• 검색 결과가 없습니다.

I compared the genomic profiles of MSCs in a group of four patients and controls. DEGs were measured by counting tags from osteoporosis samples against normal samples and were normalized using the trimmed mean of M-values normalization method (TMM) method (edgeR).21 Significantly upregulated genes of MSCs in osteoporosis were those with q-values < 0.05 and log2 fold changes (FC) >

2, whereas the control groups were defined as q-values < 0.05 and log2 FC < -2. For pathway enrichment analysis of DEGs, the right-tailed Fisher’s exact test was used to calculate a p-value to determine the probability so that the biological function assigned to a specific osteoporosis-induced network was due to chance alone.

For the RT-PCR analyses and subjects’ baseline characteristics, statistical comparisons were performed using IBM SPSS Statistics 23 (SPSS Inc., Chicago, IL, USA), and the data are presented as the means ± standard error of means (SEMs).

III. RESULTS

I compared the transcriptome of MSCs from four patients (aged 62–74 years) with severe osteoporosis and MSCs of the age-matched control group with normal BMD (60–71 years old) (Table 2). There were no statistically significant differences in the baseline characteristics such as age, height, weight, and BMI. The control group showed higher lumbar spine BMD compared to osteoporosis group but did not show statistical significancebecause of the degenerative changes of the study subjects’

spines. However, the BMDs for femur neck and total hip were significantly lower in the osteoporosis group.

Table 3 lists the 53 most highly expressed DEGs between the osteoporosis and normal subjects, which included 28 and 25 genes that were expressed at higher and lower levels, respectively, in osteoporosis subjects compared to normal subjects.

C-type lectin domain family 2 member B (CLEC2B, FC=5), endogenous retrovirus group K member 5 Gag polyprotein (LOC105374013, FC=5), NR4A1 (FC=4.2), NR4A3 (FC=4.15), and heparin-binding EGF-like growth factor (HBEGF, FC=3.17) were upregulated whereas the remaining DEGs, namely, matrix metallopeptidase 13 (MMP13, FC=−8), septin 7 (SEPT, FC=−8), solute carrier family 26 member 7 (SLC26A7, FC=−8), and sparc/osteonectin, cwcv, and kazal-like domains proteoglycan 3 (SPOCK3, FC=-8), were downregulated.

Table 2. Clinical characteristics of the study subjects for RNA-seq.

Control (n=4) Osteoporosis (n=4) p-value

Age, yrs 67.3±2.5 68.5±3.0 0.686

Height, cm 153.2±2.0 152.1±0.9 0.486

Weight, kg 76.3±2.8 63.3±3.6 0.057

BMI, kg/m2 32.6±1.2 27.3±1.3 0.114

Serum parameters

Calcium, mg/dL 9.5±0.3 9.8±0.4 0.686

Phosphorus, mg/dL 3.8±0.3 4.2±0.2 0.200

25(OH)D, ng/mL 14.2±3.0 17.0±3.2 0.486

Bone mineral density

Lumbar spine, g/cm2 1.181±0.062 0.902±0.117 0.114

Femur Neck, g/cm2 0.893±0.012 0.641±0.009 0.029

Total Hip, g/cm2 0.980±0.021 0.697±0.025 0.029

Data are mean ± SEM

Table 3. Fifty-three up- or downregulated genes in mesenchymal stem cells of osteoporosis subjects compared to controls by RNA-seq data.

Upregulated in the osteoporosis subjects

Gene symbol Entrez Gene name

log2 fold change

p-value q-value

CLEC2B C-type lectin domain family 2 member B 5 0.0002 0.04702 LOC105374013 endogenous retrovirus group K member 5 Gag

polyprotein 5 0.00005 0.01646

NR4A1 nuclear receptor subfamily 4 group A member 1 4.2 0.00005 0.01646 NR4A3 nuclear receptor subfamily 4 group A member 3 4.15 0.00005 0.01646 HBEGF heparin-binding EGF-like growth factor 3.17 0.00005 0.01646

