain upregulated genes in the PI3K-Akt signaling pathway, including Bim, IGF2, ITGA11, ITGB3, and CREB5, were considered to be closely involved in cell proliferation, apoptosis and tumor progression (Pardo et al., 2019; Shin et al., 2017; Wu et al., 2015; Yu et al., 2018). These genes were identified when RUNX1 was overexpressed in iPSCs. The present study also demonstrated that cell apoptosis was significantly increased in RUNX1-overexpressing iPSCs. Therefore, it was speculated that overexpression of RUNX1 in DS-iPSCs increased cell apoptosis by activating the PI3K-Akt signaling pathway.

Previous reports showed that Akt plays a central role in apoptosis inhibition through its regulatory effects on various downstream targets such as anti-apoptotic Bcl-2 (Yu et al., 2016). Normal mitochondrial function depends on the integrity of the mitochondrial membrane, which is strictly regulated by the Bcl-2 family of proteins (D'Orsi et al., 2017). The balance of cell death and survival signals in the Bcl-2 protein family determines the cell fate. As a pro-apoptotic member of the Bcl-2 family, Bim induces the release of cytochrome c from the mitochondria and activates the mitochondrial apoptotic pathway, which is implicated in the regulation of neuronal apoptosis (Concannon et al., 2010; Putcha et al., 2001). Bax is a pro-apoptotic protein, which can release cytochrome c from the mitochondria and induce changes in mitochondrial permeability, ultimately resulting in severe morphological changes in mitochondrial structure. Bcl-2 is an anti-apoptotic protein that regulates apoptosis by controlling the permeability of the mitochondrial membrane (Lv et al., 2018; Valiyari et al., 2017). As a result, the Bcl-2/Bax ratio can affect mitochondrial membrane permeability and is closely associated with the mitochondrial-mediated apoptotic pathway. In the present study, RUNX1 overexpression led to a significant increase in Bax expression and a significant decrease in Bcl-2 expression, thereby reducing the ratio of Bcl-2/Bax. Thus, abnormal expression of genes that belong to the Bcl-2 family, which was mediated by RUNX1 overexpression, may be an important factor for the increased apoptosis observed in DS-iPSCs. Based on the aforementioned results, it was concluded that RUNX1 could induce apoptosis by activating the mitochondrial apoptotic pathway in DS-iPSCs.

In conclusion, the present study demonstrated overexpression of RUNX1 in DS-iPSCs, and found that RUNX1 could regulate mitochondrial function and mitochondria-mediated apoptosis via the modulation of the PI3K-Akt signaling pathway in iPSCs. However, further investigation is required to assess whether RUNX1 promotes the neural differentiation of DS-iPSCs by regulating mitochondrial function.

ACKNOWLEDGMENTS

This research was supported by the National Key Research and Development Program of China (2019YFA0801402) and the National Natural Science Foundation of China (81971421, 81471485), Shanghai key clinical specialty project (shslczdzk05705), Innovative Research Team of High-Level Local Universities in Shanghai (SHSMU-ZDCX20212200).

AUTHOR CONTRIBUTIONS

Y.L., J.Y., and F.Z. conceived and performed experiments, wrote the manuscript, and secured funding. Y.L. and Y.Z. performed experiments. J.Y., Z.R., and F.Z. provided expertise and feedback. All authors have read and agreed to the published version of the manuscript.

CONFLICT OF INTEREST

The authors have no potential conflicts of interest to disclose.

Fig 1.

Figure 1.Impaired mitochondrial function in DS-iPSCs. (A) ROS levels in iPSCs were measured using a ROS assay kit. DS-iPSCs and N-iPSCs were observed under bright light and fluorescence microscopy, respectively. (B) Fluorescence intensity of DS-iPSCs and N-iPSCs was analyzed using ImageJ. ***P < 0.001 versus control samples. (C) ATP content in DS-iPSCs and N-iPSCs. **P < 0.01 versus control samples. (D) The mitochondrial membrane potential was measured with the JC-1 dye. DS-iPSCs and N-iPSCs were observed using different wavelengths under a fluorescence microscope (514 nm for green fluorescence and 585 nm for red fluorescence). (E) The ratio of red fluorescence to green fluorescence intensity was analyzed using ImageJ software. ***P < 0.001 versus control samples. DS, Down syndrome; DS-iPSCs, induced pluripotent stem cells of patients with DS; ROS, reactive oxygen species; N-iPSCs, normal induced pluripotent stem cells.
Molecules and Cells 2023; 46: 219-230https://doi.org/10.14348/molcells.2023.2095

Fig 2.

