Next Article in Journal
Inhibitors of Rho/MRTF/SRF Transcription Pathway Regulate Mitochondrial Function
Next Article in Special Issue
The Role of the NLRP3 Inflammasome in the Molecular and Biochemical Mechanisms of Cervical Ripening: A Comprehensive Review
Previous Article in Journal
Exploring the Prospective Role of Propolis in Modifying Aging Hallmarks
Previous Article in Special Issue
Biomarkers of Affective Dysregulation Associated with In Utero Exposure to EtOH
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Impact of SLC2A8 RNA Interference on Glucose Uptake and the Transcriptome of Human Trophoblast Cells

by
Aleksandra Lipka
1,*,
Łukasz Paukszto
2,
Victoria C. Kennedy
3,
Amelia R. Tanner
3,
Marta Majewska
4 and
Russell V. Anthony
3,*
1
Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-045 Olsztyn, Poland
2
Department of Botany and Nature Protection, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-727 Olsztyn, Poland
3
College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
4
Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
*
Authors to whom correspondence should be addressed.
Cells 2024, 13(5), 391; https://doi.org/10.3390/cells13050391
Submission received: 22 January 2024 / Revised: 14 February 2024 / Accepted: 22 February 2024 / Published: 24 February 2024
(This article belongs to the Special Issue Signaling Pathways in Pregnancy)

Abstract

:
While glucose is the primary fuel for fetal growth, the placenta utilizes the majority of glucose taken up from the maternal circulation. Of the facilitative glucose transporters in the placenta, SLC2A8 (GLUT8) is thought to primarily function as an intracellular glucose transporter; however, its function in trophoblast cells has not been determined. To gain insight into the function of SLC2A8 in the placenta, lentiviral-mediated RNA interference (RNAi) was performed in the human first-trimester trophoblast cell line ACH-3P. Non-targeting sequence controls (NTS RNAi; n = 4) and SLC2A8 RNAi (n = 4) infected ACH-3P cells were compared. A 79% reduction in SLC2A8 mRNA concentration was associated with an 11% reduction (p ≤ 0.05) in ACH-3P glucose uptake. NTS RNAi and SLC2A8 RNAi ACH-3P mRNA were subjected to RNAseq, identifying 1525 transcripts that were differentially expressed (|log2FC| > 1 and adjusted p-value < 0.05), with 273 transcripts derived from protein-coding genes, and the change in 10 of these mRNAs was validated by real-time qPCR. Additionally, there were 147 differentially expressed long non-coding RNAs. Functional analyses revealed differentially expressed genes involved in various metabolic pathways associated with cellular respiration, oxidative phosphorylation, and ATP synthesis. Collectively, these data indicate that SLC2A8 deficiency may impact placental uptake of glucose, but that its likely primary function in trophoblast cells is to support cellular respiration. Since the placenta oxidizes the majority of the glucose it takes up to support its own metabolic needs, impairment of SLC2A8 function could set the stage for functional placental insufficiency.

1. Introduction

The successful outcome of pregnancy requires the functional integration of three compartments: the maternal, placental, and fetal compartments. This is especially true when one considers the umbilical uptake of nutrients in support of fetal growth. Glucose, the primary energy source for fetal and placental oxidative processes [1], is entirely derived from the maternal circulation throughout most of gestation, as fetal gluconeogenesis does not begin until the very near term [2,3,4,5]. Glucose is transferred across the placenta, from maternal to fetal circulation, down a maternal–fetal glucose concentration gradient via facilitative diffusion [6]. Beyond the maternal–fetal glucose concentration gradient, placental metabolism, blood flow, and the availability and activity of specific glucose transporters can impact the efficiency of placental glucose transport [1,7].
Fourteen different facilitative glucose transporters (GLUTs) have been identified, which differ in terms of substrate specificity, kinetics, distribution, location, and regulatory mechanisms [8]. Within the human placenta, GLUT1 (SLC2A1), GLUT3 (SLC2A3), GLUT4 (SLC2A4), GLUT8 (SLC2A8), GLUT9 (SLC2A9), GLUT10 (SLC2A10), and GLUT12 (SLC2A12) have been detected [6,9]. Due to its abundance and distribution, SLC2A1 is considered the primary glucose transporter within the placenta [10,11], but is supported by SLC2A3, which has a higher affinity and transport capacity [12], at least during the first half of gestation [13,14] or in some cases of intrauterine growth restriction [15].
The maternal to fetal glucose gradient, which drives facilitated diffusion, results in part from the extensive oxidation of glucose by the placenta. The placenta is a highly metabolic organ, utilizing 80 and 72% of the glucose taken up by the uterus at mid- and late-gestation, respectively [16,17]. In contrast to SLC2A1 and SLC2A3, which are involved in glucose uptake and transfer to the fetus, SLC2A8 (GLUT8) is a class III glucose transporter, primarily localized in endosomes, lysosomes, and endoplasmic reticulum membranes [18], and is thought to catalyze hexose transport across intracellular membranes. However, in the mouse blastocyst, in response to insulin, SLC2A8 localization shifts to the plasma membrane, and is believed to be responsible for insulin-stimulated glucose uptake by the blastocyst [19,20]. Slc2A8−/− mice were viable and developed normally [21,22], but did exhibit alterations in the brain, heart, and sperm cells. The reduction in sperm cell motility with Slc2A8−/− males appears to be associated with reduced mitochondrial membrane potential and ATP availability in sperm [22], which fits with the role of SLC2A8 as an intracellular transporter. While these results do not support an important role for SLC2A8 in placental glucose transport, culturing murine embryos with Slc2A8 antisense RNA resulted in increased blastocyst apoptosis and reduced viability [20], and Limesand et al. [23] reported diminished placental expression of SLC2A8 in a sheep model of placental insufficiency resulting in intrauterine growth restriction (IUGR). As the function of SLC2A8 in placental glucose uptake and metabolism has not been fully interrogated, we hypothesized that reduction in SLC2A8 in human trophoblast cells could reduce glucose uptake and alter metabolic processes important in glucose oxidation. Therefore, our objective was to use lentiviral-mediated RNA interference (RNAi) to determine the impact of SLC2A8 deficiency on the trophoblast transcriptome and glucose uptake.

2. Materials and Methods

All procedures were approved by the Colorado State University Institutional Biosafety Committee (17-039B).

2.1. Lentivirus Vector Construction and Virus Generation

To target SLC2A8 mRNA for RNA interference, four shRNAs homologous to human SLC2A8 (Table 1) were initially inserted into pLKO.1 (plasmid #10878; Addgene, Cambridge, MA, USA). The human U6 promoter and SLC2A8 shRNA were then shuttled into pLentiLox3.7 (pLL3.7 plasmid #11795; Addgene, Cambridge, MA, USA), replacing the mouse U6 promoter, as extensively described in Baker et al. [24]. All constructs were subjected to capillary sequencing (QuintaraBio, Bay Area, CA, USA) to verify authenticity. The non-targeting sequence (NTS) control LL3.7 plasmid was previously described in Jeckel et al. [25].
The lentivirus was generated as described previously [24,26,27]. Briefly, 293FT (Thermo Fisher Scientific, Waltham, MA, USA) cells were grown to 70–80% confluence in a 15 cm tissue culture plate in high-glucose DMEM supplemented with 10% v/v fetal bovine serum (FBS; Peak Serum, Bradenton, FL, USA) and 1% Penicillin/Streptomycin/Amphotericin (PSA; Corning Life Sciences, New York, NY, USA) at 37 °C and 5% CO2. To generate second-generation lentiviral particles, pLL3.7-NTS, and pLL3.7-SLC2A8 plasmids, psPAX2 packaging plasmid (Addgene, Cambridge, MA, USA), and pMD2.G envelope plasmid (Addgene, Cambridge, MA, USA) were used. For each 15 cm plate, the transfection mix was prepared as follows: 8.82 μg of LL3.7 (NTS- or SLC2A8-shRNA), 6.66 μg of psPAX2, and 2.70 μg of pMD2.G were mixed with 180 μL of polyfect transfection reagent (Qiagen, Hilden, Germany), and the final volume was brought up to 675 μL using serum and antibiotic-free DMEM media. The transfection mixture was added to 293FT cells along with 15 mL of complete medium. After 24 h of incubation in the transfection reagent, the medium was aspirated, and fresh complete DMEM medium was added. Seventy-two hours after transfection, cell culture supernatants were collected and ultracentrifuged over a 20% w/v sucrose cushion at 47,000× g for 2 h at 4 °C. After ultracentrifugation, lentiviral pellets were resuspended in 1xPBS and stored in aliquots at −80 °C. Aliquots of the lentivirus were titered as described previously [24].

