The microRNA miR-375-3p and the Tumor Suppressor NDRG2 are Involved in Sporadic Amyotrophic Lateral Sclerosis

 

Marlena Rohma    Caroline Mayb    Katrin Marcusb    Simone Steinbachb    

Verena Theisa    Carsten Theissa    Veronika Matschkea

 

aRuhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Cytology, Bochum, Germany, bRuhr University Bochum, Medical Faculty, Medizinisches Proteom-Center, Bochum, Germany

 

 

 

 

Key Words

Wobbler mouse • p53 • miRNA • Neurodegeneration • Motor neuron

 

Abstract

Background/Aims: Amyotrophic lateral sclerosis (ALS) is the most common degenerative motor neuron disease in humans. However, the pathogenesis of ALS is not yet understood. The wobbler mouse is considered as an animal model for the sporadic form of ALS due to its spontaneous mutation in the Vps54 gene. Due to transactivation of NDRG2 by p53, this tumor suppressor might play a functional role in stress induced cell death in wobbler mice as well as ALS patients. Furthermore, deregulated microRNAs are often related to neurodegenerative diseases. Thus, the NDRG2 linked miR-375-3p was of interest for this study. Methods: Here, we investigated the relevance of NDRG2 and miR-375-3p for the pathomechanism of the motor neuronal degeneration in wobbler mice by investigating expression level via qPCR and Western Blot as well as localization of these molecules in the cervical spinal cord by in situ hybridization, immunostaining and mass spectrometric analysis. Results: We were able to show a differential regulation of the expression of NDRG2 as well as miR-375-3p in the cervical part of the spinal cord of wobbler mice. In addition, for the first time we were able to demonstrate an expression of NDRG2 in motor neurons using different techniques. Conclusion: The present study has shown NDRG2 and miR-375-3p to be promising targets for further research of the pathogenesis of sporadic ALS in the wobbler mouse model. Based on these results and in combination with previous published data we could develop a putative pro-apoptotic mechanism in the spinal cord of the wobbler mouse.

 

 

Introduction

 

The wobbler mouse, first described in 1956, is an important model for the sporadic form of amyotrophic lateral sclerosis (ALS), the most common motor neuron disease in adults [1]. 95% of ALS patients suffer from the sporadic form without any known hereditary component. Degeneration of the upper motor neurons in the cerebral cortex and the lower motor neurons in the brainstem and spinal cord are characteristic hallmarks for ALS. This degeneration leads to spasticity, generalized muscle weakness, atrophy and therefore progressive paralysis [2]. ALS patients die within 2-5 years after the appearance of the first symptoms due to a missing beneficial treatment [3].

The symptoms displayed by the homozygous wobbler mouse correspond to the human ALS phenotype [4, 5]. Therefore, the wobbler mouse is used to reveal the pathological mechanisms underlying the amyotrophic lateral sclerosis. A missense point mutation (L967Q) in the Vps54 gene of the wobbler mouse, a component of the Golgi-associated retrograde protein (GARP) complex that is involved in retrograde transport from early and late endosomes to the trans-Golgi network, was already identified, but there is still no link between the occurring symptoms and the mutation [6]. However, recently mutations in the Vps54 gene of ALS patients with moderate or even high impact were detected within the project MinE (http://databrowser.projectmine.com/) . The symptom progression of the wobbler mouse can be monitored and divided into three distinct stages [7]. From postnatal 0 (p0) to p20 there is no phenotypic difference between the wobbler mouse and their littermates. Therefore, this phase is called the presymptomatic phase. During the preclinical phase from p20 until p40 the symptoms begin to develop. The wobbler mouse stays visibly smaller than their healthy littermates, shows typical muscle weakness of the front feet and develops a head tremor [8]. From p40 the stable clinical phase starts, where the symptoms are fully developed and stagnate [9].

The N-myc downstream-regulated gene 2 (NDRG2) protein is involved in cell proliferation, differentiation, transmembrane transport but also in stress response and depression [10–12]. NDRG2 is also involved in cell death by different pathways. Within one possible signaling cascade, NDRG2 is transactivated directly by the transcription factor p53 and therefore might play a role in p53-mediated apoptosis [13, 14]. Interestingly, p53 levels are known to be elevated in the spinal cord of wobbler mice as well as ALS patients [15]. This indicates, that p53 as well as NDRG2 might play a functional role in stress induced cell death in wobbler mice as well as ALS patients. While in the past it was assumed that NDRG2 is only expressed in astrocytes and oligodendrocytes, NDRG2 was detected in pyramidal neurons of patients with Alzheimer’s disease [16, 17]. A recent study found the Death-associated protein kinase 1 (DAPK1) to be a critical regulator of NDRG2 by phosphorylating it and subsequently mediate neuronal cell death during the progression of Alzheimer’s disease [18, 19]. Another reason for NDRG2 to be an interesting target is its connection to FUS, a protein that is connected to the familial form of ALS (fALS) [20, 21]. Even though the consequence is not yet understood, it is known that depletion of FUS leads to alternative splicing of NDRG2 and ultimately cell death [20].

Neurodegenerative diseases are often related to deregulation of microRNA (miRNA, miR). These small molecules are able to prevent translation of proteins by binding to their specific target mRNA and either block the translation or degrade them [22]. The miRNA miR-375-3p is directly linked to NDRG2: microglia secret microvesicles containing miR-375-3p after overexpression of NDRG2 leading to a damaging effect on neuronal cells in vitro [23]. Contrary to this work, a neuro protective function of miR-375-3p by inhibiting p53 and therefore preventing apoptosis could be shown by Bhinge et al. [24]. They showed miR-375-3p to be essential for spinal cord motor neuron development. Downregulation of the miR-375-3p in patients with spinal muscular atrophy leads to an increase of the p53 protein level and thus to apoptosis [24]. In line with that, De Santis and colleagues have shown a decreased expression of miRNA miR-375-3p in human iPSC-derived motor neurons with a fALS typical FUS mutation [25]. They also showed an increase of proapoptotic factors, like p53, to be due to the downregulation of miR-375-3p [25].

NDRG2 overexpression leading to neuronal degeneration through increased secretion of miR-375-3p by microglia and reduction of miR-375-3p leading to elevated level of p53 and therefore apoptosis, are basic pathological mechanisms, making miR-375-3p an interesting candidate for further research in a mouse model for ALS.

In this study we investigate the relevance of NDRG2 and miR-375-3p for the pathomechanism of the motor neuronal degeneration in wobbler mice by investigating the expression level and localization of these molecules in the cervical spinal cord.

 

 

Materials and Methods

 

Animals

All experiments were conducted under established standards of the German federal state of North Rhine Westphalia, in accordance with the European Communities Council Directive 2010/63/EU on the protection of animal used for scientific purposes. Breeding, handling and genotyping of the mice, which carried the wobbler mutation, was performed as previously described [8]. For each experiment the cervical part of the spinal cords of three to five animals of each group [wild type (WT) and wobbler (WR), at three different developmental stages (p0, p20, and p40)] were collected.