Figure 2.Overexpression of RUNX1 affects mitochondrial function in iPSCs. (A) The ROS levels of N-iPSCs transfected with RUNX1 expression vector (N-iPSCs + RUNX1) or control vector (N-iPSCs + Ctrl) were observed under bright light or fluorescence microscopy, respectively. (B) The fluorescence intensity of the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups was analyzed using ImageJ. *P < 0.05 versus control group. (C) Measurement of ATP content in the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups. **P < 0.01 versus control group. (D) The mitochondrial membrane potential was measured with the JC-1 dye, and the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups were imaged by fluorescence microscopy. (E) The ratio of red fluorescence to green fluorescence intensity was analyzed using ImageJ. *P < 0.05 versus control group. (F) The membrane ROS levels of iPSCs were measured using BODIPY-C11. The cell nuclei were stained with DAPI. The N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups were observed using different wavelengths under a fluorescence microscope (581 nm for red fluorescence and 340 nm for blue fluorescence). (G) The fluorescence intensity of the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups was analyzed using ImageJ. ***P < 0.001 versus control group. (H) The lysosomes of the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups were detected with LysoTrackerTM Red. The cells were imaged using different wavelengths (577 nm for red fluorescence and 340 nm for blue fluorescence) under a fluorescence microscope. (I) The relative fluorescence intensity of the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups was analyzed using ImageJ. **P < 0.01 versus control group. RUNX1, Runt-related transcription factor 1; iPSCs, induced pluripotent stem cells; ROS, reactive oxygen species; N-iPSCs, normal induced pluripotent stem cells; Ctrl, control.
Molecules and Cells 2023; 46: 219-230https://doi.org/10.14348/molcells.2023.2095

Fig 3.

Figure 3.Inhibition of RUNX1 expression can improve the mitochondrial function of DS- iPSCs. (A) ROS levels were detected in DS-iPSCs transfected with RUNX1 shRNA vector (DS-iPSCs + shRUNX1) or control vector (DS-iPSCs + Ctrl). The results were imaged with bright light or fluorescence microscopy, respectively. (B) The fluorescence intensity of the DS-iPSCs + shRUNX1 and DS-iPSCs + Ctrl groups was analyzed using ImageJ. *P < 0.05 versus control group. (C) ATP content in the DS-iPSCs + shRUNX1 and DS-iPSCs + Ctrl groups. (D) The mitochondrial membrane potential was measured with the JC-1 dye, and the DS-iPSCs + shRUNX1 and DS-iPSCs + Ctrl groups were imaged by fluorescence microscopy. (E) The ratio of red fluorescence to green fluorescence intensity was analyzed using ImageJ software. *P < 0.05 versus control group. RUNX1, Runt-related transcription factor 1; DS, Down syndrome; DS-iPSCs; induced pluripotent stem cells of patients with DS; ROS, reactive oxygen species; shRNA, short hairpin RNA; Ctrl, control.
Molecules and Cells 2023; 46: 219-230https://doi.org/10.14348/molcells.2023.2095

Fig 4.

Figure 4.Overexpression of RUNX1 in iPSCs leads to abnormalities in the PI3K-Akt signaling pathway. (A) Heatmap based on significantly differentially expressed genes between the N-iPSCs + RUNX1 (n = 3) and N-iPSCs + Ctrl groups (n = 3). (B) Volcano plot of 51 differentially expressed genes. The red dots indicate upregulated gene expression (log2 fold-change > 2), whereas the blue dots indicate downregulated gene expression (log2 fold-change ≤ 2), and the grey dots correspond to non-significant differences. The total number of downregulated genes was 12, while the total number of upregulated genes was 39. (C) Results of Kyoto Encyclopedia of Genes and Genomes pathway analysis. The scatter plot indicates the pathway category on the left-hand side of the plot. The red dots indicate significant pathways. The most impacted upregulated pathway was the PI3K-Akt signaling pathway. (D) The relative expression levels of certain differentially expressed genes involved in the PI3K-Akt signaling pathway were evaluated by RT-qPCR. N-iPSCs + Ctrl and N-iPSCs + RUNX1 were represented with black and grey colors, respectively. *P < 0.05, **P < 0.01 versus control group. RUNX1, Runt-related transcription factor; iPSCs; induced pluripotent stem cells; N-iPSCs, normal induced pluripotent stem cells; Ctrl, control; RT-qPCR, reverse transcription-quantitative polymerase chain reaction.
Molecules and Cells 2023; 46: 219-230https://doi.org/10.14348/molcells.2023.2095