2.2. In Vitro SLC2A8 RNAi of Human Trophoblast Cells

The human first-trimester trophoblast cell line ACH-3P was used for this study to target SLC2A8 mRNA by RNAi. ACH-3P are hybrid cells generated by the fusion of primary first-trimester human trophoblast cells (12 weeks of gestation) with AC1-1 cells, a human choriocarcinoma cell line [28]. ACH-3P cells were cultured in Ham’s F-12 medium (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% v/v FBS and 1% PSA at 37 °C and 5% CO2.
ACH-3P cells were infected with either LL3.7-NTS (NTS RNAi) or the four LL3.7-SLC2A8 lentiviruses at a multiplicity of infection (MOI) of 100 and 500 transducing units per cell in a 96-well plate. Transfected cells were expanded until near-confluence was obtained in a 15 cm tissue culture plate. The concentration of SLC2A8 mRNA in transfected ACH-3P cells was assessed by quantitative real-time reverse transcriptase PCR (qPCR).

2.3. RNA Extraction and qPCR

Total RNA was isolated from cell pellets using a RNeasy Kit (Qiagen, Hilden, Germany) according to the manufacturer’s protocol. RNA quality and concentration, measured by the 260/280 nm absorbance ratio, were assessed using a plate reader (BioTek, Winooski, VT, USA), and samples were stored at −80 °C until use. Complementary DNA was generated from 1 μg of total RNA using an iScript™ Reverse Transcription Supermix (BioRad, Hercules, CA, USA). Quantitative real-time PCR was conducted using iQ™ SYBR® Green Supermix (BioRad, Hercules, CA, USA). The primer sequences used are presented in Table 2. For analysis, a PCR product for each gene was generated and cloned using the StrataClone PCR Cloning Kit (Agilent Technologies, Santa Clara, CA, USA). Each PCR product was sequenced to verify the amplification of the correct mRNA (GeneWIZ, Azenta Life Sciences, Burlington, MA, USA). By amplifying the PCR product from each gene’s plasmid, a standard curve ranging from 1 × 102 to 1 × 10−6 pg was generated. The starting quantity (pg) of each mRNA was normalized to the starting quantity of ribosomal protein S15 (RPS15). Control (NTS RNAi) and SLC2A8-shRNA treatments (SLC2A8 RNAi) were compared by unpaired Students t-test, with p < 0.05 considered as statistically significant.

2.4. Glucose Uptake Assay

Glucose uptake assay was performed with a commercially available kit (Dojindo, Rockville, MD, USA) according to the manufacturer’s protocol. Briefly, NTS RNAi and SLC2A8 RNAi ACH-3P cells were seeded in 10 replicates on a 96-well plate (black with clear bottom) with a seeding density of 10,000 cells per well and incubated overnight in 100 µL of regular Ham’s F-12 medium. The next day, the culture medium was removed and cells were washed twice with pre-warmed glucose- and serum-free medium (Gibco, Thermo Fisher Scientific, Waltham, MA, USA). Then, pre-warmed glucose- and serum-free medium was added and the plate was incubated at 37 °C for 15 min in a 5% CO2 incubator. Next, the medium was removed and a pre-warmed solution of glucose probe (fluorescent 2-deoxy-d-glucose) was added and incubated for 15 min (37 °C and 5% CO2). After incubation and probe removal, cells were washed twice with ice-cold WI solution and then incubated with an ice-cold WI solution at room temperature for 5 min. WI solution was exchanged with ice-cold WI solution and fluorescence was measured on a micro-plate reader (BioTek, Winooski, VT, USA) with excitation and emission wavelengths set to 360/40 and 460/40, respectively.

2.5. RNAseq and Data Analysis

Assessment of the RNA samples’ integrity and quality, library generation, and RNA-Seq (2 × 150 cycles, 80 million paired-end reads/sample; NovSeq6000, Illumina, San Diego, CA, USA) was conducted by the Genomics Shared Resource Core Facility, University of Colorado Anschutz Medical Campus (Aurora, CO, USA). The sequencing data from this study have been submitted to the European Nucleotide Archive under accession no. PRJEB71894 (https://www.ebi.ac.uk/ena; accessed on 16 January 2024).

2.6. Quality Control and Mapping Processes

The raw paired-end reads quality was assessed using the FastQC software v. 0.11.7 (www.bioinformatics.babraham.ac.uk, accessed on 25 April 2023). Preprocessing with Trimmomatic software v. 0.32 [29] included removal of Illumina adaptors and poly(A) stretches, exclusion of low-quality reads (Phred cutoff score ≤ 20; calculated on both ends of reads and with 10bp frameshift), and trimming of reads to equal length of 90 bp. Then, STAR software v. 2.4 (https://github.com/alexdobin/STAR, accessed on 25 April 2023) was used to map cleaned paired-end reads to the reference soft-masked human genome (Homo_sapiens.GRCh38) with ENSEMBL/GENCODE annotation (Homo_sapiens.GRCh38.105.gtf). Conversion of BAM to SAM format was performed using Samtools v. 1.12 software [30]. Next, to remove multi-mapped reads and retain uniquely aligned reads in the SAM file, the Picard tool was used. StringTie v. 1.3.3 (https://ccb.jhu.edu/software/stringtie, accessed on 28 April 2023) was applied to obtain new annotations by merging an Ensembl GTF file with reads mapped to the reference genome [31]. Count expression values were estimated by ballgown v 2.34.0 software [32] and a prepDE.py Python script (stringtie module). All transcript sequences were extracted to the FASTA file using a gffread script (https://github.com/gpertea/gffread, accessed on 6 May 2023). Memory intensive processes such as mapping, SAM to BAM conversion, and expression level calculation were performed at the Regional IT Center of University of Warmia and Mazury in Olsztyn, Poland.

2.7. Transcriptome Profiling of Protein-Coding Genes

Transcriptome profiling was performed as previously described [33,34,35]. Briefly, to obtain the stringent results of differentially expressed transcribed active regions (DE-TARs), the count gene expression matrix was calculated by the ballgown statistical method [32]. The changes in gene expression levels were considered significant when statistical test values (adjusted p-value) were lower than 0.05 and logarithmic fold change was lower than −1 or higher than 1. According to GENCODE annotation, DE-TARs were divided into protein-coding genes (DEGs), long non-coding RNAs (DElncRNAs), and other non-coding RNAs (ncRNAs). The research focused mostly on two categories (DEGs and DElncRNAs) and expression values between these transcripts. Candidate DEGs and DElncRNAs were visualized in a volcano and heatmap plots with gplots and circlize Bioconductor R packages (http://www.r-project.org/, accessed on 18 May 2023). The correlation between coding and lncRNA transcripts was calculated using Pearson correlation metric implemented in the rcorr function (Hmisc R package). The obtained DEGs were annotated by the Gene Ontology (GO) and KEGG pathway database using g.Profiler [36] v.0.2.2 software with the g:SCS algorithm (p < 0.05).

2.8. qPCR Validation of the RNAseq Results

Quantitative PCR was performed, as described earlier, using the same RNA samples as used for RNAseq. Genes for validation were selected among detected DEGs that were assigned to essential GO and KEGG processes. qPCR was performed with predesigned TaqMan assays (Thermo Fisher Scientific, Waltham, MA, USA) for MT-ND6 (Hs02596879_g1), MT-CO1 (Hs02596864_g1), IDH3G (Hs00188065_m1), MT-ND5 (Hs02596878_g1), DLD (Hs00164401_m1), CBR4 (Hs00379036_m1), SURF1 (Hs00894550_m1), MPC2 (Hs00967250_m1), NDUFA4L2 (Hs00220041_m1), and ETFRF1 (Hs01390827_g1), normalized to the level of RPS15 (Hs01358643_g1) by ΔΔCt, and statistically compared by the unpaired Student’s t-test.

3. Results

3.1. SLC2A8 RNAi in ACH-3P Cells

To target SLC2A8 mRNA for degradation, lentiviral-mediated RNAi was employed in ACH-3P cells, utilizing four distinct SLC2A8 shRNAs and a NTS shRNA control. As evidenced in Figure 1, three out of four of the shRNAs resulted in significant depletion of SLC2A8 mRNA, at both 100 and 500 MOI. The 817 shRNA (Table 1) provided the most consistent RNAi of SLC2A8, and was selected for further analysis at 500 MOI, which is now designated as SLC2A8 RNAi. The glucose uptake by SLC2A8 RNAi was compared to NTS RNAi ACH-3P cells, and the 79% reduction in SLC2A8 mRNA (Figure 1) was associated with an 11% decrease in glucose uptake (Figure 2).

3.2. RNAseq Statistics

RNAseq analysis was conducted on four replicates of ACH-3P mRNA derived from NTS RNAi and SLC2A8 RNA cell lines. High-throughput sequencing on the NovaSeq platform (Illumina) generated 949,834,284 raw paired-end reads. After trimming, 96.4% of reads with good quality were uniquely mapped (Table 3) to the reference human genome, and only 3.6% of reads were mapped to multiple loci. The mean percentage distribution of aligned bases was as follows: 2.01% were derived from intergenic regions, 3.36% from intronic regions, 27.75% from untranslated regions, and 66.68% from coding regions (Figure 3). The sequencing revealed 100,246 transcripts that belong to 31,567 transcribed active regions (TARs).