 

qPCR

Total RNA was extracted from 10-15mg cervical spinal cord tissue with the aid of the NucleoSpin miRNA Kit (#740971, Macherey-Nagel, Düren, Germany) according to manufacturer’s protocol. Afterwards the tRNA was transcribed into cDNA. For analysis of NDRG2 mRNA expression, qScript cDNA Synthesis Kit (Quanta) with 500ng tRNA was used. For miRNA expression analysis, 10ng tRNA was reverse transcribed with the miRCURY LNA RT Kit (#339340, Qiagen, Exiqon, Denmark). The quantitative real-time PCR (qPCR) was performed using GoTag qPCR Master Mix (#A6001, Promega, Madison, WI, USA) on a CFX96 Real-Time PCR Detection System (Bio-Rad, Hercules, CA, USA). The housekeeping gene GAPDH was used for mRNA normalization and U6 for miRNA normalization. Following primer were used: NDRG2-fw 5’- AGGGCGGACCTAAGTCAAAG-3’ and NDRG2-rev 5’- ATCTCCCTGCACAATCTGCC-3’, GAPDH-fw 5’- GGA GAA ACC TGC CAA GTA TGA -3’ and GAPDH-rev 5’- TCC TCA GTG TAG CCC AAG A -3’. Primer used for expression analysis of miRNA were custom designs from Exiqon (Denmark): hsa-miR-375-3p 5’- UUUGUUCGUUCGGCUCGCGUGA and hsa-miR-125b-5p 5’- UCCCUGAGACCCUAACUUGUGA-3’. Expression levels were analyzed in three independent qPCR runs in triplicates and normalized with the appropriate housekeeper. Furthermore the obtained Ct-values were analyzed using the 2−ΔΔCt method [26].

 

SDS-Page and Western Blotting

Protein from 10-15 mg cervical spinal cord tissue was extracted with cell lysis buffer (#9803, Cell Signaling Technology, Germany) supplemented with protease inhibitor (#11697498001, Sigma Aldrich, Germany). After determination of protein concentrations by using Pierce BCA Protein Assay Kit (#23225, Thermo Fisher Scientific, USA), 30µg of protein was applied per lane on an SDS gel. The samples were transferred with the Trans-Blot Turbo Transfer System (BioRad, USA) onto a nitrocellulose membrane and blocked with 1% RotiBlock (#A151, Roth, Germany) in 1x TBS for 1h at RT. Primary antibodies were incubated over night at 4°C. For detection of NDRG2, an polyclonal IgG antibody was used (rabbit polyclonal IgG anti-NDRG2 antibody 1:750, #HPA002896, Atlas Antibody, Sweden). Calnexin was used as housekeeper (rabbit polyclonal IgG anti-calnexin antibody 1:200, #sc-11397, Santa Cruz, USA). After washing with TBS-T, the membrane was incubated for one hour at RT with a HRP-coupled secondary antibody(goat anti-rabbit antibody 1:5000; #sc-2054, Santa Cruz, USA). Luminol reagent (#sc-2048, Santa Cruz, USA) was used for signal detection with an imaging system (ChemiDoc XRS+, Bio-Rad, USA).

To verify the Western Blot results, an SPL Western Blot was additionally performed (data not shown).

 

Cryosectioning

For immunostaining, in situ hybridization and laser micro dissection fresh/frozen cervical spinal cord tissue from p20 and p40 wildtype and wobbler mice was used. Therefore, the tissue was isolated and frozen in isopentane at –45°C. For immunostaining and in situ hybridization, samples were embedded in tissue freezing medium (Cryoglue #30001100, Slee, Germany) and cut on a cryostat (Leica CM 3050 S; chamber and stage temperature at -22 °C), whereas for laser micro dissection no embedding medium was used (object and chamber temperature at -15°C). Sections for in situ hybridization had a thickness of 16µm, while for immunostaining and laser micro dissection the samples were cut to a thickness of 10µm. Sections were collected on super frost glass slides (#J1800AMNZ, Menzel-Gläser, Germany). For laser micro dissection the sections were placed on 1.0 PEN membrane (#415190-9041-000, Carl Zeiss Microscopy GmbH, Germany). All slides were stored at -80°C until further use.

 

In situ hybridization

In situ hybridization was performed according to the one-day-protocol of the manual FFPE in situ hybridization using double-labeled Fluorescein or DIG miRCURY LNA microRNA Detection probes (Exiqon, Denmark). In brief, 16µm fresh/frozen sections of the cervical spinal cord (p20 and p40) were fixed with 4% PFA for 30min at RT and permeabilized with 1µg/ml Proteinase K for 10min at 37°C. Each probe (60nM mNDRG2_2, /5DigN/ACACACACACACTCTCTCTCCT/3Dig_N/, #651992-2, Exiqon, Denmark; 80nM mmu-miR-375-3p, /5DigN/TCACGCGAGCCGAACGAACAAA/3Dig_N/, #616554-360, Exiqon, Denmark) was denaturated at 90°C for 4min and supplemented afterwards with 200 µl in situ hybridization buffer (#ENZ-33808, Enzo Life Sciences, USA). A scrambled mRNA probe (#300514-15, Qiagen, Exiqon, Denmark) and a scrambled miRNA probe (#90-001, Exiqon, Denmark) were used as negative controls. This mixture was applied to the sections and incubated for 2h at 54°C in a humidified chamber. Afterwards, the tissue was washed once with 5×SSC for 5min and twice with 1×SSC and with 0.2×SSC for 5min at hybridization temperature and finally once with 0.2×SSC for 5min at RT. Blocking was performed with PBS-T supplemented with 2% sheep serum and 1% BSA for 15min at RT. For detection, incubation with an antibody against digoxigenin (Anti-Digoxigenin-AP # 11093274910, 1:800, Roche, Swiss) was performed for 1h at RT. NBT-BCIP (NBT/BCIP Ready-to-Use Tablets #11697471001, Roche, Swiss) was supplemented with levamisole (#31742, Fluka, Germany). This substrate was applied to the tissue sections and incubated for 2h at RT. The reaction was stopped with KTBT-Buffer and washed with water. Nuclear Fast Red (#N3020 Sigma-Aldrich, Germany) was used for counterstaining. The sections were washed and drained with immersing ethanol series. The samples were embedded with Eukitt® Quick-hardening mounting medium (#03989 Sigma-Aldrich, Germany) and imaged with light microscopy (Olympus microscope BX61VS, Japan; UPlanSApo 10x/0.4, Olympus, Japan).