EGR3 early growth response 3 3.13 0.00005 0.01646

NEFM neurofilament, medium polypeptide 3.13 0.00005 0.01646 NR4A2 nuclear receptor subfamily 4 group A member 2 3.09 0.00005 0.01646

IL11 interleukin 11 2.86 0.0002 0.04703

KRTAP2–2 keratin associated protein 2–2 2.53 0.00005 0.01646

CDC20 cell division cycle 20 2.39 0.00005 0.01646

DLGAP5 discs, large (Drosophila) homolog-associated

protein 5 2.37 0.00005 0.01646

ESM1 endothelial cell-specific molecule 1 2.36 0.00005 0.01646

DLX2 distal-less homeobox 2 2.33 0.0002 0.04702

GEM GTP binding protein overexpressed in skeletal

muscle 2.25 0.00015 0.0395

PTTG1 pituitary tumor-transforming 1 2.18 0.0001 0.02992

CEP55 centrosomal protein 55kDa 2.14 0.00005 0.01646

DUSP5 dual specificity phosphatase 5 2.09 0.00005 0.01646

HAS1 hyaluronan synthase 1 2.06 0.00005 0.01646

TK1 thymidine kinase 1, soluble 2.05 0.00005 0.01646

BUB1 BUB1 mitotic checkpoint serine/threonine

kinase 2.03 0.00005 0.01646

RRM2 ribonucleotide reductase regulatory subunit M2 2.02 0.0001 0.02992

CCNB1 cyclin B1 2 0.00005 0.01646

SERPINB10 serpin peptidase inhibitor, clade B (ovalbumin),

member 10 2 0.00005 0.01646

MLPH melanophilin 1.88 0.0002 0.04706

SHCBP1 SHC SH2-domain binding protein 1 1.83 0.00005 0.01646 TACC3 transforming, acidic coiled-coil containing

protein 3 1.76 0.0001 0.02992

CSRNP1 cysteine-serine-rich nuclear protein 1 1.64 0.00015 0.0395 Downregulated in the osteoporosis

subjects

Gene symbol Entrez Gene name

log2 fold change

p-value q-value

CFHR1 complement factor H-related 1 -8 0.00005 0.01646 FRMPD2 FERM and PDZ domain containing 2 -8 0.00005 0.01646

MMP13 matrix metallopeptidase 13 -8 0.00015 0.0395

PKD1 polycystic kidney disease 1 (autosomal

dominant) -8 0.00015 0.0395

RPL13 ribosomal protein L13 -8 0.00005 0.01646

SETP7 septin 7 -8 0.00005 0.01646

SLC26A7 solute carrier family 26 (anion exchanger),

member 7 -8 0.00005 0.01646

SPOCK3 sparc/osteonectin, cwcv and kazal-like domains

proteoglycan 3 -8 0.00005 0.01646

HAPLN1 hyaluronan and proteoglycan link protein 1 -7.26 0.00005 0.01646

MMP1 matrix metallopeptidase 1 -4.8 0.00005 0.01646

NPTX1 neuronal pentraxin I -4.39 0.00005 0.01646

COMP cartilage oligomeric matrix protein -3.45 0.00005 0.01646 NTRK2 neurotrophic tyrosine kinase, receptor, type 2 -2.77 0.00005 0.01646 COL11A1 collagen, type XI, alpha 1 -2.39 0.00005 0.01646

ITGA10 integrin subunit alpha 10 -2.31 0.0001 0.02992

ITGB2 integrin subunit beta 2 -2.28 0.00005 0.01646

FGFR2 fibroblast growth factor receptor 2 -2.22 0.00005 0.01646

PTGS1 prostaglandin-endoperoxide synthase 1 -2.14 0.00005 0.01646

ACAN aggrecan -1.91 0.00005 0.01646

MOXD1 monooxygenase, DBH-like 1 -1.72 0.0002 0.04702

PDK4 pyruvate dehydrogenase kinase 4 -1.72 0.00005 0.01646

ITGB8 integrin subunit beta 8 -1.6 0.00005 0.01646

FNDC1 fibronectin type III domain containing 1 -1.59 0.00015 0.0395

SYNPO2 synaptopodin 2 -1.45 0.00015 0.0395

EDIL3 EGF-like repeats and discoidin I-like domains 3 -1.42 0.0002 0.04702

The interactions among the identified DEGs were examined using IPA. RNA-seq data were mapped onto the networks to explore dynamic changes during the MSC process. The biological functions that were significantly associated with the genes in the core networks were identified by functional analysis based on the Ingenuity’s knowledge base. In all, 33 candidate network genes and 54 candidate functions/pathways genes were found. Table 4 shows the most significant gene networks of the DEGs of MSCs in osteoporosis patients. The top functions were related to cellular development, differentiation of cells, differentiation of connective tissue cells, cellular growth, and proliferation. Moreover, the differentiation of adipocytes was also related to the identified DEGs (NR4A1, NR4A2 and NR4A3).

I also investigated IPA canonical pathways in the DEGs of MSCs in osteoporosis patients (Table 5). The associated effects were categorized in the mitotic roles of polo-like kinase, integrin signaling, Signal transducer and activator of transcription 3 (STAT3) pathway, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, cell cycle: G2/M DNA damage checkpoint regulation and cell cycle regulation. In addition, pathways related to osteoblasts, osteoclasts, and chondrocytes in rheumatoid arthritis were altered. Interestingly, changes in the adipogenesis pathway were also found.

Table 4. Enriched function annotation of differentially expressed gene sets using ingenuity pathway analysis.

Functions

Annotation p-value Molecules

Cartilage

development 4.72E-08 COL11A1, DLX2, HAPLN1, HBEGF, ITGA10, ITGB8, MMP13

Development of

connective tissue 5.20E-07 COL11A1, DLX2, HAPLN1, HBEGF, IL11, ITGA10, ITGB8, MMP13

Width of hypertrophic chondrocyte layer

6.22E-06 FGFR2, ITGA10, MMP13, PKD1

Quantity of

leukocytes 2.11E-05

ACAN, BUB1, EGR3, FGFR2, HBEGF, IL11, ITGB2, ITGB8, MMP13, NR4A1, PTTG1, SHCBP1, TACC3

Quantity of T-

lymphocytes 1.26E-04 ACAN, EGR3, FGFR2, ITGB2, ITGB8, NR4A1, PTTG1, SHCBP1, TACC3

Generation of cells 7.23E-04 COL11A1, EGR3, IL11, ITGB2, ITGB8, NR4A1, NR4A2, PTGS1, TK1

Differentiation of

lymphocytes 1.17E-03 DUSP5, EGR3, ITGB2, ITGB8, NR4A1, NR4A2, TK1

Development of

blood cells 1.28E-03 EGR3, IL11, ITGB2, ITGB8, NR4A1, NR4A2, PTGS1, TK1

T cell development 1.60E-03 EGR3, ITGB2, ITGB8, NR4A1, NR4A2, PTGS1, TK1

Cellular

homeostasis 2.05E-03 EGR3, ITGB2, ITGB8, NR4A1, NR4A2, PDK4, PTGS1, TK1

Quantity of

macrophages 3.14E-03 HBEGF, IL11, ITGB2, SHCBP1

Differentiation of 3.28E-03 DUSP5, EGR3, FGFR2, IL11, ITGB2, ITGB8,

cells NR4A1, NR4A2, NR4A3, TK1 Differentiation of T-

lymphocytes 7.03E-03 EGR3, ITGB2, ITGB8, NR4A1, NR4A2 Quantity of

thymocytes 7.53E-03 EGR3, ITGB2, NR4A1, TACC3 Quantity of CD4+

T-lymphocytes 1.40E-02 ACAN, ITGB2, SHCBP1 Quantity of

phagocytes 1.44E-02 HBEGF, IL11, ITGB2, MMP13, SHCBP1 Development of

2.11E-02 FGFR2, IL11, NR4A1, NR4A2, NR4A3

Development of

lymphatic system 2.31E-02 EGR3, IL11, TK1 Differentiation of

adipocytes 2.58E-02 NR4A1, NR4A2, NR4A3

Apoptosis 2.76E-02 EGR3, FGFR2, IL11, ITGB2, NR4A1, PTTG1

Table 5. Canonical ingenuity pathway analysis of the 53 differentially expressed genes of the mesenchymal stem cells of osteoporosis subjects.

Ingenuity Canonical Pathways log(p-value) Ratio Molecules

Mitotic Roles of Polo-Like Kinase 3.11E+00 4.92E-02 CDC20, PTTG1, CCNB1

Leukocyte Extravasation Signaling 2.62E+00 2.09E-02 ITGB2, EDIL3, MMP13, MMP1

Oncostatin M Signaling 2.35E+00 5.88E-02 MMP13, MMP1

Role of IL-17F in Allergic Inflammatory

Airway Diseases 2.19E+00 4.88E-02 MMP13, IL11

Role of IL-17A in Arthritis 1.96E+00 3.70E-02 MMP13, MMP1 Granulocyte Adhesion and Diapedesis 1.90E+00 1.83E-02 ITGB2, MMP13,

MMP1

Agranulocyte Adhesion and Diapedesis 1.86E+00 1.76E-02 ITGB2, MMP13, MMP1

Integrin Signaling 1.72E+00 1.55E-02 ITGB2, ITGA10,

ITGB8

STAT3 Pathway 1.71E+00 2.74E-02 NTRK2, FGFR2

Role of Osteoblasts, Osteoclasts and

Chondrocytes in Rheumatoid Arthritis 1.63E+00 1.44E-02 MMP13, MMP1, IL11

DNA damage-induced 14-3-3σ Signaling 1.29E+00 5.56E-02 CCNB1

GADD45 Signaling 1.26E+00 5.26E-02 CCNB1

Complement System 1.07E+00 3.33E-02 ITGB2

NF-κB Signaling 1.06E+00 1.19E-02 NTRK2, FGFR2

ILK Signaling 1.04E+00 1.17E-02 ITGB2, ITGB8

IL-8 Signaling 1.02E+00 1.14E-02 ITGB2, HBEGF

IL-17A Signaling in Fibroblasts 1.01E+00 2.86E-02 MMP1 MSP-RON Signaling Pathway 8.95E-01 2.17E-02 ITGB2 Cell Cycle: G2/M DNA Damage Checkpoint

Regulation 8.78E-01 2.08E-02 CCNB1

Eicosanoid Signaling 8.02E-01 1.72E-02 PTGS1

Calcium-induced T Lymphocyte Apoptosis 7.88E-01 1.67E-02 NR4A1

CD40 Signaling 7.63E-01 1.56E-02 PTGS1

NF-κB Activation by Viruses 7.11E-01 1.37E-02 ITGB2 Role of Macrophages, Fibroblasts and

Endothelial Cells in Rheumatoid Arthritis 6.96E-01 7.12E-03 MMP13, MMP1 Cyclins and Cell Cycle Regulation 6.95E-01 1.32E-02 CCNB1

G Beta Gamma Signaling 6.57E-01 1.19E-02 HBEGF

Corticotropin Releasing Hormone Signaling 5.76E-01 9.62E-03 NR4A1

RhoA Signaling 5.55E-01 9.09E-03 SEPT7

Role of Pattern Recognition Receptors in

Recognition of Bacteria and Viruses 5.26E-01 8.40E-03 IL11

Adipogenesis pathway 4.97E-01 7.75E-03 FGFR2

Signaling by Rho Family GTPases 3.28E-01 4.69E-03 SEPT7 Protein Ubiquitination Pathway 2.84E-01 4.07E-03 CDC20 Glucocorticoid Receptor Signaling 2.63E-01 3.77E-03 MMP1

IPA software enables the systemic analysis of RNA-seq and other data in a biologic context. My up- or downregulated genes were overlaid onto a global molecular network developed from information contained in the Ingenuity Pathways Knowledge Base. Then the networks of these focus genes were algorithmically generated based on their interrelationships. Fig.2 includes several genes encoding the regulators of osteoblasts, osteoclasts, and chondrocytes in rheumatoid arthritis and glucocorticoid receptor signaling. In the network, the NR4A family is associated with the roles of these cells in rheumatoid arthritis.

To elucidate the common pathways related to osteoporosis and adipogenesis, the DEGs in the obesity-related network were identified from among the 53 DEGs using IPA (Fig.3). Numerous DEGs were related to the obesity pathway, such as the NR4A family, matrix metallopeptidase 1 (MMP1), and fibroblast growth factor receptor 2 (FGFR2). Among them, the NR4A family was highly upregulated in osteoporosis patients and was closely related to the adipogenesis pathway. In addition, TNF, which is an upstream regulator of the NR4A family, is related to the adipogenesis pathway, NF-κB signaling and glucocorticoid receptor signaling.

Fig. 2. Top networks showing interactions between selected differentially expressed genes in the mesenchymal stem cells (MSCs) of osteoporosis subjects.

Red indicates transcripts upregulated in the MSCs of osteoporosis subjects. Green indicates transcripts downregulated in the MSCs of osteoporosis subjects. White indicates transcripts not differentially expressed in my study but important in the network. The color gradient in the network indicates the strength of the expression denoted by fold changes. (→) indicates direct interaction between the transcript products; (--->) indicates indirect interaction between the transcript products; ⟲ indicates autoregulation.

Fig. 3. Differentially expressed genes (DEGs) in the obesity-related network among the 53 DEGs using ingenuity pathway analysis. Numerous DEGs were related to the obesity pathway, such as the NR4A family, matrix metallopeptidase 1 (MMP1), and fibroblast growth factor receptor 2 (FGFR2). Among them, the NR4A family was highly upregulated in osteoporosis patients and was closely related to the adipogenesis pathway. Red indicates transcripts upregulated in the mesenchymal stem cells (MSCs) of osteoporosis subjects. Green indicates transcripts downregulated in the MSCs of osteoporosis subjects. White indicates transcripts not differentially expressed in my study but important in the network.

The color gradient in the network indicates the strength of expression denoted by fold changes. (→) indicates direct interaction between the transcript products; (--->) indicates indirect interaction between the transcript products; ⟲ indicates autoregulation.

To validate this family as candidate common genes related to osteoporosis and adipogenesis, the expression levels of the mRNAs of NR4A1, NR4A2, and NR4A3 were compared using semi-quantitative RT-PCR with other independent subjects (two osteoporosis patients and controls, two obese patients and non-obese controls with similar BMDs (T-score < -1 and < -2.5]) (Fig.4). The baseline characteristics of these subjects are shown in Table 6. The expression level of NR4A1 mRNA was significantly higher in osteoporosis patients than in controls (p=0.018). However, there were also nonsignificant trends of increased expression of NR4A2 (p=0.056) and NR4A3 (p=0.664) in osteoporosis patients compared to controls. The expression levels of NR4A2 mRNA were significantly higher in obese patients than in controls (p=0.041). There were also trends of increased expression of NR4A1 (p=0.450) and NR4A3 (p=0.308) in obese patients compared to controls. I also compared the expression of the NR4A family between the MSCs of ovariectomized and sham-operated mice using RT-PCR. NR4A1 and NR4A3 were significantly upregulated in ovariectomized mice (p<0.001) (Fig.4).

Fig .4. Increased expression of the NR4A family in other osteoporosis subjects (A), obese subjects (B), and ovariectomized mice (C). Data are expressed as the expression level of NR4A1–3 relative to the expression level of actin. Sham: sham-operated DDY female mice; Ovx: ovariectomized DDY female mice.

Table 6. Clinical characteristics of the study subjects for validation of NR4A family as candidate common genes related to osteoporosis and adipogenesis.

spine, g/cm2 1.180±0.164 0.719±0.023 0.108 1.200±0.235 0.979±0.058 0.515 Femur

Neck, g/cm2 0.875±0.030 0.610±0.034 0.028 0.724±0.028 0.730±0070 0.952 Total Hip,

g/cm2 1.083±0.050 0.696±0.030 0.022 0.761±0.011 0.892±0.101 0.417 Data are mean ± SEM

IV. DISCUSSION

I identified DEGs between postmenopausal osteoporosis patients and controls with normal BMD using RNA-seq data from human MSCs. Accurate NGS technology was utilized to perform RNA-seq. With the identified DEGs, the common pathways related to osteoporosis and adipogenesis were evaluated using IPA. The NR4A family may represent common genes between osteoporosis and adipogenesis.

MSCs may be the best resource for research on the relationship between bone and fat because they are the common origins of osteoblasts and adipocytes.6 With these MSCs, RNA-seq was performed to identify DEGs between osteoporosis patients and controls using the reliable NGS technology13-15,22. The DEGs between postmenopausal osteoporosis patients and controls were rigorously identified by applying a false discovery rate of q <0.05 and log2 FC >2 or <-2. In all, 53 DEGs were identified. Well-known genes related to osteoporosis such as Runt-related transcription factor 2 (Runx2), Low-density lipoprotein receptor-related protein 5 (LRP5), and SOST23 were not different between the osteoporosis and normal subjects, suggesting that it occurs at a later stage in the osteoblast lineage than that reflected by the MSCs.24 However, pathway analysis revealed that the DEGs were closely related to the differentiation of cells, particularly for connective tissue cells, suggesting that the genetic changes in MSCs related to the differentiation of these cells could affect the pathogenesis of osteoporosis. Interestingly, enriched function annotation from IPA analyses of DEG sets revealed that the differentiation of adipocytes was also related to the identified DEGs (NR4A1, NR4A2, and NR4A3).

The RNA-seq data showed that the NR4A family was upregulated in osteoporosis patients. According to the baseline characteristics of the subjects, there were no significant differences in the weight and BMI between osteoporosis and controls;

rather, the weight and BMI seemed to be lower in osteoporosis patients. Therefore, it was not possible that the obesity of the osteoporosis group affected the upregulation of the NR4A family. Hence, it could be inferred that the genetic preference of MSCs for adipogenesis might affect the pathogenesis of osteoporosis. In fact, bone and fat cells share a common origin (i.e., MSCs), and their fates are inter-related in both the physiologic and pathologic states.6

The NR4A subfamily of orphan nuclear receptors is made up of NR4A1/NUR77/NBFIB, NR4A2/NURR1, and NR4A3/NOR1.25The NR4As are early responders to stimuli such as ß-adrenergic signaling, insulin, growth factors, glucose, lipopolysaccharide, lipids, glucagon, inflammation, and various forms of cellular stress.25-27 These transcription factors are also involved in various functions such as cell cycle regulation, apoptosis, steroidogenesis, adipogenesis, and energy metabolism.25-26,28 Moreover, there is strong upregulation of NR4As in cases of extreme obesity, and these levels normalize after fat loss.25 This suggests the potential relationship between the NR4A family and obesity. The NR4A family is also induced by parathyroid hormone in bone.29-31 In an in-vitro study, there was cross-talk betweenthe ß-catenin signaling pathway and NR4A orphan nuclear receptorsin osteoblasts. NR4A receptors repressed ß-catenin-mediatedtransactivation, which is crucial in the developmentand function of bone tissue32. This also suggests

the possible relationship between the NR4A family and bone metabolism. My IPAs also revealed a possible relationship between NR4A1 and corticotropin-releasing hormone (CRH) signaling. NR4A2 is related not only to the adipogenesis pathway but also to glucocorticoid receptor signaling through TNF. Glucocorticoids markedly enhance marrow adipogenesis at the expense of osteoblast differentiation.10 Therefore, the NR4A family might represent commonly related genes for bone and fat metabolism via CRH signaling. NR4A1 and NR4A2 enhance transcriptional activity of the CRH and proopiomelanocortin (POMC) promoters, while also being upregulated by CRH treatment in pituitary cells, thereby mediating expression of hypothalamic and pituitary stress hormones33-34. In fact, RT-PCR indicated increased expression of the NR4A family in other osteoporosis and obese subjects compared to controls, although the number of subjects for the validation was small and the results were not completely consistent. However, the osteoporosis patients used for RNA-seq were not obese according to their BMIs, and the adipogenesis pathway was upregulated in these subjects. Increased marrow adipogenesis could be suggested as a reason. Although the amount of marrow fat for the subjects was not measured in this study, an inverse association between marrow adipose tissue and measures of bone density and strength in adults has been reported.35Aging is associated with a significant increase in marrow adiposity.36Excess marrow fat has been implicated in the pathogenesis of bone fragility in premenopausal women with idiopathic osteoporosis12 and anorexianervosa.36In this study, I also compared the expression of the NR4A family between the MSCs of ovariectomized and sham-operated mice using RT-PCR to investigate if these candidate genes could be commonly related to

the postmenopausal model. NR4A1 and NR4A3 were significantly upregulated in the MSCs of ovariectomized mice. Both in vitro and in vivo studies have shown that various conditions that promote bone marrow adipogenesis, particularly estrogen withdrawal, limit osteoblastogenesis.37The absence of estrogen receptor- activation accelerates the development of marrow adipocytes in ovariectomized animals.36The upregulated NR4A family might affect the pathogenesis of osteoporosis by increased marrow adipogenesis rather than systemic obesity.

This study had some limitations. First, the sample size of four subjects for each group may appear to be small. However, Itried to recruit the subjects of each group with prominent differences in medical characteristics to identify definite DEGs.

I recruited controls with normal BMD without any medical history that would have affected bone metabolism. Moreover, the osteoporosis subjects were also carefully selected. Subjects with a definite medical history of osteoporotic fracture were recruited in this study, not only those with low BMD (T-score ≤ -2.5). Despite the small sample number, I identified distinct transcriptional changes in osteoporosis between two different groups with a reliable medical history. In addition, due to the polygenic architecture of osteoporosis, the osteoporosis trait arises in each individual from the combined effects of a large number of genetic variants.38-39 In most cases, there is sufficient statistical power to identify some of these genetic variants even with a small sample size.39Second, I only analyzed the transcriptional differences related to the molecular genetic mechanism in osteoporosis. While individual omics studies (genome/transcriptome/epigenome/proteome) are useful for identifying molecular genetic mechanisms, defects at any level (DNA, mRNA, epigenes, and

proteins) may or may not translate into the next level(s), ultimately impacting disease risk. Moreover, epigenetic factors (epigenes) and their interactions (such as those between DNA methylation and miRNA) modulated by the environment affect gene expression into mRNAs/proteins and mRNA stability.39 Further multi-omics studies are needed to verify the identified DEGs in the molecular genetic mechanism in osteoporosis.

However, this study identified DEGs related to osteoporosis with the MSCs of reliable study subjects using NGS. Using MSCs as the common origin of osteoblasts and adipocytes, I searched for a common pathway related to adipogenesis and osteoporosis via IPA. Interestingly, the NR4A family was found as a possible candidate gene for the common pathway. I also validated these genes with other MSC samples from independent subjects. To the best of my knowledge, these procedures have not been attempted to investigate a common mechanism related to osteoporosis and adipogenesis. In this regard, this study has strengths. Moreover, these procedures using MSCs might be applicable to research for common

However, this study identified DEGs related to osteoporosis with the MSCs of reliable study subjects using NGS. Using MSCs as the common origin of osteoblasts and adipocytes, I searched for a common pathway related to adipogenesis and osteoporosis via IPA. Interestingly, the NR4A family was found as a possible candidate gene for the common pathway. I also validated these genes with other MSC samples from independent subjects. To the best of my knowledge, these procedures have not been attempted to investigate a common mechanism related to osteoporosis and adipogenesis. In this regard, this study has strengths. Moreover, these procedures using MSCs might be applicable to research for common

관련 문서