3.3. SLC2A8 Deficiency Alters Trophectodermal Gene Expression

The most stringent DE statistical method, ballgown, revealed 1525 DE-TARs (|log2FC| > 1 and adjusted p ≤ 0.05). Among DE-TARs, 273 were classified as DEGs localized in a range of protein-coding genes and 147 were signed as DElncRNAs. The rest of the DE-TARs were other types of non-coding RNAs with unknown or uncertain molecular function. The logarithmic values of fold change (log2FC) and statistical significance are visualized on a Volcano plot (Figure 4). Among differentially expressed genes (DEGs) detected between NTS controls and SLC2A8-deficient ACH-3P cells, 148 were upregulated and 125 were downregulated (Figure 5).
Annotations were grouped into 10 biological process (BP), 11 molecular function (MF), and 14 cellular component (CC) categories (Figure 6). Out of the identified DEGs, 327, 326, and 314 coding and non-coding genes were annotated within functional Gene Ontology (GO) database categories BP, CC, and MF, respectively.
The DEGs enriched in GO terms were cross-overlapped (Figure 7). The genes involved in the top five BP classes carried out aerobic electron transport chain (9 DEGs out of 87 genes as a count of term size), aerobic respiration (13/192), respiratory electron transport chain (10/115), mitochondrial electron transport, NADH to ubiquinone (7/51), and oxidative phosphorylation (11/142). The top five terms of MF were oxidoreduction-driven active transmembrane transporter activity (9/71); NAD(P)H dehydrogenase (quinone) activity (7/46); catalytic activity (124/5716); oxidoreductase activity, acting on NAD(P)H, quinone or similar compound as acceptor (7/58); and protein binding (260/14,799). In the CC category, the most abundant significantly enriched terms were cytoplasm (233/12,193), respirasome (11/103), mitochondrial protein-containing complex (17/277), mitochondrial inner membrane (23/500), and inner mitochondrial membrane protein complex (12/154). The KEGG pathway enrichment analysis revealed that DEGs were categorized into 10 pathways, including the oxidative phosphorylation pathway. Annotations and functional assignments of the identified DEGs indicate that SLC2A8 deficiency mostly affects mitochondria functioning.

3.4. Validation

The genes selected for qPCR validation were chosen based on the assessment of the function, expression values, and read distribution within libraries. Four over-expressed genes (MT-ND6, MT-CO1, IDH3G, and MT-ND5) and six under-expressed genes (DLD, CBR4, SURF1, MPC2, NDUFA4L2, and ETFRF1) were tested using qPCR. Laboratory validation confirmed expression differences detected in RNAseq data for MT-ND5, MT-ND6, DLD, CBR4, SURF1, MPC2, NDUFA4L2, and ETFRF1 (Figure 8).