 

Immunofluorescence

To co-localize NDRG2 with specific cell types of the spinal cord, immunofluorescence was used. Therefore fresh/frozen 10µm slices of the cervical part of the spinal cord (p40) were post fixated with 4% PFA for 10min at RT. Permeabilization, blocking and incubating was performed according to Table 1. NDRG2 antibody was always used for duplex staining after staining of the first structural protein, as second primary antibody. For this reason, no additional permeabilization or blocking was performed. DAPI (#D9542, Sigma-Aldrich, USA; 1:1000 30 min at room temperature) was used for nuclei staining and the samples were embedded with fluoromount (#F4680, Sigma-Aldrich, USA). Finally, the samples were imaged with an inverted confocal laser scanning microscope (LSM 800, Zeiss, Germany; PlanApo 20x/0.8 DICII and PlanApo 40x/1.4 Oil DICII, Nikon instruments). Secondary antibodies were tested for specificity and showed no unspecific binding.

 

Table 1. Used conditions for duplex-immunofluorescence staining on spinal cord tissue

 

Laser micro dissection

Laser micro dissection (PALM Micro Beam, P.A.L.M.-System, Carl Zeiss Microscopy GmbH, Germany) was used to obtain pure motor neurons as well as different regions of spinal cord sections of p40 mice. To detect motor neurons, the sections were stained with cresyl violet for 30s and then briefly washed with ethanol. Motor neurons could be detected by their specific morphology in the ventral horn of the spinal cord. Using a 40x objective and the supplied software by the manufacturer (PALMRobo 4.6 pro, Carl Zeiss Microscopy GmbH) motor neurons were marked. A non-adhesive microtube cap (MicroTube 500, Carl Zeiss Microscopy GmbH) was filled with 47µl of RNase-free-water and placed over the tissue slices. Then the marked spots were cut with a laser and catapulted into the sample cup. The samples were stored at -80°C until further use.

 

Mass spectrometric analysis

The mass spectrometric analysis for identification of NDRG2 in motor neuronal cells was performed by DIA analysis.

 

Generating of a mice specific spinal cord spectral library

First a complete spinal cord of a wild type mouse was dissected. Then the spinal cord tissue was lysed and fractionated by 1D gel electrophoresis. Afterwards in-gel tryptic digestion was performed, followed by peptide extraction. Following, a DDA analysis was performed on a QExactive HF and the spectral library was generated using Spectronaut Pulsar. For a comprehensive protocol, see preparation of human substantia nigra tissue for generating a spectral library described by Barkovits et al. [27].

 

Identification of NDRG2 in motor neuronal cells y DIA

After laser microdissection, the motor neuronal cells were prepared for mass spectrometric analysis as described in detail by Aring et al. [28]. The mass spectrometric analysis itself was performed in DIA mode on a QExactive HF. For a comprehensive description of DIA measurements and data analysis see Barkovits et al. [27].

 

Statistical Analysis

For statistical analysis Prism 6 (GraphPad Inc., La Jolla, CA, USA) was used. Data represent mean values of at least three independent experiments ± standard error of the mean (SEM). Data were tested for significance using Student’s t-test and results with p-value <0.05 were considered statistically significant.

 

 

Results

 

Differential expression of NDRG2 in the cervical spinal cord of the wobbler mouse

To analyze the importance of NDRG2 for the pathomechanism of ALS in wobbler mice, we analyzed the mRNA and protein level in the cervical part of the spinal cord during all three developmental stages (p0, p20, p40) of the ALS within the wobbler mouse.

Gene expression of NDRG2 was highly significant deregulated in the cervical spinal cord of the wobbler mouse, compared to wild type mouse. Interestingly, the expression level changes during disease development from a significant downregulation at p0 to a significant upregulation at p20 and p40 (Fig. 1A). In order to specify in which region of the spinal cord the deregulation of NDRG2 mRNA at the clinical stage p40 is to be found, different areas and cells were separated with the aid of laser micro dissection and examined for mRNA expression differences by means of quantitative PCR. Significantly increased expression of NDRG2 mRNA was detected in the anterior horn, the posterior horn, and motor neurons (Fig. 1B). In contrast, the anterior column showed nearly no difference, the posterior column showed a significantly decreased NDRG2 mRNA expression (Fig. 1B). Here we could show for the first time, that NDRG2 mRNA is detectible in motor neuronal cells of the spinal cord in mice. Protein level of NDRG2 was also found to be differentially expressed in the cervical spinal cord of the wobbler mouse. The banding pattern in the Western Blot indicates the existence of both isoforms of NDRG2 in the spinal cord of the mice (Fig. 1C [29]). At the presymptomatic phase no significant difference of NDRG2 protein expression was found. Only a slight increase could be shown. A significantly down regulated expression of NDRG2 protein could be observed in p20, whereas a significantly up regulated expression was found in p40 (Fig. 1D). The Western Blot results were validated by SPL-Western Blot.

 

Fig. 1. Differential expression of NDRG2 gene and protein level in the cervical spinal cord of the wobbler mouse. (A) Expression levels of NDRG2 mRNA from presymptomatic to stationary phase of wild type (WT) and wobbler (WR) spinal cord were investigated by qPCR. Significant deregulation of NDRG2 was detected at all developmental stages. (B) Quantification of NDRG2 mRNA level in different regions of the spinal cord after laser micro dissection at p40. Significant deregulation was observed in all regions except the anterior column. For relative quantification of NDRG2 expression, the 2-ΔΔCt method was conducted using the housekeeping gene GAPDH for normalization. Data are provided as means ± SEM. Data were tested for significance using Student's t-test. Significant differences are indicated by *p<0.1, **p<0.01, ****p<0.0001. n= 5-7. (C) Representative Western Blot image of NDRG2 expression levels in WT and WR mouse spinal cord at three developmental stages of the wobbler motor neuron disease. Two bands indicate two isoforms of NDRG2. Calnexin was used as control protein. (D) Quantitative analysis of NDRG2 protein expression level in the cervical spinal cord from presymptomatic to stationary phase of WT and WR spinal cord. A significant deregulation was found in developmental stages p20 and p40, while there is no significant deregulation at time point p0. Data are provided as means ± SEM. Data were tested for significance using Student's t-test. Significant differences are indicated by *p<0.1. n = 3-5.

 

Noticeable is the negative correlation between the gene and protein expression of NDRG2 in the cervical spinal cord at the developmental stages p0 and p20. At the final clinical stage, where the symptoms are fully developed and stagnate, gene as well as protein levels are significantly increased.

 

Localization of NDRG2 in motor neurons

NDRG2 was always discussed as a marker for astrocytes, since it was only found to be expressed in astrocytes and oligodendrocytes but not in neurons in the brain [17, 30]. Furthermore NDRG2 was found to be present in pyramidal neurons under pathological conditions of patients with Alzheimer’s disease [16]. To locate NDRG2 in the spinal cord of wild type and wobbler mice, we performed in situ hybridization, immunofluorescence and laser micro dissection followed by mass spectrometry.