Fig 5.

Figure 5.Effect of overexpression of RUNX1 on the PI3K-Akt signaling pathway and cell apoptosis. (A) The expression levels and phosphorylation of Akt, S6 and 4E-binding protein were identified using western blotting in the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups. (B) Fluorescence activated cell sorting analysis was used to assess cell apoptosis, and the data were expressed as the percentage of annexin-V FITC-positive cells, including viable apoptotic cells (PE-positive cells, Q2) and non-viable apoptotic cells (PE-negative cells, Q3). (C) Apoptotic rate in the N-iPSCs + RUNX1 and N-iPSCs + Ctrl groups. **P < 0.01 versus control group. (D) The relative expression levels of certain apoptotic biomarkers were assessed by RT-qPCR. N-iPSCs + Ctrl and N-iPSCs + RUNX1 were represented with black and grey colors, respectively. *P < 0.05, **P < 0.01 versus control group. RUNX1, Runt-related transcription factor 1; N-iPSCs, normal induced pluripotent stem cells; Ctrl, control; PE, phycoerythrin; Q, quadrant; RT-qPCR, reverse transcription-quantitative polymerase chain reaction.
Molecules and Cells 2023; 46: 219-230https://doi.org/10.14348/molcells.2023.2095

References

  1. Aburawi E.H. and Souid A.K. (2012). Lymphocyte respiration in children with Trisomy 21. BMC Pediatr. 12, 193.
    Pubmed KoreaMed CrossRef
  2. Alafnan A., Hussain T., Rizvi S.M.D., Moin A., and Alamri A. (2021). Prostate apoptotic induction and NFκB suppression by dammarolic acid: mechanistic insight into onco-therapeutic action of an aglycone asiaticoside. Curr. Issues Mol. Biol. 43, 932-940.
    Pubmed KoreaMed CrossRef
  3. Arrázola M.S., Andraini T., Szelechowski M., Mouledous L., Arnauné-Pelloquin L., Davezac N., Belenguer P., Rampon C., and Miquel M.C. (2019). Mitochondria in developmental and adult neurogenesis. Neurotox. Res. 36, 257-267.
    Pubmed CrossRef
  4. Beckervordersandforth R. (2017). Mitochondrial metabolism-mediated regulation of adult neurogenesis. Brain Plast. 3, 73-87.
    Pubmed KoreaMed CrossRef
  5. Bhatti J.S., Bhatti G.K., and Reddy P.H. (2017). Mitochondrial dysfunction and oxidative stress in metabolic disorders - a step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 1863, 1066-1077.
    Pubmed KoreaMed CrossRef
  6. Bourquin J.P., Subramanian A., Langebrake C., Reinhardt D., Bernard O., Ballerini P., Baruchel A., Cavé H., Dastugue N., and Hasle H., et al. (2006). Identification of distinct molecular phenotypes in acute megakaryoblastic leukemia by gene expression profiling. Proc. Natl. Acad. Sci. U. S. A. 103, 3339-3344.
    Pubmed KoreaMed CrossRef
  7. Concannon C.G., Tuffy L.P., Weisová P., Bonner H.P., Dávila D., Bonner C., Devocelle M.C., Strasser A., Ward M.W., and Prehn J.H. (2010). AMP kinase-mediated activation of the BH3-only protein Bim couples energy depletion to stress-induced apoptosis. J. Cell Biol. 189, 83-94.
    Pubmed KoreaMed CrossRef
  8. Coskun P.E. and Busciglio J. (2012). Oxidative stress and mitochondrial dysfunction in Down's syndrome: relevance to aging and dementia. Curr. Gerontol. Geriatr. Res. 2012, 383170.
    Pubmed KoreaMed CrossRef
  9. D'Orsi B., Mateyka J., and Prehn J.H.M. (2017). Control of mitochondrial physiology and cell death by the Bcl-2 family proteins Bax and Bok. Neurochem. Int. 109, 162-170.