4. Discussion

The main function of the placenta is to provide an appropriate environment for the developing fetus. This function is realized via supplying oxygen and essential nutrients from the maternal circulation, and by extracting carbon dioxide and metabolic waste products from the fetus through the placenta to the maternal circulation [37]. Additionally, the placenta produces hormones and growth factors that are released into the maternal and fetal circulation [38]. Performing these functions is related to a high metabolic rate and oxygen consumption within the placenta. Placental metabolism changes throughout pregnancy and adapts to homoeostatic challenges that come from not only the mother and fetus, but also from the placenta itself. The principal and essential energy substrate for normal placental and fetal metabolism and growth is glucose [39,40]. To meet the high glucose demand of the placenta and developing fetus, its supply is regulated by a compound mechanism that keeps its metabolism relatively constant [1,37]. A part of this mechanism comprises glucose transporters (GLUTs) that mediate facilitated glucose diffusion. GLUTs vary in terms of their substrate specificity, distribution, and regulatory mechanisms [6]. Currently, there are 14 different GLUT proteins that have been characterized in humans, and they are categorized into three main classes based on their sequence homology and functional characteristics. Each GLUT protein has distinct tissue distribution and specific functional characteristics, allowing them to facilitate glucose transport in various physiological contexts. Some GLUTs are constitutively active, while others are insulin-regulated or specific for transporting fructose or urate [8]. Based on current knowledge, the placental glucose transport system is mainly based on SLC2A1 (GLUT1) action. However, it is important to note that other glucose transporters, such as GLUT3 (SLC2A3), GLUT4 (SLC2A4), GLUT8 (SLC2A8), GLUT9 (SLC2A9), GLUT10 (SLC2A10), and GLUT12 (SLC2A12), are also present in the placenta [6,9], which implies they may also be involved in facilitating the uptake of glucose from the maternal bloodstream into placental cells and then supplying the growing fetus. Additionally, such a multitude of different glucose transporters within the placenta may indicate less obvious functions, not necessarily related to direct involvement in glucose uptake. Herein, we report the impact of glucose transporter 8 (SLC2A8) deficiency on the transcriptomic profile of the first-trimester human trophoblast cell line, ACH-3P, and cellular glucose uptake.
Glucose transporter 8 is encoded by the SLC2A8 gene and has been identified in various tissues, including the placenta. Interestingly, SLC2A8 is a dual-specificity glucose and fructose transporter, and its cellular localization and function differs among tissue types [18,41]. In the blastocyst, SLC2A8 participates in insulin-stimulated cell-membrane hexose transport [19], while in other tissues such as the brain, SLC2A8 is involved exclusively in intracellular hexose transport with no evidence of membrane localization [42]. In the ovine placenta, SLC2A8 is expressed within chorionic epithelium and exhibits increased expression over late gestation [23]. These authors [23] conclude that the decreased placental SLC2A8 concentration observed in an ovine model of placental insufficiency and IUGR may contribute to the reduction in placental glucose transport. Our data indicate that lentiviral-mediated RNA interference resulting in a 79% decrease in the SLC2A8 mRNA, resulting in an 11% reduction in glucose uptake, which is consistent with their [23] findings. On the other hand, Janzen et al. [43] reported that human IUGR placenta affected by IUGR showed increased SLC2A8 expression and significant differences in SLC2A8 between basal and chorionic plate regions of the placenta [43]. However, Stanirowski et al. [44] examined placental abundance of SLC2A1, SLC2A3, SLC2A8, and SLC2A12 at term, in pregnancies affected by IUGR, SGA (small for gestational age), or macrosomia, and found reduced density of placental SLC2A8 only in SGA pregnancies. In placentas affected by IUGR, SLC2A1 and SLC2A3 were significantly altered, while SLC2A8 remained similar to that in uncompromised pregnancies. No significant differences in any of the four transporters examined were detected in the placentas of macrosomic fetuses [44]. In order to fully address the discrepancies between these studies, the composition of the research cohorts should be considered, as well as the inclusion and exclusion criteria. However, even with that information, not knowing the relative glucose concentrations and placental transfer rate makes it difficult to draw firm conclusions.
Among glucose transporters, SLC2A8 is a class III transporter that is thought to be primarily involved in intracellular transport, rather than glucose uptake across the plasma membrane [18]. Interestingly, in tumors, SLC2A8 mRNA exists mostly as an untranslated splice variant [45]. It is unclear if this is also the case in normal tissues, or how SLC2A8 mRNA concentration reflects translated SLC2A8. The possibility that SLC2A8 mRNA in trophoblast cells is mostly untranslated could explain the discrepancy between the 79% reduction in SLC2A8 mRNA we obtained and the 11% reduction in glucose uptake. Alternatively, the reduction in glucose uptake may not have directly resulted from SLC2A8-mediated glucose uptake, but indirectly from a reduction in cellular glucose demand for oxidative purposes. The plasticity by which glucose transporters respond to reduced glucose uptake was recently demonstrated by Lynch et al. [14], in which SLC2A3 RNAi resulted in reduced placental glucose uptake in vivo, and a converse increase in SLC2A1 was observed, likely as an attempt to provide adequate glucose to the fetus. The interplay between the various placental glucose transporters, in response to intracellular glucose availability, has not been thoroughly investigated.
Analysis of the SLC2A8 RNAi transcriptome indicated that diminished SLC2A8 mRNA mostly affected processes associated with mitochondria function in terms of cellular respiration, oxidative phosphorylation (OXPHOS), and ATP synthesis. This is consistent with previous reports showing that SLC2A8 mostly functions to facilitate hexose transport through intracellular membranes, such as the mitochondrial membrane, endoplasmic reticular membrane, and lysosomal membrane [8,18]. Moreover, the sperm cells of SLC2A8−/− mice had reduced ATP concentrations and low motility [22], which could result from dysregulated mitochondria function and energy metabolism. Our results may indicate that the reduced glucose uptake due to diminished SLC2A8 expression results from impaired oxidation of glucose for ATP generation.
Among DEGs that enriched biological processes and metabolic functions, 14 genes were overlapping. Within this group, six genes were downregulated (ETFRF1, NDUFA4L2, MPC2, SURF1, CBR4, and DLD) and eight were upregulated (MT-ND3, MT-ND4, MT-ND5, MT-ND6, MT-CO1, MT-ATP6, IDH3G, and FMO5). As mitochondria function in the generation of ATP, they are considered the center of metabolism for nearly all eukaryotic cells. However, mitochondria may also participate in a wide range of essential functions related to cellular metabolism, signaling, and programmed cell death [46]. Furthermore, mitochondria function in modulating calcium signaling, which is a universal secondary messenger [47]. Diminished SLC2A8 expression in trophoblast ACH-3P cells dysregulated MT-ND3, MT-ND4, MT-ND5, and MT-ND6, which are the genes encoding subunits of mitochondrial complex I, the first enzyme of the respiratory chain. Complex I is considered the largest and most complex component of the respiratory chain, but at present, the functions of the individual subunits are largely unclear [48]. The primary functions of complex I are to oxidize NADH, generated through the Krebs cycle, and to reduce ubiquinone to ubiquinol. Additionally, complex I is associated with the regulation of reactive oxygen species (ROS), which are important molecules in various signaling pathways, including apoptosis [49]. Disorders of complex I assembly or function affect mitochondria as a whole and may cause major disruption to energy conversion. Severe impairment of mitochondria function is associated with various metabolic disorders, as well as other diseases such as seizures, ataxia, cortical blindness, dystonia, diabetic mellitus, short stature, cardiomyopathy, sensorineural hearing loss, and kidney failure [50]. Therefore, it may be possible that dysregulation of SLC2A8 expression and glucose uptake leads to disturbed mitochondria function, resulting in pregnancy disorders associated with placental insufficiency and glucose supply below demand.
Among genes that were dysregulated in the currently obtained RNAseq data, several were involved in the formation of cytochrome c oxidase (COX, also known as complex IV), the last enzyme in the respiratory electron transport chain. The COX assembly is a multistep process that involves more than 30 diverse factors, each of which is important for proper COX functioning. SURF1 Cytochrome COxidase Assembly Factor (SURF1) encodes one of the assembly proteins involved in the formation of COX. SURF1 disorders are most common and responsible for severe forms of COX deficiency, such as Leigh syndrome [51,52]. Additionally, dysregulation of the Mitochondrially Encoded Cytochrome COxidase I (MT-CO1), which encodes one of the central subunits of the COX catalytic core, was detected in the current research. Our current results indicate that in the trophoblast ACH-3P cells, the response to reduced SLC2A8 involves downregulation of the NDUFA4 Mitochondrial Complex Associated Like 2 (NDUFA4L2), a subsequent component of mitochondrial respiratory chain complex IV. NDUFA4L2 is suggested to regulate mitochondrial and lysosomal activities [53]. NDUFA4L2 placental expression is upregulated in response to maternal nutrient restriction in sheep [54]. Additionally, in a sheep model of IUGR, impaired NDUFA4L2 expression was associated with reduction of OXPHOS in skeletal muscle, that in turn was identified as a result of prolonged decrease in the tricarboxylic acid cycle (TCA) and electron transport chain activity [55,56].
Our results indicate that SLC2A8 deficiency impacted mitochondria functioning multidimensionally, as not only were major mitochondrial enzymes impaired, but carriers of electrons and substrates were also affected. Electron transfer flavoprotein regulatory factor 1 (ETFRF1) is associated with the OXPHOS complexes, and interacts with and deflavinates the electron transferring flavoprotein that shuttles electrons to coenzyme Q [57]. Research on energy restriction during late gestation and the muscle and blood transcriptome of beef calves revealed that impaired glucose metabolism may affect genes involved in cellular respiration, including ETFRF1 [58]. ETFRF1 was also listed among the genes that were dysregulated in subcutaneous and perirenal adipose tissue of sheep as a resultant of pre- and early postnatal malnutrition [59]. In the current research, ETFRF1 expression was downregulated as a consequence of the diminished SLC2A8 expression. Mitochondrially encoded ATP synthase membrane subunit 6 (MT-ATP6) contributes to proton-transporting ATP synthase activity. Interestingly, MT-ATP6 was listed among mitochondrial and glycolysis-regulatory gene expression profiles that are associated with IUGR [60]. Expression of the MPC2, the subunit of Mitochondrial Pyruvate Carrier (MPC), was also reduced in the current study. MPC2 creates MPC together with MPC1, and abnormal expression of each of these will lead to MPC dysfunction, which in turn will dysregulate the balance of glycolysis and OXPHOS [61,62].
It should be mentioned that due to the identified dysregulation of genes such as Carbonyl Reductase 4 (CBR4), dihydrolipoamide dehydrogenase (DLD), Flavin Containing Dimethylaniline Monooxygenase 5 (FMO5), and Isocitrate Dehydrogenase (NAD(+)) 3 Non-Catalytic Subunit Gamma (IDH3G), other mitochondrial aspects can also be affected by SLC2A8 deficiency. Among these, mitochondrial fatty acid synthesis [63,64], as well as disruption of multiple enzyme complexes [65], lipid homeostasis, the uptake and metabolism of glucose, the generation of cytosolic NADPH, the one-carbon metabolism [66], and the TCA cycle, [67], should be listed. An overview of the genes dysregulated due to diminished SLC2A8 expression supports the conclusion that SLC2A8 is important for proper mitochondria functioning. Impaired expression of the indicated genes, regardless of the direction of change, may cause structural as well as functional mitochondria disruption, manifested by impaired nutrient oxidation and therefore impaired ATP synthesis.
Further functional studies are needed to fully assess SLC2A8 function in the placenta; however, our results regarding reduced glucose uptake and the impact on the transcriptome resulting from SLC2A8 RNAi, indicate it is reasonable to speculate that SLC2A8 deficiency could significantly impair placental function. Placental glucose transfer to the fetus depends on the maternal concentration of glucose, the uptake of glucose by the uteroplacental unit, and the maternal/fetal glucose concentration gradient [37,68]. The maternal/fetal concentration gradient, while dependent on the maternal [69] and fetal [37] glucose concentrations, in many ways is also determined by the rate of placental glucose oxidation [16,17]. Collectively, our RNAseq results could infer that a deficiency in placental SLC2A8 would likely impair ATP generation, general metabolic processes, and the redox balance, likely resulting in oxidative stress within the placenta. Placental oxidative stress would have detrimental effects on placental function and fetal development in general, and is considered a factor regulating gene expression and downstream activities such as trophoblast proliferation, invasion, and angiogenesis [70]. Furthermore, the impact of the oxidative stress on placental function is dependent on when during gestation it occurs; however, oxidative stress has been associated with preeclampsia, IUGR, and even pregnancy loss [71,72]. While many factors can influence placental function and placental glucose utilization, our results suggest that SLC2A8, as an intracellular glucose transporter, may play a key role in regulating placental metabolism.

5. Conclusions

Using lentiviral-mediated RNA interference in human trophoblast cells, we investigated the impact of specifically reducing the availability of SLC2A8. While SLC2A8 RNAi diminished glucose uptake by these cells, the response was not dramatic and may have been an indirect response. By contrast, there was a major impact on the trophoblast transcriptome, revealing potential impairment in mitochondrial function and oxidative processes. These results align with previous reports in the testes of Slc2A8−/− mice, resulting in reduced ATP production. As the mammalian placenta utilizes the majority of oxygen and glucose taken up from the maternal circulation, for oxidative processes, our results suggest that SLC2A8 deficiency would likely impair intracellular glucose transport and oxidation, resulting in functional placental insufficiency. Since functional placental insufficiency is a major cause of IUGR, a more thorough understanding of the regulation of placental metabolism is needed, and is highlighted by our results. It should also be kept in mind that placental insufficiency can have lasting effects, impacting the postnatal period and adulthood [73].

Author Contributions

Conceptualization, A.L. and R.V.A.; methodology, R.V.A.; software, Ł.P.; validation, A.L. and V.C.K.; data analysis, A.L., Ł.P., M.M. and R.V.A.; investigation, A.L., V.C.K. and A.R.T.; resources, R.V.A.; writing—original draft preparation, A.L. and R.V.A.; writing—review and editing, A.L., Ł.P., V.C.K., A.R.T., M.M. and R.V.A.; visualization, A.L. and Ł.P.; supervision, R.V.A.; funding acquisition, R.V.A. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Institutes of Health grants HD094952 and HD093701. A.L. was supported with the Fulbright Senior Award (PL/2020/43/SR) funded by the Polish-U.S. Fulbright Commission and the Bekker Programme funded by the Polish National Agency For Academic Exchange (PPN/BEK/2019/1/00248/U/00001).

Institutional Review Board Statement

All procedures were approved by the Colorado State University Institutional Biosafe-ty Committee (17-039B).

Informed Consent Statement

Not applicable.

Data Availability Statement

The sequencing data from this study have been submitted to the European Nucleotide Archive under accession no. PRJEB71894 (https://www.ebi.ac.uk/ena, 16 January 2024).