In situ hybridization was performed with a labeled probe detecting the NDRG2 mRNA. Staining was carried out in the cervical spinal cord of wobbler and wild type mice at the age of p20 and p40. A prominent staining of single cells can be found in the grey matter, whereas the white matter only revealed scattered light signals (Fig. 2A). At higher magnification of the anterior horn, a clear signal can be detected in large cells with a large nucleus that can be assigned to motor neuronal cells (Fig. 2B). Comparing wild type and wobbler sections during two different stages p20 and p40, no differences in NDRG2 mRNA localization can be found. Negative controls with a scrambled mRNA probe showed no staining (Supplementary Fig. 1 - for all supplemental material see www.cellphysiolbiochem.com).

 

Fig. 2. Expression pattern of NDRG2 mRNA in the cervical spinal cord of wild type and wobbler mice. (A) Exemplary overview of in situ hybridization with NDRG2-probe in a cross section of the cervical spinal cord of wild type (WT) and wobbler (WR) mice at the developmental stage p20 and p40. There are no visible staining pattern differences between WT and WR. Scale bar = 1 mm. (B) Cross section of the cervical spinal cord of a wild type mouse at p40. Exemplary large-scale picture of the anterior horn after in situ hybridization with NDRG2-probe. Arrows indicate motor neurons with a clearly visible prominent staining for NDRG2. Scale bar = 500 µm. All pictures were taken with a light microscope (Olympus microscope BX61VS, Japan) and a 20x objective (UPlanSApo 20x/0.4, Olympus, Japan). For counterstaining Nuclear Fast Red was used.

 

To verify the localization of NDRG2 on protein level, immunofluorescence was carried out to co-localize NDRG2 with different markers: GFAP representing astrocytes (Fig. 3A), ChAT for motor neurons (Fig. 3B) and CD68 as a marker for microglial cells (Fig. 3C). While the immunofluorescence showed nearly no co-localization of NDRG2 with GFAP and CD68, there is a clear visible overlap in the signals for ChAT and NDRG2 (Fig. 3A-C). Therefore, an expression of NDRG2 in motor neurons could be considered.

 

Fig. 3. Co-localization of NDRG2 with specific cell marker in immunofluorescence staining. Exemplary immunofluorescence staining of the following marker to define specific cell types: GFAP as marker for astrocytes (A), ChAT as marker for motor neurons (B) and CD68 as marker for microglial cells (C). A clear co-localization is visible for ChAT and NDRG2, whereas the other marker show nearly no co-localization. White areas in the merge channel represent a co-localization of both markers used. Nuclei were stained with DAPI (blue). Pictures were taken using a confocal laser scanning microscope (LSM 800, Zeiss, Germany with a 20x and a 40x objective (PlanApo 20x/0.8 DICII and PlanApo 40x/1.4 Oil DICII, Nikon instruments ). White arrows indicate co-localized signals.

 

Since expression of NDRG2 in neuronal cells has always been negated, mass spectrometric analysis after laser microdissection should verify the expression of NDRG2 protein in mouse motor neuronal cells. Using DIA mass spectrometric analysis, NDRG2 could be detected with a sequence coverage of 70%. Two of 14 identified peptides had a Q-value ≤ 0.01 (ILLDQGQTHSVETPYGSVTFTVYGTPKPK; LDPTQTSFLK) and one had a Q-value of ≤ 0.05 (CPVMLVVGDQAPHEDAVVECNSKLDPTQTSFLK). This indicates an NDRG2 expression in motor neurons of the spinal cord in wobbler and wild type mice.

 

miR-375-3p deregulation and expression pattern in spinal cord of mice

Overexpression of NDRG2 in a microglial cell line leads to an increase of miR-375-3p expression and secretion followed by neuronal degeneration [23]. This observations, and the fact that miR-375-3p suppresses the expression of the apoptosis initiating transcription factor p53, makes miR-375-3p an interesting miRNA in regard to motor neuron degeneration [24]. We also analyzed miR-375-3p by using the LNA technology by Exiqon (Denmark) to carry out a qPCR. Gene expression of miR-375-3p was significantly deregulated in the cervical spinal cord of the wobbler mouse, compared to wild type mouse. The expression level changes during disease development from a significant upregulation at p0 and p20 to a significant downregulation at p40 (Fig. 4A). A prominent staining of single cells can be found in the grey matter, whereas the white matter revealed no light signals (Fig. 4B). At higher magnification of the anterior horn, a clear signal can be detected in large cells with a large nucleus that can be assigned to motor neuronal cells (Fig. 4C). Looking at the posterior horn in a larger magnification, a clear signal can also be detected in smaller cells. Comparing wild type and wobbler sections during two different stages p20 and p40, no differences in NDRG2 mRNA localization can be found. Negative controls with a scrambled miRNA probe showed no staining (Supplementary Fig. 1).

 

Fig. 4. Qualitative and quantitative analysis of miR-375-3p expression level in the cervical spinal cord of the wobbler (WR) in comparison to wild type (WT) mouse. (A) Expression levels of miR-375-3p from presymptomatic to stationary phase of wild type (WT) and wobbler (WR) spinal cord were investigated by qPCR. Significant deregulation of miR-375-3p was detected at all developmental stages. For relative quantification of miR-375-3p expression, the 2-ΔΔCt method was conducted using the housekeeping gene U6 for normalization. Data are provided as means ± SEM. Data were tested for significance using Student's t-test. Significant differences are indicated by **p<0.01, ***p<0.001. n = 6-10. (B) Exemplary overview of in situ hybridization with miR-375-3p-probe in a cross section of the cervical spinal cord of wild type (WT) and wobbler (WR) mice at the developmental stage p20 and p40. There are no visible staining pattern differences between WT and WR. Scale bar = 1mm. (C) Cross section of the cervical spinal cord of a wild type mouse at p40. Exemplary large-scale picture of the anterior horn after in situ hybridization with miR-375-3p-probe. Arrows indicate motor neurons with a clearly visible prominent staining for miR-375-3p. Scale bar = 500 µm. All pictures were taken with a light microscope (Olympus microscope BX61VS, Japan) and a 20x objective (UPlanSApo 20x/0.4, Olympus, Japan). For counterstaining Nuclear Fast Red was used.

 

 

Discussion

 

The protein NDRG2 is mentioned to have many different functions. Among others, NDRG2 is involved in stress response [10, 12]. It was shown, that NDRG2 has a direct binding side for p53, playing a role in p53 mediated apoptosis [13, 14]. A possible mechanism for neurodegeneration was shown to be caused by an interplay between NDRG2 and a miRNA, miR-375-3p. miR-375-3p is expressed in dependence of NDRG2 expression level in N9 microglial cells causing damage to neuronal cells after secretion [23]. In contrast to that, miR-375-3p was shown to prevent apoptosis by inhibiting p53 [24]. Both molecules were already mentioned in context of familial ALS and therefore present interesting targets for further investigations [20, 25].

The aim of this study was to investigate the relevance of these two molecules for the pathomechanism of the motor neuronal degeneration in wobbler mice.