    Pubmed CrossRef
  10. Delabar J.M., Theophile D., Rahmani Z., Chettouh Z., Blouin J.L., Prieur M., Noel B., and Sinet P.M. (1993). Molecular mapping of twenty-four features of Down syndrome on chromosome 21. Eur. J. Hum. Genet. 1, 114-124.
    Pubmed CrossRef
  11. Edwards H., Xie C., LaFiura K.M., Dombkowski A.A., Buck S.A., Boerner J.L., Taub J.W., Matherly L.H., and Ge Y. (2009). RUNX1 regulates phosphoinositide 3-kinase/AKT pathway: role in chemotherapy sensitivity in acute megakaryocytic leukemia. Blood 114, 2744-2752.
    Pubmed KoreaMed CrossRef
  12. El Hajj N., Dittrich M., Böck J., Kraus T.F., Nanda I., Müller T., Seidmann L., Tralau T., Galetzka D., and Schneider E., et al. (2016). Epigenetic dysregulation in the developing Down syndrome cortex. Epigenetics 11, 563-578.
    Pubmed KoreaMed CrossRef
  13. Flippo K.H. and Strack S. (2017). Mitochondrial dynamics in neuronal injury, development and plasticity. J. Cell Sci. 130, 671-681.
    Pubmed KoreaMed CrossRef
  14. Grabherr M.G., Haas B.J., Yassour M., Levin J.Z., Thompson D.A., Amit I., Adiconis X., Fan L., Raychowdhury R., and Zeng Q., et al. (2011). Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat. Biotechnol. 29, 644-652.
    Pubmed KoreaMed CrossRef
  15. Gutti U., Komati J.K., Kotipalli A., Saladi R.G.V., and Gutti R.K. (2018). Justicia adhatoda induces megakaryocyte differentiation through mitochondrial ROS generation. Phytomedicine 43, 135-139.
    Pubmed CrossRef
  16. Halevy T., Biancotti J.C., Yanuka O., Golan-Lev T., and Benvenisty N. (2016). Molecular characterization of Down syndrome embryonic stem cells reveals a role for RUNX1 in neural differentiation. Stem Cell Reports 7, 777-786.
    Pubmed KoreaMed CrossRef
  17. Hart J.R. and Vogt P.K. (2011). Phosphorylation of AKT: a mutational analysis. Oncotarget 2, 467-476.
    Pubmed KoreaMed CrossRef
  18. Hirata K., Nambara T., Kawatani K., Nawa N., Yoshimatsu H., Kusakabe H., Banno K., Nishimura K., Ohtaka M., and Nakanishi M., et al. (2020). 4-Phenylbutyrate ameliorates apoptotic neural cell death in Down syndrome by reducing protein aggregates. Sci. Rep. 10, 14047.
    Pubmed KoreaMed CrossRef
  19. Hu F. and Liu F. (2011). Mitochondrial stress: a bridge between mitochondrial dysfunction and metabolic diseases? Cell. Signal. 23, 1528-1533.
    Pubmed KoreaMed CrossRef
  20. Izzo A., Mollo N., Nitti M., Paladino S., Calì G., Genesio R., Bonfiglio F., Cicatiello R., Barbato M., and Sarnataro V., et al. (2018). Mitochondrial dysfunction in down syndrome: molecular mechanisms and therapeutic targets. Mol. Med. 24, 2.
    Pubmed KoreaMed CrossRef
  21. Jia L., Jiang Y., Li X., and Chen Z. (2020). Purβ promotes hepatic glucose production by increasing Adcy6 transcription. Mol. Metab. 31, 85-97.
    Pubmed KoreaMed CrossRef
  22. Johnson J., Mercado-Ayon E., Mercado-Ayon Y., Dong Y.N., Halawani S., Ngaba L., and Lynch D.R. (2021). Mitochondrial dysfunction in the development and progression of neurodegenerative diseases. Arch. Biochem. Biophys. 702, 108698.
    Pubmed CrossRef
  23. Kann O. and Kovács R. (2007). Mitochondria and neuronal activity. Am. J. Physiol. Cell Physiol. 292, C641-C657.
    Pubmed CrossRef
  24. Laufer B.I., Hwang H., Jianu J.M., Mordaunt C.E., Korf I.F., Hertz-Picciotto I., and LaSalle J.M. (2021). Low-pass whole genome bisulfite sequencing of neonatal dried blood spots identifies a role for RUNX1 in Down syndrome DNA methylation profiles. Hum. Mol. Genet. 29, 3465-3476.
    Pubmed KoreaMed CrossRef
  25. Li H., Liu J., Wang Y., Fu Z., Hüttemann M., Monks T.J., Chen A.F., and Wang J.M. (2017). MiR-27b augments bone marrow progenitor cell survival via suppressing the mitochondrial apoptotic pathway in Type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 313, E391-E401.
    Pubmed KoreaMed CrossRef
  26. Li X., Sun Y., Jin Q., Song D., and Diao Y. (2019). Kappa opioid receptor agonists improve postoperative cognitive dysfunction in rats via the JAK2/STAT3 signaling pathway. Int. J. Mol. Med. 44, 1866-1876.
    Pubmed KoreaMed CrossRef
  27. Li Z., Jiang T., Lu Q., Xu K., He J., Xie L., Chen Z., Zheng Z., Ye L., and Xu K., et al. (2020). Berberine attenuated the cytotoxicity induced by t-BHP via inhibiting oxidative stress and mitochondria dysfunction in PC-12 cells. Cell. Mol. Neurobiol. 40, 587-602.
    Pubmed CrossRef
  28. Lim H.J., Crowe P., and Yang J.L. (2015). Current clinical regulation of PI3K/PTEN/Akt/mTOR signalling in treatment of human cancer. J. Cancer Res. Clin. Oncol. 141, 671-689.
    Pubmed CrossRef
  29. Liu S., Zhang Y., Huang C., and Lin S. (2020). miR-215-5p is an anticancer gene in multiple myeloma by targeting RUNX1 and deactivating the PI3K/AKT/mTOR pathway. J. Cell. Biochem. 121, 1475-1490.
    Pubmed CrossRef
  30. Lv J., Liang Y., Tu Y., Chen J., and Xie Y. (2018). Hypoxic preconditioning reduces propofol-induced neuroapoptosis via regulation of Bcl-2 and Bax and downregulation of activated caspase-3 in the hippocampus of neonatal rats. Neurol. Res. 40, 767-773.
    Pubmed CrossRef
  31. McCubrey J.A., Steelman L.S., Chappell W.H., Abrams S.L., Montalto G., Cervello M., Nicoletti F., Fagone P., Malaponte G., and Mazzarino M.C., et al. (2012). Mutations and deregulation of Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascades which alter therapy response. Oncotarget 3, 954-987.
    Pubmed KoreaMed CrossRef
  32. Nishimura A., Hirabayashi S., Hasegawa D., Yoshida K., Shiraishi Y., Ashiarai M., Hosoya Y., Fujiwara T., Harigae H., and Miyano S., et al. (2021). Acquisition of monosomy 7 and a RUNX1 mutation in Pearson syndrome. Pediatr. Blood Cancer 68, e28799.
    Pubmed CrossRef
  33. Ohnishi T., Ohnishi S.T., and Salerno J.C. (2018). Five decades of research on mitochondrial NADH-quinone oxidoreductase (complex I). Biol. Chem. 399, 1249-1264.
    Pubmed CrossRef
  34. Pardo M., Cheng Y., Sitbon Y.H., Lowell J.A., Grieco S.F., Worthen R.J., Desse S., and Barreda-Diaz A. (2019). Insulin growth factor 2 (IGF2) as an emergent target in psychiatric and neurological disorders. Review. Neurosci. Res. 149, 1-13.
    Pubmed CrossRef
  35. Patel A., Rees S.D., Kelly M.A., Bain S.C., Barnett A.H., Thalitaya D., and Prasher V.P. (2011). Association of variants within APOE, SORL1, RUNX1, BACE1 and ALDH18A1 with dementia in Alzheimer's disease in subjects with Down syndrome. Neurosci. Lett. 487, 144-148.
    Pubmed CrossRef
  36. Pecze L. and Szabo C. (2021). Meta-analysis of gene expression patterns in Down syndrome highlights significant alterations in mitochondrial and bioenergetic pathways. Mitochondrion 57, 163-172.
    Pubmed CrossRef
  37. Pelleri M.C., Cicchini E., Locatelli C., Vitale L., Caracausi M., Piovesan A., Rocca A., Po-letti G., Seri M., and Strippoli P., et al. (2016). Systematic reanalysis of partial trisomy 21 cases with or without Down syndrome suggests a small region on 21q22.13 as critical to the phenotype. Hum. Mol. Genet. 25, 2525-2538.
    Pubmed KoreaMed CrossRef
  38. Perluigi M., Pupo G., Tramutola A., Cini C., Coccia R., Barone E., Head E., Butterfield D.A., and Di Domenico F. (2014). Neuropathological role of PI3K/Akt/mTOR axis in Down syndrome brain. Biochim. Biophys. Acta 1842, 1144-1153.
    Pubmed KoreaMed CrossRef
  39. Putcha G.V., Moulder K.L., Golden J.P., Bouillet P., Adams J.A., Strasser A., and Johnson E.M. (2001). Induction of BIM, a proapoptotic BH3-only BCL-2 family member, is critical for neuronal apoptosis. Neuron 29, 615-628.
    Pubmed CrossRef
  40. Qiu J.J., Liu Y.N., Ren Z.R., and Yan J.B. (2017). Dysfunctions of mitochondria in close association with strong perturbation of long noncoding RNAs expression in down syndrome. Int. J. Biochem. Cell Biol. 92, 115-120.
    Pubmed CrossRef
  41. Salemi M., Giambirtone M., Barone C., Salluzzo M.G., Russo R., Giudice M.L., Cutuli S., Ridolfo F., and Romano C. (2018). Mitochondrial mRNA expression in fibroblasts of Down syndrome subjects. Hum. Cell 31, 179-181.
    Pubmed CrossRef
  42. Salemi M., Ridolfo F., Salluzzo M.G., Cannarrella R., Giambirtone M., Caniglia S., Tirolo C., Ferri R., and Romano C. (2020). Humanin gene expression in fibroblast of Down syndrome subjects. Int. J. Med. Sci. 17, 320-324.
    Pubmed KoreaMed CrossRef
  43. Shah S.I., Paine J.G., Perez C., and Ullah G. (2019). Mitochondrial fragmentation and network architecture in degenerative diseases. PLoS One 14, e0223014.
    Pubmed KoreaMed CrossRef
  44. Shin J., Shim H.G., Hwang T., Kim H., Kang S.H., Dho Y.S., Park S.H., Kim S.J., and Park C.K. (2017). Restoration of miR-29b exerts anti-cancer effects on glioblastoma. Cancer Cell Int. 17, 104.
    Pubmed KoreaMed CrossRef
  45. Vacca R.A., Bawari S., Valenti D., Tewari D., Nabavi S.F., Shirooie S., Sah A.N., Volpicella M., Braidy N., and Nabavi S.M. (2019). Down syndrome: neurobiological alterations and therapeutic targets. Neurosci. Biobehav. Rev. 98, 234-255.
    Pubmed CrossRef
  46. Valenti D., Braidy N., De Rasmo D., Signorile A., Rossi L., Atanasov A.G., Volpicella M., Henrion-Caude A., Nabavi S.M., and Vacca R.A. (2018). Mitochondria as pharmacological targets in Down syndrome. Free Radic. Biol. Med. 114, 69-83.
    Pubmed CrossRef
  47. Valiyari S., Salami M., Mahdian R., Shokrgozar M.A., Oloomi M., Mohammadi Farsani A., and Bouzari S. (2017). sIL-24 peptide, a human interleukin-24 isoform, induces mitochondrial-mediated apoptosis in human cancer cells. Cancer Chemother. Pharmacol. 80, 451-459.
    Pubmed CrossRef
  48. Wong R.S. (2011). Apoptosis in cancer: from pathogenesis to treatment. J. Exp. Clin. Cancer Res. 30, 87.
    Pubmed KoreaMed CrossRef
  49. Wu J., Zheng C., Wang X., Yun S., Zhao Y., Liu L., Lu Y., Ye Y., Zhu X., and Zhang C., et al. (2015). MicroRNA-30 family members regulate calcium/calcineurin signaling in podocytes. J. Clin. Invest. 125, 4091-4106.
    Pubmed KoreaMed CrossRef
  50. Ye Z., Xia P., Cheng Z.G., and Guo Q. (2015). Neuroprotection induced by sevoflurane-delayed post-conditioning is attributable to increased phosphorylation of mitochondrial GSK-3β through the PI3K/Akt survival pathway. J. Neurol. Sci. 348, 216-225.
    Pubmed CrossRef
  51. Yoshikawa M., Hirabayashi M., Ito R., Ozaki S., Aizawa S., Masuda T., Senzaki K., and Shiga T. (2015). Contribution of the Runx1 transcription factor to axonal pathfinding and muscle innervation by hypoglossal motoneurons. Dev. Neurobiol. 75, 1295-1314.
    Pubmed CrossRef
  52. Yoshikawa M., Masuda T., Kobayashi A., Senzaki K., Ozaki S., Aizawa S., and Shiga T. (2016). Runx1 contributes to the functional switching of bone morphogenetic protein 4 (BMP4) from neurite outgrowth promoting to suppressing in dorsal root ganglion. Mol. Cell. Neurosci. 72, 114-122.
    Pubmed CrossRef
  53. Yu G., Yin C., Jiang L., Zheng Z., Wang Z., Wang C., Zhou H., Jiang X., Liu Q., and Meng F. (2016). Amyloid precursor protein cooperates with c-KIT mutation/overexpression to regulate cell apoptosis in AML1-ETO-positive leukemia via the PI3K/AKT signaling pathway. Oncol. Rep. 36, 1626-1632.
    Pubmed CrossRef
  54. Yu M., Du G., Xu Q., Huang Z., Huang X., Qin Y., Han L., Fan Y., Zhang Y., and Han X., et al. (2018). Integrated analysis of DNA methylome and transcriptome identified CREB5 as a novel risk gene contributing to recurrent pregnancy loss. EBioMedicine 35, 334-344.
    Pubmed KoreaMed CrossRef
  55. Yuan Y., Chen Y., Peng T., Li L., Zhu W., Liu F., Liu S., An X., Luo R., and Cheng J., et al. (2019). Mitochondrial ROS-induced lysosomal dysfunction impairs autophagic flux and contributes to M1 macrophage polarization in a diabetic condition. Clin. Sci. (Lond.) 133, 1759-1777.
    Pubmed CrossRef
  56. Zamponi E. and Helguera P.R. (2019). The shape of mitochondrial dysfunction in Down syndrome. Dev. Neurobiol. 79, 613-621.
    Pubmed CrossRef
  57. Zhang W., Hu X., Shen Q., and Xing D. (2019). Mitochondria-specific drug release and reactive oxygen species burst induced by polyprodrug nanoreactors can enhance chemotherapy. Nat. Commun. 10, 1704.
    Pubmed KoreaMed CrossRef
  58. Zhao X., Song X., Bai X., Fei N., Huang Y., Zhao Z., Du Q., Zhang H., Zhang L., and Tong D. (2016). miR-27b attenuates apoptosis induced by transmissible gastroenteritis virus (TGEV) infection via targeting runt-related transcription factor 1 (RUNX1). PeerJ 4, e1635.
    Pubmed KoreaMed CrossRef
Mol. Cells
Nov 30, 2023 Vol.46 No.11, pp. 655~725
COVER PICTURE
Kim et al. (pp. 710-724) demonstrated that a pathogen-derived Ralstonia pseudosolanacearum type III effector RipL delays flowering time and enhances susceptibility to bacterial infection in Arabidopsis thaliana. Shown is the RipL-expressing Arabidopsis plant, which displays general dampening of the transcriptional program during pathogen infection, grown in long-day conditions.

Supplementary File

Share this article on

  • line

Related articles in Mol. Cells

Molecules and Cells

eISSN 0219-1032
qr-code Download