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Hay, W.W. Placental-Fetal Glucose Exchange and Fetal Glucose Metabolism. Trans. Am. Clin. Climatol. Assoc. 2006, 117, 321–339, discussion 339–340. [Google Scholar]
  2. Hauguel, S.; Desmaizieres, V.; Challier, J.C. Glucose Uptake, Utilization, and Transfer by the Human Placenta as Functions of Maternal Glucose Concentration. Pediatr. Res. 1986, 20, 269–273. [Google Scholar] [CrossRef]
  3. Zeng, Z.; Liu, F.; Li, S. Metabolic Adaptations in Pregnancy: A Review. Ann. Nutr. Metab. 2017, 70, 59–65. [Google Scholar] [CrossRef]
  4. Anand, R.S.; Ganguli, S.; Sperling, M.A. Effect of Insulin-Induced Maternal Hypoglycemia on Glucose Turnover in Maternal and Fetal Sheep. Am. J. Physiol.-Endocrinol. Metab. 1980, 238, E524–E532. [Google Scholar] [CrossRef]
  5. Marconi, A.M.; Cetin, I.; Davoli, E.; Baggiani, A.M.; Fanelli, R.; Fennessey, P.V.; Battaglia, F.C.; Pardi, G. An Evaluation of Fetal Glucogenesis in Intrauterine Growth-Retarded Pregnancies. Metabolism 1993, 42, 860–864. [Google Scholar] [CrossRef]
  6. Illsley, N.P. Glucose Transporters in the Human Placenta. Placenta 2000, 21, 14–22. [Google Scholar] [CrossRef]
  7. Stanirowski, P.J.; Szukiewicz, D.; Pazura-Turowska, M.; Sawicki, W.; Cendrowski, K. Placental Expression of Glucose Transporter Proteins in Pregnancies Complicated by Gestational and Pregestational Diabetes Mellitus. Can. J. Diabetes 2018, 42, 209–217. [Google Scholar] [CrossRef]
  8. Navale, A.M.; Paranjape, A.N. Glucose Transporters: Physiological and Pathological Roles. Biophys. Rev. 2016, 8, 5–9. [Google Scholar] [CrossRef]
  9. Illsley, N.P.; Baumann, M.U. Human Placental Glucose Transport in Fetoplacental Growth and Metabolism. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165359. [Google Scholar] [CrossRef]
  10. Jansson, T.; Ylvén, K.; Wennergren, M.; Powell, T.L. Glucose Transport and System A Activity in Syncytiotrophoblast Microvillous and Basal Plasma Membranes in Intrauterine Growth Restriction. Placenta 2002, 23, 392–399. [Google Scholar] [CrossRef]
  11. Jansson, T.; Wennergren, M.; Illsley, N.P. Glucose Transporter Protein Expression in Human Placenta throughout Gestation and in Intrauterine Growth Retardation. J. Clin. Endocrinol. Metab. 1993, 77, 1554–1562. [Google Scholar] [CrossRef]
  12. Simpson, I.A.; Dwyer, D.; Malide, D.; Moley, K.H.; Travis, A.; Vannucci, S.J. The Facilitative Glucose Transporter GLUT3: 20 Years of Distinction. Am. J. Physiol. Endocrinol. Metab. 2008, 295, 242–253. [Google Scholar] [CrossRef] [PubMed]
  13. Brown, K.; Heller, D.S.; Zamudio, S.; Illsley, N.P. Glucose Transporter 3 (GLUT3) Protein Expression in Human Placenta across Gestation. Placenta 2011, 32, 1041–1049. [Google Scholar] [CrossRef] [PubMed]
  14. Lynch, C.S.; Kennedy, V.C.; Tanner, A.R.; Ali, A.; Winger, Q.A.; Rozance, P.J.; Anthony, R.V. Impact of Placental SLC2A3 Deficiency during the First-Half of Gestation. Int. J. Mol. Sci. 2022, 23, 12530. [Google Scholar] [CrossRef]
  15. Janzen, C.; Lei, M.Y.Y.; Cho, J.; Sullivan, P.; Shin, B.C.; Devaskar, S.U. Placental Glucose Transporter 3 (GLUT3) Is up-Regulated in Human Pregnancies Complicated by Late-Onset Intrauterine Growth Restriction. Placenta 2013, 34, 1072–1078. [Google Scholar] [CrossRef]
  16. Meschia, G.; Battaglia, F.C.; Hay, W.W.; Sparks, J.W. Utilization of Substrates by the Ovine Placenta in Vivo. Fed. Proc. 1980, 39, 245–249. [Google Scholar]
  17. Bell, A.W.; Kennaugh, J.M.; Battaglia, F.C.; Makowski, E.L.; Meschia, G. Metabolic and Circulatory Studies of Fetal Lamb at Midgestation. Am. J. Physiol.-Endocrinol. Metab. 1986, 250, E538–E544. [Google Scholar] [CrossRef]
  18. Schmidt, S.; Joost, H.G.; Schürmann, A. GLUT8, the Enigmatic Intracellular Hexose Transporter. Am. J. Physiol. Endocrinol. Metab. 2009, 296, 614–618. [Google Scholar] [CrossRef]
  19. Carayannopoulos, M.O.; Chi, M.M.Y.; Cui, Y.; Pingsterhaus, J.M.; McKnight, R.A.; Mueckler, M.; Devaskar, S.U.; Moley, K.H. GLUT8 Is a Glucose Transporter Responsible for Insulin-Stimulated Glucose Uptake in the Blastocyst. Proc. Natl. Acad. Sci. USA 2000, 97, 7313–7318. [Google Scholar] [CrossRef]
  20. Pinto, A.B.; Carayannopoulos, M.O.; Hoehn, A.; Dowd, L.; Moley, K.H. Glucose Transporter 8 Expression and Translocation Are Critical for Murine Blastocyst Survival. Biol. Reprod. 2002, 66, 1729–1733. [Google Scholar] [CrossRef]
  21. Membrez, M.; Hummler, E.; Beermann, F.; Haefliger, J.-A.; Savioz, R.; Pedrazzini, T.; Thorens, B. GLUT8 Is Dispensable for Embryonic Development but Influences Hippocampal Neurogenesis and Heart Function. Mol. Cell. Biol. 2006, 26, 4268–4276. [Google Scholar] [CrossRef]
  22. Gawlik, V.; Schmidt, S.; Scheepers, A.; Wennemuth, G.; Augustin, R.; Aumüller, G.; Moser, M.; Al-Hasani, H.; Kluge, R.; Joost, H.G.; et al. Targeted Disruption of Slc2a8 (GLUT8) Reduces Motility and Mitochondrial Potential of Spermatozoa. Mol. Membr. Biol. 2008, 25, 224–235. [Google Scholar] [CrossRef]
  23. Limesand, S.W.; Regnault, T.R.H.; Hay, W.W. Characterization of Glucose Transporter 8 (GLUT8) in the Ovine Placenta of Normal and Growth Restricted Fetuses. Placenta 2004, 25, 70–77. [Google Scholar] [CrossRef]
  24. Baker, C.M.; Goetzmann, L.N.; Cantlon, J.D.; Jeckel, K.M.; Winger, Q.A.; Anthony, R.V. Development of Ovine Chorionic Somatomammotropin Hormone-Deficient Pregnancies. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2016, 310, R837–R846. [Google Scholar] [CrossRef]
  25. Jeckel, K.M.; Boyarko, A.C.; Bouma, G.J.; Winger, Q.A.; Anthony, R.V. Chorionic Somatomammotropin Impacts Early Fetal Growth and Placental Gene Expression. J. Endocrinol. 2018, 237, 301–310. [Google Scholar] [CrossRef]
  26. Ali, A.; Anthony, R.V.; Bouma, G.J.; Winger, Q.A. LIN28-Let-7 Axis Regulates Genes in Immortalized Human Trophoblast Cells by Targeting the ARID3B-Complex. FASEB J. 2019, 33, 12348–12363. [Google Scholar] [CrossRef]
  27. Gates, K.C.; Goetzmann, L.N.; Cantlon, J.D.; Jeckel, K.M.; Anthony, R.V. Effect of Proline Rich 15-Deficiency on Trophoblast Viability and Survival. PLoS ONE 2017, 12, e0174976. [Google Scholar] [CrossRef] [PubMed]
  28. Hiden, U.; Wadsack, C.; Prutsch, N.; Gauster, M.; Weiss, U.; Frank, H.-G.; Schmitz, U.; Fast-Hirsch, C.; Hengstschläger, M.; Pötgens, A.; et al. The First Trimester Human Trophoblast Cell Line ACH-3P: A Novel Tool to Study Autocrine/Paracrine Regulatory Loops of Human Trophoblast Subpopulations–TNF-Alpha Stimulates MMP15 Expression. BMC Dev. Biol. 2007, 7, 137. [Google Scholar] [CrossRef] [PubMed]
  29. Bolger, A.M.; Lohse, M.; Usadel, B. Trimmomatic: A Flexible Trimmer for Illumina Sequence Data. Bioinformatics 2014, 30, 2114–2120. [Google Scholar] [CrossRef] [PubMed]
  30. Li, H.; Handsaker, B.; Wysoker, A.; Fennell, T.; Ruan, J.; Homer, N.; Marth, G.; Abecasis, G.; Durbin, R. The Sequence Alignment/Map Format and SAMtools. Bioinformatics 2009, 25, 2078–2079. [Google Scholar] [CrossRef] [PubMed]