Using molecular techniques, we were able to show a differential expression of NDRG2 and miR-375-3p in the spinal cord of the wobbler mouse. Already immediately after birth, without pathological symptoms, the wobbler mouse exhibit significant deregulation of NDRG2 mRNA and miR-375-3p expression. Furthermore, we were able to demonstrate for the first time the expression of NDRG2 on mRNA as well as on protein level in motor neuronal cells of the spinal cord under physiological and pathological conditions. In previous publications NDRG2 was discussed exclusively as an astrocyte marker [17, 30]. The only exceptional case showed Mitchelmore and colleagues in 2004. In this study, NDRG2 expression could be shown in cortical pyramidal neurons under pathological conditions of Alzheimer's disease [16].

We observed a negative correlation of NDRG2 mRNA and protein expression at p0 and p20. Several publication show, that a correlation of mRNA expression and protein abundance is not always given. In Crohn’s disease, for instance, an increase of a specific gene does not correlate with the protein level [31]. Thus, analyzing both molecule levels is even more important. Possible reasons for absence of correlation are plenty, but can be resumed into two main causes: Post-transcriptional mechanisms and variations in in vivo half-lives [32]. Regulatory proteins as well as non-coding RNA, e.g. miRNA, take influence on translation by either promoting the binding of RNA and ribosome or blocking it [33]. Additionally, the translation efficiency depends on the density of ribosomes as well as the secondary structure of the RNA [33, 34]. Secondly, the turnover of protein and RNA can vary significantly and depend on different conditions [35].

Recently, the role of NDRG2 in the central nervous system has attracted a lot of attention. Pan et al. showed NDRG2 overexpressed leading to decreased intracellular ATP and NADPH generation as well as an elevated ROS-level [36]. Thus, leading to significantly decreased proliferation and increased apoptosis of hepatoma cells [36]. A contrary effect can be observed, if NDRG2 expression is inhibited [37]. Ma and colleagues showed a significantly decreased ROS level after reduced NDRG2 expression in astrocytes [37]. Therefore, a direct correlation between the expression level of NDRG2, oxidative stress and the associated apoptosis of cells can be hypothesized.

A recent publication showed an elevated ROS level in the cervical spinal cord of wobbler mice at p40 [38]. Deregulation of NAD+ metabolism through a deregulated Nmnat2 expression was shown and discussed as a reason for the elevated ROS levels. The up-regulation of NDRG2 expression in the spinal cord of the wobbler mice at p40 shown in the current study, could be another reason for increased ROS and degeneration in the tissue of this ALS model. This confirms the assumption that ALS is a multifactorial disease [39].

In several publications NDRG2 was shown to be involved in the p53 mediated apoptosis [13, 14]. Liu and colleagues also found a direct p53-binding site in intron 1 of the NDRG2 gene. This leads to a p53-dependent transactivation of NDRG2 [13]. Interestingly, they also indicated NDRG2 to be required for p53 mediated apoptosis [13]. The actual role in this process is not yet understood. In various tumor cell lines, NDRG2 was proposed to play a crucial role by inhibiting cell proliferation when overexpressed [40, 41]. Since motor neurons do not proliferate but show overexpression of NDRG2 in wobbler mouse, a different approach concerning the involvement of NDRG2 in the p53 mediated apoptosis should be considered. An overexpression of NDRG2 is known to increase the Bax/Bcl-2 ratio, which is determining cell death while both Bax and Bcl-2 are regulated by p53 [42–44]. In astrocytes, Li et al. showed a pro-apoptotic effect of Bax after overexpression of NDRG2,  whereas Bcl-2 was uninfluenced [43]. This indicates, that NDRG2 might influence downstream targets of p53 and therefore contribute to the p53 mediated apoptosis.

In accordance with the here performed in situ hybridization, Bhinge et al. showed miR-375-3p to be expressed in motor neurons. Additionally different studies showed that miR-375-3p is essential for motor neuron development [24, 25]. Furthermore, both Binghe et al. and De Santis et al. showed in different disease models that down-regulation of miR-375-3p leads to an increase in proapoptotic factors, such as p53 [20, 24, 25]. Farther, Wang et al. demonstrated that miR-375-3p suppresses epithelial apoptosis by inhibiting JAK2/Stat3-signaling pathway [46]. Thus, miR-375-3p appears to regulate various target structures that intervene at different sites in an apoptotic pathway. In this study, we could show a significant decrease of miR-375-3p within the spinal cord of wobbler mice at the stable clinical phase (p40), whereas at p0 and p20 a significant increase of miR-375-3p was observed. According to Bhinge et al., De Santis et al., and Wang et al., miR-375-3p is able to protect cells from apoptosis, which might explain the late start of symptoms in the wobbler mouse. However, a significant down regulation of miR-375-3p could lead to an increase of p53 expression and thus apoptosis [21, 22, 25]. It is also known that p53 is increased in both ALS patients and the wobbler mouse [15, 47]. Here, our results complement the interaction of the two molecules by elucidating the expression levels of miR-375-3p. Therefore, miR-375-3p seem to have an anti-apoptotic function in motor neurons of wobbler mice. Otherwise, it was shown that this miRNA can have a negative effect on neuronal cells. Tang and colleagues have revealed that overexpression of NDRG2 in N9 microglia leads to increased secretion of miR-375-3p. These miRNA-filled vesicles have a negative influence on neuronal cells in vitro in form of increased degeneration [23]. This suggests an apoptotic effect of miR-375-3p.

Some miRNAs are known to exert opposing effects depending on their expression locus. miRNA-204, for example, shows contrary effects in form of tumor suppressor or oncomiR in different prostate cancer cell lines [48, 49].

Since both molecules, NDRG2 and miR-375-3p, have a connection to p53, we used published data and our results to establish a model for a putative pro-apoptotic mechanism in the spinal cord of the wobbler mice (Fig. 5). Responding to stress signals like elevated ROS level [38], p53 is activated, and triggers apoptosis by initiating the expression of pro-apoptotic genes like Bax and Apaf-1 [50, 51]. P53 is known to be highly increased in the spinal cord of the wobbler mouse as well as ALS patients [15, 47]. MiR-375-3p inhibits the expression of p53, thereby affecting NDRG2 transcription [13, 24]. In addition to that, Pan et al. showed that an increased NDRG2 level lead to an elevated ROS level [36]. Fig. 5 shows a simplified mechanism that might occur in the spinal cord of wobbler mice at the clinical stage leading to increased apoptosis. miR-375-3p is decreased at this stage. Deregulation of miR-375-3p leads to an inefficient inhibition of translation of p53 compared to wildtype resulting in increased p53. This leads to an increased protein level of p53 and thus to an increased NDRG2 transcription in the spinal cord of the wobbler mouse. Finally, the overexpression of NDRG2 protein leads to elevated ROS, and thus maintains the vicious circle.

 

Fig. 5. Putative pro-apoptotic mechanism in the spinal cord of the wobbler mouse at the clinical stage p40. P53 is known to bind to NDRG2 and induce its transcription. Therefore NDRG is associated with the apoptotic pathway of p53 [13]. MiR-375-3p inhibits the translation of p53 by directly targeting its mRNA [24]. Additionally, p53 expression is known to be increased in the spinal cord of the wobbler mice [15]. NDRG2 mRNA and protein expression was shown to be upregulated in the spinal cord of wobbler mice at p40. Since the expression of miR-375-3p is decreased, translation of p53 is not effectively inhibited, which affects NDRG2 transcription. High level of NDRG2 can lead to elevated ROS level, which in turn can activate p53. The increased p53 and NDRG2 level lead to increased apoptosis in the spinal cord of the wobbler mice.

 

Based on this model we can discuss the results for the other stages p0 and p20. At p0 the phenotype of the wobbler mouse is unobtrusive. Here we could show a decreased NDRG2 mRNA expression without differences on protein level. Furthermore, we could show an increased miR-375-3p expression, that could lead to decreased p53 protein expression. Thus, pathologic mechanisms would not overweight. At the onset of disease at p20, we could show an increased NDRG2 mRNA and miR375-3p expression, whereas protein expression of NDRG2 was decreased. This results in a still efficient inhibition of p53 translation as well as normal ROS level. The increased miR-375-3p could be a protective mechanism, preventing the cell to induce apoptosis by decreasing p53 and therefore maintaining the level of NDRG2. Additionally, a negative correlation between NDRG2 mRNA and protein level is obvious. This could raise the question of NDRG2 specific miRNAs in the protective mechanism.

 

 

Conclusion

 

The present study has revealed NDRG2 and miR-375-3p to be promising targets for research in regard to the pathogenesis of sALS in the wobbler mouse model. Here a deregulation of the tumor suppressor NDRG2 as well as miR-375-3p during the progression of disease in the spinal cord could be shown. Elevated NDRG2 levels lead to increased ROS formation, which serve as a stress signal for the activation of p53. Due to downregulation of miR-375-3p, inefficient inhibition of p53 leads to overexpression of NDRG2, increasing ROS production and maintaining a vicious cycle.

 

 

Acknowledgements

 

The authors gratefully acknowledge A. Lodwig, C. Grzelak, and A. Harbecke for technical assistance, as well as A. Lenz for secretarial work. We acknowledge support by the DFG Open Access Publication Funds of the Ruhr-Universität Bochum.

This study was supported also by FoRUM - F874N-2016 (Ruhr-University Bochum) to V.M. and C.T., the Heinrich and Alma Vogelsang scholarship to M.R., the German Federal Ministry of Education and Research (WTZ with Brasil, FKZ 01DN14023) and PURE (233-1.08.03.03-031-68079, Ministry of Culture and Science Northrhine Westfalia) to C.M. and K.M..

 

 

Disclosure Statement

 

The authors state that there are no personal or institutional conflicts of interest.

 

 

References

 

1 Falconer DS: Wobbler (wr). Mouse News Lett 1956;15:22.

 

2 Cozzolino M, Carrì MT: Mitochondrial dysfunction in ALS. Prog Neurobiol 2012;97:54-66.
https://doi.org/10.1016/j.pneurobio.2011.06.003
PMID: 21827820

 

3 Kiernan MC, Vucic S, Cheah BC, Turner MR, Eisen A, Hardiman O, Burrell JR, Zoing MC: Amyotrophic lateral sclerosis. Lancet 2011;377:942-955.
https://doi.org/10.1016/S0140-6736(10)61156-7
PMID: 21296405

 

4 Duchen LW, Strich SJ: An hereditary motor neurone disease with progressive denervation of muscle in the mouse. J Neurol Neurosurg Psychiatry 1968;31:535-542.
https://doi.org/10.1136/jnnp.31.6.535
PMID: 5709840 PMCid:PMC496420

 

5 Moser JM, Bigini P, Schmitt-John T: The wobbler mouse, an ALS animal model. Mol Genet Genomics 2013;288:207-229.
https://doi.org/10.1007/s00438-013-0741-0
PMID: 23539154 PMCid:PMC3664746

 

6 Schmitt-John T, Drepper C, Mussmann A, Hahn P, Kuhlmann M, Thiel C, Hafner M, Lengeling A, Heimann P, Jones JM, Meisler MH, Jockusch H: Mutation of Vps54 causes motor neuron disease and defective spermiogenesis in the wobbler mouse. Nat Genet 2005;37:1213-1215.
https://doi.org/10.1038/ng1661
PMID: 16244655

 

7 Boillée S, Peschanski M, Junier MP: The Wobbler Mouse. Mol Neurobiol 2003;28:65-106.
https://doi.org/10.1385/MN:28:1:65
PMID: 14514986

 

8 Ott B, Dahlke C, Meller K, Napirei M, Schmitt-John T, Brand-Saberi B, Theiss C, Saberi D: Implementation of a manual for working with wobbler mice and criteria for discontinuation of the experiment. Ann Anat 2015;200:118-124.
https://doi.org/10.1016/j.aanat.2015.03.007
PMID: 25929815

 

9 Saberi D, Ott B, Dahlke C, Matschke V, Schmitt-John T, Theiss C: The Spatiotemporal Pattern of Degeneration in the Cerebellum of the Wobbler Mouse. J Neuropathol Exp Neurol 2016;75:347-357.
https://doi.org/10.1093/jnen/nlw005
PMID: 26945034

 

10 Ichikawa T, Nakahata S, Tamura T, Manachai N, Morishita K: The loss of NDRG2 expression improves depressive behavior through increased phosphorylation of GSK3β. Cell Signal 2015;27:2087-2098.
https://doi.org/10.1016/j.cellsig.2015.07.012
PMID: 26208882

 

11 Melotte V, Qu X, Ongenaert M, van Criekinge W, Bruine AP, Baldwin HS, van Engeland M: The N-myc downstream regulated gene (NDRG) family: diverse functions, multiple applications. FASEB J 2010;24:4153-4166.
https://doi.org/10.1096/fj.09-151464
PMID: 20667976

 

12 Yao L, Zhang J, Liu X: NDRG2: a Myc-repressed gene involved in cancer and cell stress. Acta Biochim Biophys Sin (Shanghai) 2008;40:625-635.
https://doi.org/10.1111/j.1745-7270.2008.00434.x
PMID: 18604454

 

13 Liu N, Wang L, Li X, Yang Q, Liu X, Zhang J, Wu Y, Ji S, Zhang Y, Yang A, Han H, Yao L: N-Myc downstream-regulated gene 2 is involved in p53-mediated apoptosis. Nucleic Acids Res 2008;36:5335-5349.
https://doi.org/10.1093/nar/gkn504
PMID: 18689861 PMCid:PMC2532733

 

14 Ma J, Liu W, Yan X, Wang Q, Zhao Q, Xue Y, Ren H, Wu L, Cheng Y, Li S, Miao L, Yao L, Zhang J: Inhibition of Endothelial Cell Proliferation and Tumor Angiogenesis by Up-Regulating NDRG2 Expression in Breast Cancer Cells. PLoS One 2012;7:e32368.
https://doi.org/10.1371/journal.pone.0032368
PMID: 22393400 PMCid:PMC3290656

 

15 Eve DJ, Dennis JS, Citron BA: Transcription factor p53 in degenerating spinal cords. Brain Res 2007;1150:174-181.
https://doi.org/10.1016/j.brainres.2007.02.088
PMID: 17434459

 

16 Mitchelmore C, Buchmann-Moller S, Rask L, West MJ, Troncoso JC, Jensen NA: NDRG2: a novel Alzheimer's disease associated protein. Neurobiol Dis 2004;16:48-58.
https://doi.org/10.1016/j.nbd.2004.01.003
PMID: 15207261

 

17 Okuda T, Kokame K, Miyata T: Differential expression patterns of NDRG family proteins in the central nervous system. J Histochem Cytochem 2008;56:175-182.
https://doi.org/10.1369/jhc.7A7323.2007
PMID: 17998568 PMCid:PMC2324175

 

18 Kim BM, You MH, Chen CH, Lee S, Hong Y, Hong Y, Kimchi A, Zhou XZ, Lee TH: Death-associated protein kinase 1 has a critical role in aberrant tau protein regulation and function. Cell Death Dis 2014;5:e1237-e1237.
https://doi.org/10.1038/cddis.2014.216
PMID: 24853415 PMCid:PMC4047864

 

19 You MH, Kim BM, Chen CH, Begley MJ, Cantley LC, Lee TH: Death-associated protein kinase 1 phosphorylates NDRG2 and induces neuronal cell death. Cell Death Differ 2017;24:238-250.
https://doi.org/10.1038/cdd.2016.114
PMID: 28141794 PMCid:PMC5299707

 

20 Lagier-Tourenne C, Polymenidou M, Hutt KR, Vu AQ, Baughn M, Huelga SC, Clutario KM, Ling SC, Liang TY, Mazur C, Wancewicz E, Kim AS, Watt A, Freier S, Hicks GG, Donohue JP, Shiue L, Bennett CF, Ravits J, Cleveland DW, et al.: Divergent roles of ALS-linked proteins FUS/TLS and TDP-43 intersect in processing long pre-mRNAs. Nat Neurosci 2012;15:1488-1497.
https://doi.org/10.1038/nn.3230
PMID: 23023293 PMCid:PMC3586380

 

21 Kwiatkowski TJ, Bosco DA, LeClerc AL, Tamrazian E, Vanderburg CR, Russ C, Davis A, Gilchrist J, Kasarskis EJ, Munsat T, Valdmanis P, Rouleau GA, Hosler BA, Cortelli P, de Jong PJ, Yoshinaga Y, Haines JL, Pericak-Vance MA, Yan J, Ticozzi N, et al.: Mutations in the FUS/TLS Gene on Chromosome 16 Cause Familial Amyotrophic Lateral Sclerosis. Science 2009;323:1205-1208.
https://doi.org/10.1126/science.1166066
PMID: 19251627

 

22 Bartel DP: MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281-297.
https://doi.org/10.1016/S0092-8674(04)00045-5
PMID: 14744438

 

23 Tang L, Wu Y, Fang C, Qu P, Gao Z: NDRG2 promoted secreted miR-375 in microvesicles shed from M1 microglia, which induced neuron damage. Biochem Biophys Res Commun 2015;469:392-398.
https://doi.org/10.1016/j.bbrc.2015.11.098
PMID: 26631961

 

24 Bhinge A, Bithell A, Buckley NJ, Namboori SC, Soldati C, Stanton LW: MiR-375 is Essential for Human Spinal Motor Neuron Development and May Be Involved in Motor Neuron Degeneration. Stem Cells 2016;34:124-134.
https://doi.org/10.1002/stem.2233
PMID: 26507573

 

25 De Santis R, Santini L, Colantoni A, Peruzzi G, de Turris V, Alfano V, Bozzoni I, Rosa A: FUS Mutant Human Motoneurons Display Altered Transcriptome and microRNA Pathways with Implications for ALS Pathogenesis. Stem Cell Rep 2017;9:1450-1462.
https://doi.org/10.1016/j.stemcr.2017.09.004
PMID: 28988989 PMCid:PMC5830977

 

26 Livak KJ, Schmittgen TD: Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2−ΔΔCT Method. Methods 2001;25:402-408.
https://doi.org/10.1006/meth.2001.1262
PMID: 11846609

 

27 Barkovits K, Linden A, Galozzi S, Schilde L, Pacharra S, Mollenhauer B, Stoepel N, Steinbach S, May C, Uszkoreit J, Eisenacher M, Marcus K: Characterization of Cerebrospinal Fluid via Data-Independent Acquisition Mass Spectrometry. J Proteome Res 2018;17:3418-3430.
https://doi.org/10.1021/acs.jproteome.8b00308
PMID: 30207155

 

28 Aring L, Steinbach S, Marcus K, May C: Isolation of Distinct Types of Neurons from Fresh Brain Tissue Using Laser Microdissection in Combination with High-Performance Liquid Chromatography-Mass Spectrometry; in Murray GI (ed): Laser Capture Microdissection. New York, NY, Springer New York, 2018, pp 247-260.
https://doi.org/10.1007/978-1-4939-7558-7_14
PMID: 29344865

 

29 Boulkroun S, Fay M, Zennaro M-C, Escoubet B, Jaisser F, Blot-Chabaud M, Farman N, Courtois-Coutry N: Characterization of rat NDRG2 (N-Myc downstream regulated gene 2), a novel early mineralocorticoid-specific induced gene. J Biol Chem 2002;277:31506-31515.
https://doi.org/10.1074/jbc.M200272200
PMID: 12072429

 

30 Flügge G, Araya-Callis C, Garea-Rodriguez E, Stadelmann-Nessler C, Fuchs E: NDRG2 as a marker protein for brain astrocytes. Cell Tissue Res 2014;357:31-41.
https://doi.org/10.1007/s00441-014-1837-5
PMID: 24816982 PMCid:PMC4077251

 

31 Taquet N, Dumont S, Vonesch JL, Hentsch D, Reimund JM, Muller CD: Differential between Protein and mRNA Expression of CCR7 and SSTR5 Receptors in Crohn's Disease Patients. Mediators Inflamm 2009;2009:285812.
https://doi.org/10.1155/2009/285812
PMID: 20150960 PMCid:PMC2817506

 

32 Greenbaum D, Colangelo C, Williams K, Gerstein M: Comparing protein abundance and mRNA expression levels on a genomic scale. Genome Biol 2003;4:117.
https://doi.org/10.1186/gb-2003-4-9-117
PMID: 12952525 PMCid:PMC193646

 

33 Maier T, Güell M, Serrano L: Correlation of mRNA and protein in complex biological samples. FEBS Lett 2009;583:3966-3973.
https://doi.org/10.1016/j.febslet.2009.10.036
PMID: 19850042

 

34 MacKay VL, Li X, Flory MR, Turcott E, Law GL, Serikawa KA, Xu XL, Lee H, Goodlett DR, Aebersold R, Zhao LP, Morris DR: Gene expression analyzed by high-resolution state array analysis and quantitative proteomics: response of yeast to mating pheromone. Mol Cell Proteomics 2004;3:478-489.
https://doi.org/10.1074/mcp.M300129-MCP200
PMID: 14766929

 

35 Glickman MH, Ciechanover A: The ubiquitin-proteasome proteolytic pathway: destruction for the sake of construction. Physiol Rev 2002;82:373-428.
https://doi.org/10.1152/physrev.00027.2001
PMID: 11917093

 

36 Pan T, Zhang M, Zhang F, Yan G, Ru Y, Wang Q, Zhang Y, Wei X, Xu X, Shen L, Zhang J, Wu K, Yao L, Li X: NDRG2 overexpression suppresses hepatoma cells survival during metabolic stress through disturbing the activation of fatty acid oxidation. Biochem Biophys Res Commun 2017;483:860-866.
https://doi.org/10.1016/j.bbrc.2017.01.018
PMID: 28069379

 

37 Ma YL, Zhang LX, Liu GL, Fan Y, Peng Y, Hou WG: N-Myc Downstream-Regulated Gene 2 (Ndrg2) Is Involved in Ischemia-Hypoxia-Induced Astrocyte Apoptosis: a Novel Target for Stroke Therapy. Mol Neurobiol 2017;54:3286-3299.
https://doi.org/10.1007/s12035-016-9814-5
PMID: 27154863

 

38 Röderer P, Klatt L, John F, Theis V, Winklhofer KF, Theiss C, Matschke V: Increased ROS Level in Spinal Cord of Wobbler Mice due to Nmnat2 Downregulation. Mol Neurobiol 2018;55:8414-8424.
https://doi.org/10.1007/s12035-018-0999-7
PMID: 29549647

 

39 Eisen A: Amyotrophic lateral sclerosis is a multifactorial disease. Muscle Nerve 1995;18:741-752.
https://doi.org/10.1002/mus.880180711
PMID: 7783764

 

40 Deng Y, Yao L, Chau L, Ng SS, Peng Y, Liu X, Au WS, Wang J, Li F, Ji S, Han H, Nie X, Li Q, Kung HF, Leung SY, Lin MC: N-Myc downstream-regulated gene 2(NDRG2) inhibits glioblastoma cell proliferation. Int J Cancer 2003;106:342-347.
https://doi.org/10.1002/ijc.11228
PMID: 12845671

 

41 Zhang J, Liu J, Li X, Li F, Wang L, Zhang J, Liu X, Shen L, Liu N, Deng Y, Yang A, Han H, Zhao M, Yao L.: The physical and functional interaction of NDRG2 with MSP58 in cells. Biochem Biophys Res Commun 2007;352:6-11.
https://doi.org/10.1016/j.bbrc.2006.10.141
PMID: 17109818

 

42 Hemann MT, Lowe SW: The p53-Bcl-2 connection. Cell Death Differ 2006;13:1256-1259.
https://doi.org/10.1038/sj.cdd.4401962
PMID: 16710363 PMCid:PMC4590992

 

43 Li Y, Xu N, Cai L, Gao Z, Shen L, Zhang Q, Hou W, Zhong H, Wang Q, Xiong L: NDRG2 Is a Novel p53-Associated Regulator of Apoptosis in C6-Originated Astrocytes Exposed to Oxygen-Glucose Deprivation. PLoS One 2013;8:e57130.
https://doi.org/10.1371/journal.pone.0057130
PMID: 23451161 PMCid:PMC3579777

 

44 Yang J, Li Y, Wu L, Zhang Z, Han T, Guo H, Jiang N, Tao K, Ti Z, Liu X, Yao L, Dou K: NDRG2 in rat liver regeneration: Role in proliferation and apoptosis: NDRG2 regulates rat liver regeneration. Wound Repair Regen 2010;18:524-531.
https://doi.org/10.1111/j.1524-475X.2010.00614.x
PMID: 20840522

 

45 Micheli F, Palermo R, Talora C, Ferretti E, Vacca A, Napolitano M: Regulation of proapoptotic proteins Bak1 and p53 by miR-125b in an experimental model of Alzheimer's disease: Protective role of 17β-estradiol. Neurosci Lett 2016;629:234-240.
https://doi.org/10.1016/j.neulet.2016.05.049
PMID: 27235580

 

46 Wang T, Chen D, Wang P, Xu Z, Li Y: miR-375 prevents nasal mucosa cells from apoptosis and ameliorates allergic rhinitis via inhibiting JAK2/STAT3 pathway. Biomed Pharmacother 2018;103:621-627.
https://doi.org/10.1016/j.biopha.2018.04.050
PMID: 29677549

 

47 Ranganathan S, Bowser R: p53 and Cell Cycle Proteins Participate in Spinal Motor Neuron Cell Death in ALS. Open Pathol J 2010;4:11-22.
https://doi.org/10.2174/1874375701004010011
PMID: 21572928 PMCid:PMC3092395

 

48 Ding M, Lin B, Li T, Liu Y, Li Y, Zhou X, Miao M, Gu J, Pan H, Yang F, Li T, Liu XY, Li R: A dual yet opposite growth-regulating function of miR-204 and its target XRN1 in prostate adenocarcinoma cells and neuroendocrine-like prostate cancer cells. Oncotarget 2015;6:7686-7700.
https://doi.org/10.18632/oncotarget.3480
PMID: 25797256

 

49 Turner DP, Findlay VJ, Moussa O, Semenchenko VI, Watson PM, LaRue AC, Desouki MM, Fraig M, Watson DK: Mechanisms and functional consequences of PDEF protein expression loss during prostate cancer progression. Prostate 2011;71:1723-1735.
https://doi.org/10.1002/pros.21389
PMID: 21446014 PMCid:PMC3128180

 

50 Leu JI, Dumont P, Hafey M, Murphy ME, George DL: Mitochondrial p53 activates Bak and causes disruption of a Bak-Mcl1 complex. Nat Cell Biol 2004;6:443-450.
https://doi.org/10.1038/ncb1123
PMID: 15077116

 

51 Moll UM, Wolff S, Speidel D, Deppert W: Transcription-independent pro-apoptotic functions of p53. Curr Opin Cell Biol 2005;17:631-636.
https://doi.org/10.1016/j.ceb.2005.09.007
PMID: 16226451