  31. Pertea, M.; Pertea, G.M.; Antonescu, C.M.; Chang, T.-C.; Mendell, J.T.; Salzberg, S.L. StringTie Enables Improved Reconstruction of a Transcriptome from RNA-Seq Reads. Nat. Biotechnol. 2015, 33, 290–295. [Google Scholar] [CrossRef]
  32. Frazee, A.C.; Pertea, G.; Jaffe, A.E.; Langmead, B.; Salzberg, S.L.; Leek, J.T. Ballgown Bridges the Gap between Transcriptome Assembly and Expression Analysis. Nat. Biotechnol. 2015, 33, 243–246. [Google Scholar] [CrossRef]
  33. Lipka, A.; Jastrzebski, J.P.; Paukszto, L.; Makowczenko, K.G.; Lopienska-Biernat, E.; Gowkielewicz, M.; Lepiarczyk, E.; Wiszpolska, M.; Majewski, M.K.; Majewska, M. Sex-Biased LncRNA Signature in Fetal Growth Restriction (FGR). Cells 2021, 10, 921. [Google Scholar] [CrossRef]
  34. Majewska, M.; Lipka, A.; Paukszto, L.; Jastrzebski, J.P.; Myszczynski, K.; Gowkielewicz, M.; Jozwik, M.; Majewski, M.K. Transcriptome Profile of the Human Placenta. Funct. Integr. Genom. 2017, 17, 551–563. [Google Scholar] [CrossRef]
  35. Majewska, M.; Lipka, A.; Paukszto, L.; Jastrzebski, J.P.; Szeszko, K.; Gowkielewicz, M.; Lepiarczyk, E.; Jozwik, M.; Majewski, M.K. Placenta Transcriptome Profiling in Intrauterine Growth Restriction (IUGR). Int. J. Mol. Sci. 2019, 20, 1510. [Google Scholar] [CrossRef]
  36. Reimand, J.; Arak, T.; Adler, P.; Kolberg, L.; Reisberg, S.; Peterson, H.; Vilo, J. G:Profiler—A Web Server for Functional Interpretation of Gene Lists. Nucleic Acids Res. 2016, 44, W83–W89. [Google Scholar] [CrossRef] [PubMed]
  37. Hay, W.W.; Molina, R.A.; DiGiacomo, J.E.; Meschia, G. Model of Placental Glucose Consumption and Glucose Transfer. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 1990, 258, R569–R577. [Google Scholar] [CrossRef] [PubMed]
  38. Burton, G.J.; Fowden, A.L. Review: The Placenta and Developmental Programming: Balancing Fetal Nutrient Demands with Maternal Resource Allocation. Placenta 2012, 33, S23–S27. [Google Scholar] [CrossRef] [PubMed]
  39. Hay, W.W. Energy and Substrate Requirements of the Placenta and Fetus. Proc. Nutr. Soc. 1991, 50, 321–336. [Google Scholar] [CrossRef]
  40. Vaughan, O.R.; Sferruzzi-Perri, A.N.; Coan, P.M.; Fowden, A.L. Environmental Regulation of Placental Phenotype: Implications for Fetal Growth. Reprod. Fertil. Dev. 2012, 24, 80–96. [Google Scholar] [CrossRef] [PubMed]
  41. Doege, H.; Schürmann, A.; Bahrenberg, G.; Brauers, A.; Joost, H.G. GLUT8, a Novel Member of the Sugar Transport Facilitator Family with Glucose Transport Activity. J. Biol. Chem. 2000, 275, 16275–16280. [Google Scholar] [CrossRef]
  42. Sankar, R.; Thamotharan, S.; Shin, D.; Moley, K.H.; Devaskar, S.U. Insulin-Responsive Glucose Transporters-GLUT8 and GLUT4 Are Expressed in the Developing Mammalian Brain. Mol. Brain Res. 2002, 107, 157–165. [Google Scholar] [CrossRef]
  43. Janzen, C.; Lei, M.Y.Y.; Jeong, I.S.D.; Ganguly, A.; Sullivan, P.; Paharkova, V.; Capodanno, G.; Nakamura, H.; Perry, A.; Shin, B.-C.; et al. Humanin (HN) and Glucose Transporter 8 (GLUT8) in Pregnancies Complicated by Intrauterine Growth Restriction. PLoS ONE 2018, 13, e0193583. [Google Scholar] [CrossRef]
  44. Stanirowski, P.J.; Lipa, M.; Bomba-Opoń, D.; Wielgoś, M. Expression of Placental Glucose Transporter Proteins in Pregnancies Complicated by Fetal Growth Disorders. In Advances in Protein Chemistry and Structural Biology; Elsevier: Amsterdam, The Netherlands, 2021; Volume 123, pp. 95–131. ISBN 9780128220870. [Google Scholar]
  45. Alexander, C.M.; Martin, J.A.; Oxman, E.; Kasza, I.; Senn, K.A.; Dvinge, H. Alternative Splicing and Cleavage of GLUT8. Mol. Cell Biol. 2020, 41, e00480-20. [Google Scholar] [CrossRef]
  46. Pagliarini, D.J.; Rutter, J. Hallmarks of a New Era in Mitochondrial Biochemistry. Genes. Dev. 2013, 27, 2615–2627. [Google Scholar] [CrossRef]
  47. Modesti, L.; Danese, A.; Angela Maria Vitto, V.; Ramaccini, D.; Aguiari, G.; Gafà, R.; Lanza, G.; Giorgi, C.; Pinton, P. Mitochondrial Ca2+ Signaling in Health, Disease and Therapy. Cells 2021, 10, 1317. [Google Scholar] [CrossRef]
  48. Saneto, R.P. Mitochondrial Diseases: Expanding the Diagnosis in the Era of Genetic Testing. J. Transl. Genet. Genom. 2020, 4, 384–428. [Google Scholar] [CrossRef]
  49. Henriques, B.J.; Katrine Jentoft Olsen, R.; Gomes, C.M.; Bross, P. Electron Transfer Flavoprotein and Its Role in Mitochondrial Energy Metabolism in Health and Disease. Gene 2021, 776, 145407. [Google Scholar] [CrossRef] [PubMed]
  50. Mallik, B.; Frank, C.A. Roles for Mitochondrial Complex I Subunits in Regulating Synaptic Transmission and Growth. Front. Neurosci. 2022, 16, 846425. [Google Scholar] [CrossRef] [PubMed]
  51. Kovářová, N.; Čížková Vrbacká, A.; Pecina, P.; Stránecký, V.; Pronicka, E.; Kmoch, S.; Houštěk, J. Adaptation of Respiratory Chain Biogenesis to Cytochrome c Oxidase Deficiency Caused by SURF1 Gene Mutations. Biochim. Biophys. Acta Mol. Basis Dis. 2012, 1822, 1114–1124. [Google Scholar] [CrossRef] [PubMed]
  52. Brischigliaro, M.; Zeviani, M. Cytochrome c Oxidase Deficiency. Biochim. Biophys. Acta Bioenerg. 2021, 1862, 148335. [Google Scholar] [CrossRef] [PubMed]
  53. Kubala, J.M.; Laursen, K.B.; Schreiner, R.; Williams, R.M.; van der Mijn, J.C.; Crowley, M.J.; Mongan, N.P.; Nanus, D.M.; Heller, D.A.; Gudas, L.J. NDUFA4L2 Reduces Mitochondrial Respiration Resulting in Defective Lysosomal Trafficking in Clear Cell Renal Cell Carcinoma. Cancer Biol. Ther. 2023, 24, 2170669. [Google Scholar] [CrossRef] [PubMed]
  54. Steinhauser, C.B.; Lambo, C.A.; Askelson, K.; Burns, G.W.; Behura, S.K.; Spencer, T.E.; Bazer, F.W.; Satterfield, M.C. Placental Transcriptome Adaptations to Maternal Nutrient Restriction in Sheep. Int. J. Mol. Sci. 2021, 22, 7654. [Google Scholar] [CrossRef] [PubMed]
  55. Pendleton, A.L.; Antolic, A.T.; Kelly, A.C.; Davis, M.A.; Camacho, L.E.; Doubleday, K.; Anderson, M.J.; Langlais, P.R.; Lynch, R.M.; Limesand, S.W. Lower Oxygen Consumption and Complex I Activity in Mitochondria Isolated from Skeletal Muscle of Fetal Sheep with Intrauterine Growth Restriction. Am. J. Physiol. Endocrinol. Metab. 2020, 319, E67–E80. [Google Scholar] [CrossRef] [PubMed]
  56. Pendleton, A.L.; Wesolowski, S.R.; Regnault, T.R.H.; Lynch, R.M.; Limesand, S.W. Dimming the Powerhouse: Mitochondrial Dysfunction in the Liver and Skeletal Muscle of Intrauterine Growth Restricted Fetuses. Front. Endocrinol. 2021, 12, 612888. [Google Scholar] [CrossRef] [PubMed]
  57. Floyd, B.J.; Wilkerson, E.M.; Veling, M.T.; Minogue, C.E.; Xia, C.; Beebe, E.T.; Wrobel, R.L.; Cho, H.; Kremer, L.S.; Alston, C.L.; et al. Mitochondrial Protein Interaction Mapping Identifies New Regulators of Respiratory Chain Function of the Project and Its Design HHS Public Access. Mol. Cell 2016, 63, 621–632. [Google Scholar] [CrossRef] [PubMed]
  58. Sanglard, L.P.; Nascimento, M.; Moriel, P.; Sommer, J.; Ashwell, M.; Poore, M.H.; Duarte, M.D.S.; Serão, N.V.L. Impact of Energy Restriction during Late Gestation on the Muscle and Blood Transcriptome of Beef Calves after Preconditioning. BMC Genom. 2018, 19, 702. [Google Scholar] [CrossRef]
  59. Ahmad, S.; Drag, M.H.; Salleh, S.M.; Cai, Z.; Nielsen, M.O. Transcriptomics Analysis of Differentially Expressed Genes in Subcutaneous and Perirenal Adipose Tissue of Sheep as Affected by Their Pre- and Early Postnatal Malnutrition Histories. BMC Genom. 2021, 22, 338. [Google Scholar] [CrossRef]
  60. Jones, R.; Peña, J.; Mystal, E.; Marsit, C.; Lee, M.J.; Stone, J.; Lambertini, L. Mitochondrial and Glycolysis-Regulatory Gene Expression Profiles Are Associated with Intrauterine Growth Restriction. J. Matern.-Fetal Neonatal Med. 2020, 33, 1336–1345. [Google Scholar] [CrossRef]
  61. Bender, T.; Martinou, J.C. The Mitochondrial Pyruvate Carrier in Health and Disease: To Carry or Not to Carry? Biochim. Biophys. Acta Mol. Cell Res. 2016, 1863, 2436–2442. [Google Scholar] [CrossRef] [PubMed]
  62. Feng, J.; Ma, Y.; Chen, Z.; Hu, J.; Yang, Q.; Ding, G. Mitochondrial Pyruvate Carrier 2 Mediates Mitochondrial Dysfunction and Apoptosis in High Glucose-Treated Podocytes. Life Sci. 2019, 237, 116941. [Google Scholar] [CrossRef]
  63. Venkatesan, R.; Sah-Teli, S.K.; Awoniyi, L.O.; Jiang, G.; Prus, P.; Kastaniotis, A.J.; Hiltunen, J.K.; Wierenga, R.K.; Chen, Z. Insights into Mitochondrial Fatty Acid Synthesis from the Structure of Heterotetrameric 3-Ketoacyl-ACP Reductase/3R-Hydroxyacyl-CoA Dehydrogenase. Nat. Commun. 2014, 5, 4805. [Google Scholar] [CrossRef]
  64. Hiltunen, J.K.; Autio, K.J.; Schonauer, M.S.; Kursu, V.A.S.; Dieckmann, C.L.; Kastaniotis, A.J. Mitochondrial Fatty Acid Synthesis and Respiration. Biochim. Biophys. Acta Bioenerg. 2010, 1797, 1195–1202. [Google Scholar] [CrossRef]
  65. Blackburn, P.; Gass, J.; Pinto e Vairo, F.; Farnham, K.; Atwal, H.; Macklin, S.; Klee, E.; Atwal, P. Maple Syrup Urine Disease: Mechanisms and Management. Appl. Clin. Genet. 2017, 10, 57–66. [Google Scholar] [CrossRef]
  66. Phillips, I.R.; Veeravalli, S.; Khadayate, S.; Shephard, E.A. Metabolomic and Transcriptomic Analyses of Fmo5-/- Mice Reveal Roles for Flavin-Containing Monooxygenase 5 (FMO5) in NRF2-Mediated Oxidative Stress Response, Unfolded Protein Response, Lipid Homeostasis, and Carbohydrate and One-Carbon Metabolism. PLoS ONE 2023, 18, e0286692. [Google Scholar] [CrossRef] [PubMed]
  67. Al-Khallaf, H. Isocitrate Dehydrogenases in Physiology and Cancer: Biochemical and Molecular Insight. Cell Biosci. 2017, 7, 37. [Google Scholar] [CrossRef] [PubMed]
  68. Hay, W.W., Jr.; Meznarich, H.K. Effect of Maternal Glucose Concentration on Uteroplacental Glucose Consumption and Transfer in Pregnant Sheep. Proc. Soc. Exp. Biol. Med. 1989, 190, 63–69. [Google Scholar] [CrossRef] [PubMed]
  69. Simmons, M.A.; Battaglia, F.C.; Meschia, G. Placental Transfer of Glucose. J. Dev. Physiol. 1979, 1, 227–243. [Google Scholar] [PubMed]
  70. Wu, F.; Tian, F.-J.; Lin, Y. Oxidative Stress in Placenta: Health and Diseases. Biomed. Res. Int. 2015, 2015, 293271. [Google Scholar] [CrossRef] [PubMed]
  71. Ball, E.; Bulmer, J.N.; Ayis, S.; Lyall, F.; Robson, S.C. Late Sporadic Miscarriage Is Associated with Abnormalities in Spiral Artery Transformation and Trophoblast Invasion. J. Pathol. 2006, 208, 535–542. [Google Scholar] [CrossRef] [PubMed]
  72. Kadyrov, M.; Kingdom, J.C.P.; Huppertz, B. Divergent Trophoblast Invasion and Apoptosis in Placental Bed Spiral Arteries from Pregnancies Complicated by Maternal Anemia and Early-Onset Preeclampsia/Intrauterine Growth Restriction. Am. J. Obstet. Gynecol. 2006, 194, 557–563. [Google Scholar] [CrossRef] [PubMed]
  73. Cleal, J.K.; Poore, K.R.; Lewis, R.M. The Placental Exposome, Placental Epigenetic Adaptations and Lifelong Cardio-Metabolic Health. Mol. Aspects Med. 2022, 87, 101095. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effect of lentivirus-mediated RNA interference on SLC2A8 mRNA level in ACH-3P cells. Concentration of SLC2A8 mRNA was measured by qPCR. Data are shown as mean values ± SEM. * p ≤ 0.05 when SLC2A8-deficient cell lines are compared with controls (NTS RNAi).
Figure 1. Effect of lentivirus-mediated RNA interference on SLC2A8 mRNA level in ACH-3P cells. Concentration of SLC2A8 mRNA was measured by qPCR. Data are shown as mean values ± SEM. * p ≤ 0.05 when SLC2A8-deficient cell lines are compared with controls (NTS RNAi).
Cells 13 00391 g001
Figure 2. Effect of lentivirus-mediated SLC2A8 RNAi on glucose uptake in ACH-3P cells. Data are shown as mean values ± SEM. * p ≤ 0.05 when SLC2A8-deficient cell lines are compared with controls (NTS RNAi).
Figure 2. Effect of lentivirus-mediated SLC2A8 RNAi on glucose uptake in ACH-3P cells. Data are shown as mean values ± SEM. * p ≤ 0.05 when SLC2A8-deficient cell lines are compared with controls (NTS RNAi).
Cells 13 00391 g002
Figure 3. Expression profile overview. (A) Graphical representation of the first (PC1) and second (PC2) principal components affecting the sample expression pattern of control (NTS RNAi) and SLC2A8-deficient ACH-3P libraries (SLC2A8 RNAi). (B) distribution of reads mapped to the human genome within intergenic, intronic, UTR, and coding regions.
Figure 3. Expression profile overview. (A) Graphical representation of the first (PC1) and second (PC2) principal components affecting the sample expression pattern of control (NTS RNAi) and SLC2A8-deficient ACH-3P libraries (SLC2A8 RNAi). (B) distribution of reads mapped to the human genome within intergenic, intronic, UTR, and coding regions.
Cells 13 00391 g003
Figure 4. Volcano plot depicts log2FC plotted against log-normalized p-values of lncRNA (A) and protein-coding genes (B) in the deficiency of SLC2A8 expression. The dotted horizontal line indicates a negative logarithmic adjusted p ≤ 0.05 cut-off. Dotted vertical lines indicate cut-off values of logFC. (A) Orange dots represent upregulated differentially expressed lncRNA (DElncRNA); purple dots refer to downregulated DElncRNA. (B) Red dots illustrate upregulated differentially expressed genes (DEGs); green dots represent downregulated DEGs. Black dots are not significant transcripts, according to the ballgown method (A,B).
Figure 4. Volcano plot depicts log2FC plotted against log-normalized p-values of lncRNA (A) and protein-coding genes (B) in the deficiency of SLC2A8 expression. The dotted horizontal line indicates a negative logarithmic adjusted p ≤ 0.05 cut-off. Dotted vertical lines indicate cut-off values of logFC. (A) Orange dots represent upregulated differentially expressed lncRNA (DElncRNA); purple dots refer to downregulated DElncRNA. (B) Red dots illustrate upregulated differentially expressed genes (DEGs); green dots represent downregulated DEGs. Black dots are not significant transcripts, according to the ballgown method (A,B).
Cells 13 00391 g004
Figure 5. Circular heatmap visualization of differentially expressed genes (DEGs) and differentially expressed lncRNA (DElncRNA) resulting from the deficiency in SLC2A8 in ACH-3P cells. The eight tracks visualize the normalized (Z-score; red–green scale) expression profiles for DEGs and DElncRNA in each of NTS RNAi and SLC2A8 RNAi samples. The most inner track shows the correlation links between the co-expressed DEGs and DElncRNA, whereas blue links depict positive and yellow negative (<−0.9) Euclidean correlation > 0.9.
Figure 5. Circular heatmap visualization of differentially expressed genes (DEGs) and differentially expressed lncRNA (DElncRNA) resulting from the deficiency in SLC2A8 in ACH-3P cells. The eight tracks visualize the normalized (Z-score; red–green scale) expression profiles for DEGs and DElncRNA in each of NTS RNAi and SLC2A8 RNAi samples. The most inner track shows the correlation links between the co-expressed DEGs and DElncRNA, whereas blue links depict positive and yellow negative (<−0.9) Euclidean correlation > 0.9.
Cells 13 00391 g005
Figure 6. Enrichment ontology visualization. (A) Gene Ontology (GO) bubble chart of the assigned ontology terms (biological process—BP, cellular components—CC, and metabolic function—MF). Circle size is proportional to the logarithmic scale of adjusted p-value in enrichment GO analysis. Z-score is calculated from the number of up- and downregulated genes enriched in each GO term. (B) Circos visualization of selected GO processes related to mitochondria function. Red dots illustrate upregulated differentially expressed genes (DEGs); blue dots represent downregulated DEGs.
Figure 6. Enrichment ontology visualization. (A) Gene Ontology (GO) bubble chart of the assigned ontology terms (biological process—BP, cellular components—CC, and metabolic function—MF). Circle size is proportional to the logarithmic scale of adjusted p-value in enrichment GO analysis. Z-score is calculated from the number of up- and downregulated genes enriched in each GO term. (B) Circos visualization of selected GO processes related to mitochondria function. Red dots illustrate upregulated differentially expressed genes (DEGs); blue dots represent downregulated DEGs.
Cells 13 00391 g006
Figure 7. Circos plot represents four significantly enriched Gene Ontology (GO) terms associated with detected differentially expressed genes (DEGs). Gene symbols with logarithmic values (blue-red scale) of fold change (logFC) are located on the left side of the circos. Four color links merge genes with GO annotations.
Figure 7. Circos plot represents four significantly enriched Gene Ontology (GO) terms associated with detected differentially expressed genes (DEGs). Gene symbols with logarithmic values (blue-red scale) of fold change (logFC) are located on the left side of the circos. Four color links merge genes with GO annotations.
Cells 13 00391 g007
Figure 8. The mRNA expression of selected genes obtained using real-time PCR. The expression values were normalized to RPS15 housekeeping gene expression. * p ≤ 0.05, ** p ≤ 0.01 when SLC2A8-deficient samples are compared with controls (NTS RNAi). Abbreviations: MT-ND6 (Mitochondrially Encoded NADH:Ubiquinone Oxidoreductase Core Subunit 6); MT-CO1 (Mitochondrially Encoded Cytochrome C Oxidase I); IDH3G (Isocitrate Dehydrogenase (NAD(+)) 3 Non-Catalytic Subunit Gamma); MT-ND5 (Mitochondrially Encoded NADH:Ubiquinone Oxidoreductase Core Subunit 5); DLD (Dihydrolipoamide Dehydrogenase); CBR4 (Carbonyl Reductase 4); SURF1 (SURF1 Cytochrome C Oxidase Assembly Factor); MPC2 (Mitochondrial Pyruvate Carrier 2); NDUFA4L2 (NDUFA4 Mitochondrial Complex Associated Like 2); ETFRF1 (Electron Transfer Flavoprotein Regulatory Factor 1).
Figure 8. The mRNA expression of selected genes obtained using real-time PCR. The expression values were normalized to RPS15 housekeeping gene expression. * p ≤ 0.05, ** p ≤ 0.01 when SLC2A8-deficient samples are compared with controls (NTS RNAi). Abbreviations: MT-ND6 (Mitochondrially Encoded NADH:Ubiquinone Oxidoreductase Core Subunit 6); MT-CO1 (Mitochondrially Encoded Cytochrome C Oxidase I); IDH3G (Isocitrate Dehydrogenase (NAD(+)) 3 Non-Catalytic Subunit Gamma); MT-ND5 (Mitochondrially Encoded NADH:Ubiquinone Oxidoreductase Core Subunit 5); DLD (Dihydrolipoamide Dehydrogenase); CBR4 (Carbonyl Reductase 4); SURF1 (SURF1 Cytochrome C Oxidase Assembly Factor); MPC2 (Mitochondrial Pyruvate Carrier 2); NDUFA4L2 (NDUFA4 Mitochondrial Complex Associated Like 2); ETFRF1 (Electron Transfer Flavoprotein Regulatory Factor 1).
Cells 13 00391 g008
Table 1. SLC2A8-targeting shRNA sequences.
Table 1. SLC2A8-targeting shRNA sequences.
GeneAccession NumberOligo NameOligonucleotide Sequence (5′ → 3′)
SLC2A8Y17801.1650 FCCGGGCTTCTCATGTGCTTCATGCCTTCAAGAGAGGCATGAAGCACATGAGAAGCTTTTTG
650 RAATTCAAAAAGCTTCTCATGTGCTTCATGCCTCTCTTGAAGGCATGAAGCACATGAGAAGC
817 FCCGGGCATCTACAAGCCCTTCATCATTCAAGAGATGATGAAGGGCTTGTAGATGCTTTTTG
817 RAATTCAAAAAGCATCTACAAGCCCTTCATCATCTCTTGAATGATGAAGGGCTTGTAGATGC
1452 FCCGGGGAAAGACTCTGGAACAAATCTTCAAGAGAGATTTGTTCCAGAGTCTTTCCTTTTTG
1452 RAATTCAAAAAGGAAAGACTCTGGAACAAATCTCTCTTGAAGATTTGTTCCAGAGTCTTTCC
1863 F CCGGGCCTTATCGGGAAGGAAATTTTTCAAGAGAAAATTTCCTTCCCGATAAGGCTTTTTG
1863 RAATTCAAAAAGCCTTATCGGGAAGGAAATTTTCTCTTGAAAAATTTCCTTCCCGATAAGGC
Table 2. Primers used for qPCR of SLC2A8 mRNA.
Table 2. Primers used for qPCR of SLC2A8 mRNA.
GeneAccession NumberFwd (5′ → 3′)Rev (5′ → 3′)Amplicon Size (bp)
SLC2A8Y17801.1ATGTGCTTCATGCCCGAGACCTGGATGACACCCACGACGA311
RPS15NM_001018TTCCGCAAGTTCACCTACCCGGGCCGGGCATGCTTTACG361
Table 3. Sequencing, quality control, and mapping metrics for control and SLC2A8-deficient ACH-3P cells.
Table 3. Sequencing, quality control, and mapping metrics for control and SLC2A8-deficient ACH-3P cells.
Control ACH-3P CellsSLC2A8-Deficient ACH-3P Cells
12341234
Row reads101,985,422119,212,310117,319,316122,204,792122,233,048138,670,320127,242,2421.01 × 108
Mapped reads58,421,94467,375,77265,818,39066,997,25267,896,14874,225,83869,811,36854,391,282
Uniquely mapped reads56,626,19465,017,28463,508,56064,650,00665,217,45271,508,53067,218,32252,358,886
Multi-mapped reads1,789,4242,348,6642,301,0562,338,9342,668,1142,706,9062,584,9222,024,950
Too many loci632698248774831210,58210,40281247446
Expressed transcripts58,14760,40559,87260,47660,60662,10661,76458,481
Expressed genes24,08425,15424,97124,90924,91025,19424,96524,357
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lipka, A.; Paukszto, Ł.; Kennedy, V.C.; Tanner, A.R.; Majewska, M.; Anthony, R.V. The Impact of SLC2A8 RNA Interference on Glucose Uptake and the Transcriptome of Human Trophoblast Cells. Cells 2024, 13, 391. https://doi.org/10.3390/cells13050391

AMA Style

Lipka A, Paukszto Ł, Kennedy VC, Tanner AR, Majewska M, Anthony RV. The Impact of SLC2A8 RNA Interference on Glucose Uptake and the Transcriptome of Human Trophoblast Cells. Cells. 2024; 13(5):391. https://doi.org/10.3390/cells13050391

Chicago/Turabian Style

Lipka, Aleksandra, Łukasz Paukszto, Victoria C. Kennedy, Amelia R. Tanner, Marta Majewska, and Russell V. Anthony. 2024. "The Impact of SLC2A8 RNA Interference on Glucose Uptake and the Transcriptome of Human Trophoblast Cells" Cells 13, no. 5: 391. https://doi.org/10.3390/cells13050391

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop