Acessibilidade / Reportar erro

HOX gene analysis in the osteogenic differentiation of human mesenchymal stem cells

Abstract

Human bone marrow-derived mesenchymal stem cells (hMSCs) have the capacity to differentiate into osteoblasts during osteogenesis. Several studies attempted to identify osteogenesis-related genes in hMSCs. Although HOX genes are known to play a pivotal role in skeletogenesis, their function in the osteogenesis of hMSCs has not yet been investigated in detail. Our aim was to characterize the expression of 37 HOX genes by multiplex RT-PCR to identify the ones most probably involved in osteogenic differentiation. The results showed that the expression patterns of four HOX genes were altered during this process. In particular, the expression levels of HOXC13 and HOXD13 were dramatically changed. Real-time PCR and Western blot analysis were performed in order to further analyze the expression of HOXC13 and HOXD13 . The qRT-PCR results showed that transcription of HOXC13 was up-regulated by up to forty times, whereas that of HOXD13 was down-regulated by approximately five times after osteogenic differentiation. The Western blot results for the HOXC13 and HOXD13 proteins also corresponded well with the real-time PCR result. These findings suggest that HOXC13 and HOXD13 might be involved in the osteogenic differentiation of hMSCs.

human mesenchymal stem cells; HOX genes; osteogenic differentiation; stem cell differentiation; gene profiling


ABSTRACT

GENETICS

HOX gene analysis in the osteogenic differentiation of human mesenchymal stem cells

Song Wha Chae1 * * These authores contributed equally to this work.

Bo Keun Jee1 * * These authores contributed equally to this work.

Joo Yong Lee1

Chang Whan Han2

Yang-Whan Jeon3

Young Lim4

Kweon-Haeng Lee1,5

Hyoung Kyun Rha1

Gue-Tae Chae6

1Neuroscience Genome Research Center, The Catholic University of Korea, Seoul, Republic of Korea

2Department of Orthopedic Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea

3Department of Psychiatry, Our Lady of Mercy Hospital, The Catholic University of Korea, Incheon, Republic of Korea

4Department of Occupational and Environmental Medicine, St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea

5Department of Pharmacology, The Catholic University of Korea, Seoul, Republic of Korea

6Institute of Hansen's Disease, The Catholic University of Korea, Seoul, Republic of Korea

Human bone marrow-derived mesenchymal stem cells (hMSCs) have the capacity to differentiate into osteoblasts during osteogenesis. Several studies attempted to identify osteogenesis-related genes in hMSCs. Although HOX genes are known to play a pivotal role in skeletogenesis, their function in the osteogenesis of hMSCs has not yet been investigated in detail. Our aim was to characterize the expression of 37 HOX genes by multiplex RT-PCR to identify the ones most probably involved in osteogenic differentiation. The results showed that the expression patterns of four HOX genes were altered during this process. In particular, the expression levels of HOXC13 and HOXD13 were dramatically changed. Real-time PCR and Western blot analysis were performed in order to further analyze the expression of HOXC13 and HOXD13. The qRT-PCR results showed that transcription of HOXC13 was up-regulated by up to forty times, whereas that of HOXD13 was down-regulated by approximately five times after osteogenic differentiation. The Western blot results for the HOXC13 and HOXD13 proteins also corresponded well with the real-time PCR result. These findings suggest that HOXC13 and HOXD13 might be involved in the osteogenic differentiation of hMSCs.

Bone marrow-derived stem cells can be divided into two major types: hematopoietic stem cells and nonhematopoietic, or mesenchymal, stem cells. Human bone marrow-derived mesenchymal stem cells (hMSCs) have the capacity for self-renewal and multilineage differentiation. Under the appropriate conditions, they can also give rise to mesenchymal tissues such as muscle, bone, fat, and cartilage (Pittenger et al., 1999). Due to their ability to differentiate into osteoblasts, chondrocytes, adipocytes, tenocytes and myoblasts, hMSCs hold promise for clinical applications in regenerative medicine (Song et al., 2006).

Because osteoblastic cells play a major role in the processes of normal bone growth, remodeling and fracture repair, many researchers have used the process of osteogenesis to study the differentiation and characteristics of stem cells (Kraus and Kirker-Head, 2006). To obtain osteoblastic cells, MSCs are incubated with a mixture medium containing dexamethasone, β-glycerophosphate and ascorbic acid for a period of 2~3 weeks (Bobis et al., 2006).

HOX genes were initially identified by their homology with the Drosophila HOM genes (Levine et al., 1984; Acampora et al., 1989; Duboule and Dolle, 1989). These genes encode homeodomain transcription factors related to anterior-posterior axis patterning that takes place during embryonic development (van den Akker et al., 2001). The homeodomain contains a 180-base-pair homeobox sequence that encodes a conserved 60 amino acid region and acts as a DNA-binding domain via a helix-turn-helix motif (Gehring et al., 1994). In vertebrates, 39 HOX genes have been identified. These are distributed over four homologous HOX clusters termed HOXA, B, C, and D. These loci are located on four different chromosomal locations and are comprised of nine to eleven genes (Akin and Nazarali, 2005). It is well known that HOX proteins participate in many common developmental processes during normal embryogenesis. Several reports have indicated that HOX genes play a regulatory role in skeletogenesis (Goff and Tabin, 1997; Kanzler et al., 1998; van den Akker et al., 2001; Remacle et al., 2004).

Although HOX genes are known to play an essential role in skeletal development and bone formation, there is no report regarding the screening of HOX groups that are involved in the osteogenesis of hMSCs. Thus, in the present study, the expression profile of HOX genes during osteogenic differentiation of hMSCs was investigated by multiplex PCR and the results showed significant changes in the expression of four of them during this process. Of these four genes, the expression of HOXC13 and HOXD13 showed the most dramatic changes. Therefore, the expression levels of HOXC13 and HOXD13 were evaluated by qRT-PCR and Western blot analysis, and the results were similar to the multiplex PCR result. This suggests that the other two genes (HOXA1 and HOXC11) are also involved in osteogenesis.


Figure 1 Phenotypic characterization of hMSCs using flow cytometric analysis. FACS analysis showed that the cells were negative for CD11b, CD34 and CD45 expression and positive for CD29, CD73 and CD105, which are phenotypes currently known to be characteristic of hMSCs. The gray line indicates the control of the CD marker isotypes.


Figure 2 Osteogenic differentiation of hMSCs. (A) ALP staining and von Kossa staining. Osteogenic differentiation was confirmed by ALP staining. The cells stained positively for endogenous ALP activity during 21 days of culture in osteogenic media. The von Kossa staining also showed an increase in calcium deposition during differentiation. Scale bar = 30 μm. (B) Expression of osteogenesis-specific genes (ALP, BSP, and OCN) was observed by RT-PCR at day 0, 10 and 21 of the culture period. GAPDH was used as control. The expression levels of the osteogenesis-specific genes increased during osteogenic differentiation. The early osteogenesis marker ALP showed increased expression at day 10, and the expression of late osteogenesis markers (BSP, OCN) was observed at day 21. (C) Immunoblotting analysis of OCN and OPN expression. Whole-cell proteins obtained on day 0, day 10 and day 21 were blotted onto a nitrocellulose membrane. The expression levels of the OCN and OPN proteins increased during osteogenic differentiation. β-actin was used as control.

Figure 3
HOX multiplex PCR. (A) The mRNA expression of HOXA1, HOXC11 and HOXC13 increased during osteogenesis. The HOXC13 gene showed the most significant up-regulation. The expression of HOXA1 gradually increased during osteogenic differentiation. The expression of HOXC11 increased by approximately 30% and then returned to the expression level observed on day 0. (B) The mRNA expression of HOXD13 clearly decreased during osteogenic differentiation. The expression of HOXD13 decreased by approximately 70% on day 10; on day 21, the expression of HOXD13 was half the value observed on day 10. GAPDH was used as control. The graphs represent the mean ± SEM of six separate experiments. Asterisk (*) indicates a significant increase between two samples (p < 0.05).


Figure 4 Real-time PCR analysis of HOXC13 and HOXD1. The data were presented as fold changes relative to day 0. The mRNA expression of HOXC13 was five times higher on day 10 and 42 times higher on day 21 compared to the expression in a control. The expression of HOXD13 decreased rapidly at day 10 and slowly increased at day 21. The real-time PCR data were normalized with GAPDH expression. Asterisk (*) indicates a significant increase between two samples (p < 0.05).


Figure 5 Immunoblotting analysis of HOXC13 and HOXD13. Whole cell proteins obtained on day 0, day 10 and day 21 were blotted onto a nitrocellulose membrane. The protein level of HOXC13 showed a significant increase on day 21, whereas that of HOXD13 decreased gradually during osteogenic differentiation. β-actin was used as control. Asterisk (*) indicates a significant increase between two samples (p < 0.05).

The research protocol was reviewed and approved by the human ethical care committee at St. Mary's Hospital, Catholic University in Daejeon, Republic of Korea. The hMSCs were isolated from the bone marrow of six individuals, as described below (Choi et al., 2006). All experiments were performed with hMSCs obtained after the third cell passage.

hMSCs were analyzed by FACS-Calibur (Becton Dickinson, San Jose, CA) as previously described (Choi et al., 2006). FACS analysis was performed using fluorescein isothiocyanate (FITC)-conjugated anti-CD11b, CD29, CD34, CD45, CD73 and CD105 antibodies (BD Bioscience, San Diego, CA) to confirm that the phenotype of the hMSCs was maintained after expansion in the culture. The samples were incubated with antibodies against each surface marker for 30 min, and this treatment was followed by FACS.

To induce osteogenic differentiation, hMSCs at the third passage were plated with Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS) in a 250-ml tissue culture flask (Nunc, Roskilde, Denmark). The cells were then incubated at 37 °C in 5% CO2 for 24 h. The medium was replaced with high-glucose DMEM containing 10% FBS, 0.1 μM dexamethasone, 10 mM β-glycerophosphate, and 0.3 mM ascorbic acid (Sigma, St. Louis, MO) for osteogenic differentiation. This osteogenic medium was replaced every 2 days for 21 days.

About 3 x 105 cells were seeded onto each well of a 6-well plate. After incubation for 12 h at 37 °C in 5% CO2, the medium was replaced with osteogenic differentiation medium, replaced again every 2 days for periods of 10 and 21 days. The 10-day and 21-day differentiated and undifferentiated hMSCs were washed twice with ice-cold PBS (phosphate buffered saline), fixed with 2% paraformaldehyde/ 0.1 M sodium cacodylate for 10 min, and washed with 0.1 M cacodylic acid. The cells were incubated with ALP substrate solution (5 mg naphthol AS-TR phosphate in 25 mL water plus 10 mg Fast red TR in 24 mL of 0.1 M Tris buffer, pH 9.5) for 1 h at room temperature. Cells were photographed using a Nikon TE-300 (Tokyo, Japan) inverted light microscope.

Approximately 3 x 105 cells were seeded onto each well of a 6-well plate. After incubation for 12 h at 37 °C in 5% CO2, the medium was replaced with osteogenic differentiation medium and thereafter replaced every 2 days for periods of 10 and 21 days. Day-10 and day-21 differentiated and undifferentiated hMSCs were washed with distilled water, fixed with 4% formalin, and then treated with 5% silver nitrate. Then the cells were exposed to UV light for 1 h, 5% thiosulfate was added, and the cells were placed at room temperature after a washing step with distilled water. The samples were photographed with a Nikon TE-300 (Tokyo, Japan) inverted light microscope.

RNA isolation and reverse transcriptase-polymerase chain reaction (RT-PCR)

RT-PCR analysis was performed as described by Jee et al. (2006). After the induction of osteogenic differentiation for 21 days, total RNA was isolated from the cells using an RNeasy Mini Kit (QIAGEN, Valencia, CA). Two micrograms of total RNA were reverse-transcribed in order to synthesize cDNA, using an AccuPower RTPReMix kit (Bioneer, Inc., Rockville, MD). The subsequent PCR amplification was performed with 1 μL of RT reaction mixture, using the following thermocycling profile: 1 cycle at 94 °C for 5 min, followed by 30 cycles of 92 °C for 1 min, 52 °C for 1 min and 72 °C for 1 min, and a final cycle at 72 °C for 10 min. The primer sequences used are listed in Table 1. The GAPDH (glyceraldehyde-3-phosphate dehydrogenase) gene was used as an internal control. The PCR products were run on a 1% agarose gel and then analyzed under UV light after staining with ethidium bromide. The gel was photographed and then quantitatively measured by scanning densitometry. The experiments were performed with three different RNA samples.

Immunoblotting analysis was performed as previously described (Jee et al., 2007). After incubation with osteogenic differentiation medium for 21 days, the cells were lysed on ice for 30 min in RIPA buffer (50 mM Tris-HCl pH 7.5, 0.15 M NaCl, 1% NP-40, 0.1% SDS, 1 mM DTT and 1 mM PMSF) containing a mixture of protease inhibitors (Roche, Mannheim, Germany). The insoluble materials were separated using 10% polyacrylamide gels containing 10% sodium dodecyl sulfate (SDS), 1.5 M Tris-HCl, 0.035% N, N, N', N'-tetra-methylenediamine and 7 mg ammonium persulfate. The separated proteins were transferred to a nitrocellulose membrane (Whatman, Dassel, Germany) at 36 mA in a transfer buffer that contained 39 mM glycine, 48 mM Tris base, 0.037% SDS, and 20% methanol. The membranes were sequentially incubated with anti-OPN (osteopontin), OCN (osteocalcin), HOXC13 monoclonal antibodies (mAb; Abnova, Taipei, Taiwan), and HOXD13 mAb (Santa Cruz Biotechnology, Santa Cruz, CA) at 1:1000 dilutions. A horseradish peroxidase-conjugated anti-rabbit IgG was used as a secondary antibody at a dilution of 1:1500 (Pierce Biotechnology, Rockford, IL). Detection was performed with an electrochemiluminescence detection reagent (Amersham Biosciences, Uppsala, Sweden). In some cases, the Western blots were stripped and re-blotted with antibody, according to the manufacturer's instructions.

Multiplex PCR was performed using the GeneXP Human HOX Assay Kit (Seegene, Seoul, Republic of Korea). After the induction of osteogenic differentiation for 21 days, total RNA was isolated from the cells using an RNeasy Mini Kit (QIAGEN, Valencia, CA). Two micrograms of total RNA were reverse-transcribed in order to synthesize cDNA using an AccuPower RTPReMix kit (Bioneer, Inc., Rockville, MD). The synthesized cDNAs were used as templates for multiplex PCR,according to the manufacturer's instructions (http://www.seegene.com). PCR was carried out under the following conditions: 1 cycle at 94 °C for 15 min, followed by 40 cycles of 94 °C for 0.5 min, 63 °C for 1.5 min, and 72 °C for 1.5 min, and a final cycle at 72 °C for 10 min. GAPDH was used as an internal control. Electrophoresis was carried out on a 2% agarose gel. The multiplex PCR products were analyzed with the Alpha EaseFC software (Alpha Innotech, San Leandro, CA). The experiment was performed six times on each individual (Table 2).

After the induction osteogenic differentiation for 21 days, total RNA was isolated using an RNeasy Mini Kit (QIAGEN, Valencia, CA). Two micrograms of total RNA were reverse-transcribed in order to synthesize cDNA, using an AccuPower RTPReMix kit (Bioneer, Inc., Rockville, MD). For relative quantification, the reactions were performed in a total volume of 20 μL, containing 15 μL of LightCycler® FastStart DNA Master SYBR Green 1 (Roche Diagnostics, Mannheim, Germany), 10 ng of cDNA, and 10 pmol of each primer. Real-time quantitative PCR was carried out with specific primers, in a LightCycler Instrument (Roche Diagnostics, Mannheim, Germany). The samples were analyzed in triplicate. The primer sequences used are listed in Table 1. GAPDH was used as an internal control. For quantification, the data were analyzed using the LightCycler analysis software (Roche Diagnostics, Mannheim, Germany). Relative quantification of target gene expression was evaluated using the comparative CT method (Wang et al., 2004). The ΔCT value was determined by subtracting the target CT of each sample from its respective GAPDH CT value. Calculation of ΔCT involves using the mean ΔCT value of the control gene as an arbitrary constant to subtract from all other ΔCT mean values. Fold-changes in gene expression of the target gene were equivalent to 2 -ΔΔCT. The values obtained were then entered into a Student's t test. P values less than 0.05 were considered significant.

To investigate differentially expressed HOX genes during osteogenic differentiation from hMSCs, the data obtained from multiplex PCR were examined by variance analysis (ANOVA) with SPSS 12.0 software for Windows (SPSS, Chicago, IL). Tukey's HSD test was used for post hoc comparisons. For all statistical tests, an error probability of p < 0.05 was regarded as significant.

In an effort to explore the characterization of hMSCs, flow cytometry was used to examine the expression of the surface antigens CD11b, CD29, CD34, CD45, CD73, and CD105 in the isolated hMSCs. The isolated hMSCs were submitted to FACS analysis and found to be positive for CD29 (68 ± 2.5%), CD73 (96.9 ± 2.7%) and CD105 (91.5 ± 2.5%), and negative for CD11b, CD34 and CD45. These results show that the hMSCs were successfully isolated and that the culture-expanded hMSCs maintained their phenotype (Figure 1).

ALP and von Kossa staining were used to examine the differentiation of hMSCs into osteoblasts in the osteogenic medium. Although ALP staining at day 10 showed a weak color signal, the intensity of ALP activity increased remarkably by day 21. The intensity of von Kossa staining also peaked at day 21 (Figure 2A). RT-PCR was performed using osteogenic markers to confirm hMSC osteogenesis (Table 1). The mRNA expression levels of the osteogenic markers, which included bone sialoprotein (BSP), OCN and ALP, were significantly higher at day 21 than at day 0 (Figure 2B). Immunoblot analysis was performed using OCN and OPN in order to obtain further confirmation of osteogenesis. The results of von Kossa staining and RT-PCR were identical to the result observed with ALP and showed that the expression of the OCN and OPN proteins increased as differentiation progressed (Figure 2C). All of the corresponding results confirmed that the hMSCs were successfully differentiated into osteoblasts.

Multiplex PCR was used to assess the expression levels of HOX genes during osteogenic differentiation. The expression patterns of the 37 HOX genes were screened at day 0, day 10, and day 21 in both undifferentiated and differentiated hMSCs. The expression of the 37 HOX genes at the level of transcription is listed in Table 2. The HOXA1, HOXA4, HOXA6, HOXA9, HOXA11, HOXB9, HOXC5, HOXC6, HOXC12, HOXC13, HOXD4, and HOXD10 genes were up-regulated under the osteogenesis-induced condition. On the other hand, HOXB4 and HOXD13 were down-regulated during osteogenesis. The other HOX genes showed no significant changes in their mRNA expression levels.

Statistical analysis revealed that four HOX genes showed significant differences in expression at the transcription level. The HOXA1, HOXC11 and HOXC13 genes were found to be up-regulated. The expression of HOXC13 was unaltered between day 0 and day 10 and only increased after day 10. The expression of HOXA1 gradually increased for 21 days, but the increase in the expression of HOXC13 was more dramatic. The mRNA level of HOXC11 fluctuated during osteogenesis. The expression of HOXC11 increased during the first 10 days of osteogenic differentiation, but then decreased over the next 11 days (Figure 3A). The expression of HOXD13 was down-regulated during the osteogenesis of hMSCs. The mRNA level of HOXD13 decreased gradually over the 21-day period (Figure 3B).

The expression of HOXC13 and HOXD13 showed the most dramatic change after 21 days of differentiation. The expression of HOXC13 increased by approximately 91%, whereas that of HOXD13 decreased by 50% after osteogenesis. Real-time quantitative PCR and immunoblotting analysis were carried out in order to further confirm the increased expression of HOXC13 and HOXD13. The results of qPCR showed that the expression of HOXC13 was five times higher at day 10 and forty-two times higher at day 21 than in the undifferentiated state, respectively, whereas the mRNA expression of HOXD13 showed a five-fold decrease at day 10 (Figure 4). These qPCR results of the HOX genes were in agreement with those of multiplex PCR.

The expression levels of these two HOX genes were then submitted to immunoblot analysis to further evaluate their protein level in the osteogenic differentiation of hMSCs. The results showed increased expression of the HOXC13 protein and decreased expression of the HOXD13 protein after 21 days of differentiation (Figure 5). This result was in agreement with those of multiplex and real-time PCR.

Many factors are known to regulate osteogenesis (Bobis et al., 2006). The important factors involved in osteogenic regulation include bone morphogenetic protein (BMP), transforming growth factor (TGF), insulin-like growth factor (IGF), brain-derived growth factor (BDGF), fibroblast growth factor (FGF), leptin and parathyroid hormone-related peptide (PTHrP). These proteins regulate the expression of signals needed for bone remodeling. In addition, many reports have suggested that various transcription factors participate in osteogenesis. Among them, Cbfa1/ Runx2, Osterix, ΔFosB, Fra-1, Aj18, Osf1, Msx2, Dlx5 and TWIST have been shown to play pivotal roles.

Several studies have also reported that HOX genes are involved in osteogenesis. These reports showed that HOXA2 plays several important roles in the process of skeletogenesis (Gendron-Maguire et al., 1993; Rijli et al., 1993; Kanzler et al., 1998). Another study found, using quantitative RT-PCR (Dobreva et al., 2006), that the expression of HOXA2 was up-regulated during osteogenesis. HOXA10 has been shown to contribute to osteogenic lineage determination (Hassan et al., 2007). HOXC8 was reported to be involved in the regulation of osteogenesis through bone morphogenic protein (BMP) pathways (Juan et al., 2006). However, no significant changes in the expression of HOXA2, HOXA10 and HOXC8 were observed in the present study. The differences in these results may be due to the fact that HOXA2 may have been induced during mouse embryogenesis, and HOXA10 and HOXC8 expression were likely induced by BMP. However, in the present study, mesenchymal stem cells were used, and osteogenic differentiation was induced in vitro using dexamethasone, β-glycerophosphate and ascorbic acid.

Knowledge regarding the expression patterns of the HOX genes during osteogenic differentiation may reveal the signal pathway of osteogenesis and may also help in the potential therapeutic application of hMSCs. However, a report regarding the expression profile of HOX genes during osteogenesis has not yet been published. In the present report, 37 HOX genes were investigated in order to determine their expression patterns during the osteogenesis of hMSCs. For this purpose, we performed multiplex PCR, real-time PCR and Western blot analysis. Based on the results, we suggest that four HOX genes, HOXA1, HOXC13, HOXC11 and HOXD13, might be involved in the osteogenic differentiation of hMSCs. HOXA1 is a key gene in skull development, and it is a retinoic acid (RA) direct target gene (Ijichi and Ijichi, 2002). Mice with mutations in the HOXA1 hexapeptide motif show skeletal defects (Remacle et al., 2004). Similar results were reported by Martinez-Ceballos et al. (2005), who showed that the disruption of the HOXA1 gene results in abnormal ossification of the skull. Andrews et al. (1994) reported that osteogenic protein-1 (OP-1), a member of the TGF-β superfamily, induces HOXA1. In addition, recent microarray analyses revealed that BSP and Col1a1, both key markers of osteogenesis, are the target molecules of HOXA1 (Martinez-Ceballos et al., 2005). The results of multiplex PCR showed that HOXA1 was significantly increased during osteogenesis. The results of the present study and those of previous reports suggest that HOXA1 is an important factor involved in the osteogenesis of hMSCs.

In the present study, the expression of HOXC13 showed the largest increase. However, there are no previous reports suggesting a relationship between HOXC13 and osteogenesis. Kulessa et al. (2000) reported that the overexpression of the BMP inhibitor resulted in the down-regulation of HOXC13 expression in mutant mice. Based on the findings of the present study, it seems likely that HOXC13 contributes to the osteogenesis of hMSCs via the BMP pathway.

The HOXC11 gene encodes a transcription factor known to be involved in the definition of segment identities along the anterio-posterior axis. The expression of HOXC11 is detected in the mesenchyme posterior to the region forming the femur and fibula (Hostikka and Capecchi, 1998). There is a report suggesting that HOXC11 is involved in chondrogenesis, which is regulated by BMP2 and BMP7 (Papenbrock et al., 2000). However, there is no clear evidence that HOXC11 contributes to osteogenesis, thus HOXC11 may be related to the osteogenesis of hMSCs. In particular, HOXC11 may only be involved in the early stages of the osteogenic process from the hMSCs stage to the osteoblast progenitor cell stage, and not from the osteoblast progenitor cell stage to the osteoblast stage, once the expression level drops after day 10 (Figure 3A).

Williams et al. (2005) recently demonstrated that the interaction between the mouse HOXD13 protein and Smad1 might reciprocally antagonize the expression of Runx2, which is a key molecule in mammalian osteogenesis (Williams et al., 2005). This implies that the expression of HOXD13 may decrease as osteogenesis progresses, which is in agreement with the results of the present study. In light of previous reports on HOXD13 and of the present results, it is likely that the decrease in HOXD13 expression during osteogenesis is required for the promotion of osteogenic differentiation (Shi et al., 1999; Yang et al., 2000; Liu et al., 2004; Williams et al., 2005; Li et al., 2006).

There are few studies regarding the HOX genes involved in the differentiation of hMSCs. The results of the present study show that the mRNA expression levels of four HOX genes noticeably changed during the osteogenic differentiation of hMSCs. Although the roles of the four genes in the osteogenic differentiation of hMSCs have yet to be clarified, the present study represents a first step elucidating the relationship between HOX gene expression and the differentiation of hMSCs, making part of the signalling pathway in osteogenic differentiation from hMSCs. Functional studies, such as a gene siRNA-mediatd gene silencing or gene transfection, are needed in order to further investigate the role of the HOX genes in osteogenic differentiation.

Acknowledgments

This research was supported by a grant from the Seoul Research and Business Development Program (10548) funded by the Seoul Metropolitan Government, Republic of Korea.

References

Acampora D., D'Esposito M., Faiella A., Pannese M., Migliaccio E., Morelli F., Stornaiuolo A., Nigro V., Simeone A., Boncinelli E. (1989) The human HOX gene family. Nucleic Acids Res 17:10385-10402.

Akin Z.N., Nazarali A.J. (2005) Hox genes and their candidate downstream targets in the developing central nervous system. Cell Mol Neurobiol 25:697-741.

Andrews P.W., Damjanov I., Berends J., Kumpf S., Zappavigna V., Mavilio F., Sampath K. (1994) Inhibition of proliferation and induction of differentiation of pluripotent human embryonal carcinoma cells by osteogenic protein-1 (or bone morphogenetic protein-7). Lab Invest 71:243-251.

Bobis S., Jarocha D., Majka M. (2006) Mesenchymal stem cells: Characteristics and clinical applications. Folia Histochem Cytobiol 44:215-230.

Choi C.B., Cho Y.K., Prakash K.V., Jee B.K., Han C.W., Paik Y.K., Kim H.Y., Lee K.H., Chung N., Rha H.K. (2006) Analysis of neuron-like differentiation of human bone marrow mesenchymal stem cells. Biochem Biophys Res Commun 350:138-146.

Dobreva G., Chahrour M., Dautzenberg M., Chirivella L., Kanzler B., Farinas I., Karsenty G., Grosschedl R. (2006) SATB2 is a multifunctional determinant of craniofacial patterning and osteoblast differentiation. Cell 125:971-986.

Duboule D., Dolle P. (1989) The structural and functional organization of the murine HOX gene family resembles that of Drosophila homeotic genes. EMBO J 8:1497-1505.

Gehring W.J., Qian Y.Q., Billeter M., Furukubo-Tokunaga K., Schier A.F., Resendez-Perez D., Affolter M., Otting G., Wuthrich K. (1994) Homeodomain-DNA recognition. Cell 78:211-223.

Gendron-Maguire M., Mallo M., Zhang M., Gridley T. (1993) Hoxa-2 mutant mice exhibit homeotic transformation of skeletal elements derived from cranial neural crest. Cell 75:1317-1331.

Goff D.J., Tabin C.J. (1997) Analysis of Hoxd-13 and Hoxd-11 misexpression in chick limb buds reveals that Hox genes affect both bone condensation and growth. Development 124:627-636.

Hassan M.Q., Tare R., Lee S.H., Mandeville M., Weiner B., Montecino M., van Wijnen A.J., Stein J.L., Stein G.S., Lian J.B. (2007) HOXA10 controls osteoblastogenesis by directly activating bone regulatory and phenotypic genes. Mol Cell Biol 27:3337-3352.

Hostikka S.L., Capecchi M.R. (1998) The mouse Hoxc11 gene: Genomic structure and expression pattern. Mech Dev 70:133-145.

Ijichi S., Ijichi N. (2002) Minor form of trigonocephaly is an autistic skull shape. A suggestion based on homeobox gene variants and MECP2 mutations. Med Hypotheses 58:337-339.

Jee B.K., Lee J.Y., Lim Y., Lee K.H., Jo Y.H. (2007) Effect of KAI1/CD82 on the beta1 integrin maturation in highly migratory carcinoma cells. Biochem Biophys Res Commun 359:703-708.

Jee B.K., Park K.M., Surendran S., Lee W.K., Han C.W., Kim Y.S., Lim Y. (2006) KAI1/CD82 suppresses tumor invasion by MMP9 inactivation via TIMP1 up-regulation in the H1299 human lung carcinoma cell line. Biochem Biophys Res Commun 342:655-661.

Juan A.H., Lei H., Bhargava P., Lebrun M., Ruddle F.H. (2006) Multiple roles of hoxc8 in skeletal development. Ann N Y Acad Sci 1068:87-94.

Kanzler B., Kuschert S.J., Liu Y.H., Mallo M. (1998) Hoxa-2 restricts the chondrogenic domain and inhibits bone formation during development of the branchial area. Development 125:2587-2597.

Kraus K.H., Kirker-Head C. (2006) Mesenchymal stem cells and bone regeneration. Vet Surg 35:232-242.

Kulessa H., Turk G., Hogan B.L. (2000) Inhibition of Bmp signaling affects growth and differentiation in the anagen hair follicle. EMBO J 19:6664-6674.

Levine M., Rubin G.M., Tjian R. (1984) Human DNA sequences homologous to a protein coding region conserved between homeotic genes of Drosophila. Cell 38:667-673.

Li X., Nie S., Chang C., Qiu T., Cao X. (2006) Smads oppose Hox transcriptional activities. Exp Cell Res 312:854-864.

Liu Z., Shi W., Ji X., Sun C., Jee W.S., Wu Y., Mao Z., Nagy T.R., Li Q., Cao X. (2004) Molecules mimicking Smad1 interacting with Hox stimulate bone formation. J Biol Chem 279:11313-11319.

Martinez-Ceballos E., Chambon P., Gudas L.J. (2005) Differences in gene expression between wild type and Hoxa1 knockout embryonic stem cells after retinoic acid treatment or leukemia inhibitory factor (LIF) removal. J Biol Chem 280:16484-16498.

Papenbrock T., Visconti R.P., Awgulewitsch A. (2000) Loss of fibula in mice overexpressing Hoxc11. Mech Dev 92:113-123.

Pittenger M.F., Mackay A.M., Beck S.C., Jaiswal R.K., Douglas R., Mosca J.D., Moorman M.A., Simonetti D.W., Craig S., Marshak D.R. (1999) Multilineage potential of adult human mesenchymal stem cells. Science 284:143-147.

Remacle S., Abbas L., De Backer O., Pacico N., Gavalas A., Gofflot F., Picard J.J., Rezsohazy R. (2004) Loss of function but no gain of function caused by amino acid substitutions in the hexapeptide of Hoxa1 in vivo. Mol Cell Biol 24:8567-8575.

Rijli F.M., Mark M., Lakkaraju S., Dierich A., Dolle P., Chambon P. (1993) A homeotic transformation is generated in the rostral branchial region of the head by disruption of Hoxa-2, which acts as a selector gene. Cell 75:1333-1349.

Shi X., Yang X., Chen D., Chang Z., Cao X. (1999) Smad1 interacts with homeobox DNA-binding proteins in bone morphogenetic protein signaling. J Biol Chem 274:13711-13717.

Song L., Webb N.E., Song Y., Tuan R.S. (2006) Identification and functional analysis of candidate genes regulating mesenchymal stem cell self-renewal and multipotency. Stem Cells 24:1707-1718.

van den Akker E., Fromental-Ramain C., de Graaff W., Le Mouellic H., Brulet P., Chambon P., Deschamps J. (2001) Axial skeletal patterning in mice lacking all paralogous group 8 Hox genes. Development 128:1911-1921.

Wang G., Brennan C., Rook M., Wolfe J.L., Leo C., Chin L., Pan H., Liu W.H., Price B., Makrigiorgos G.M. (2004) Balanced-PCR amplification allows unbiased identification of genomic copy changes in minute cell and tissue samples. Nucleic Acids Res 32:e76

Williams T.M., Williams M.E., Heaton J.H., Gelehrter T.D., Innis J.W. (2005) Group 13 HOX proteins interact with the MH2 domain of R-Smads and modulate Smad transcriptional activation functions independent of HOX DNA-binding capability. Nucleic Acids Res 33:4475-4484.

Yang X., Ji X., Shi X., Cao X. (2000) Smad1 domains interacting with Hoxc-8 induce osteoblast differentiation. J Biol Chem 275:1065-1072.

Table 1
- Primer sequences.

Table 2
- HOX multiplex PCR. A total of thirty-seven HOX genes were examined using multiplex RT-PCR. The relative expression values are expressed as mean ± SEM. P values were calculated using ANOVA.

Received: October 26, 2007; Accepted: January 22, 2008

Gue-Tae Chae and Kweon-Haeng Lee. Institute of Hansen's Disease, The Catholic University of Korea, 505, Banpo-dong, Seocho-ku, Seoul, Republic of Korea. E-mail: ngrc@catholic.ac.kr.

  • Introduction
  • Materials and Methods
  • Research protocol
  • Flow cytometric analysis (FACS) of hMSCs
  • Osteogenic differentiation
  • Alkaline phosphatase (ALP) staining
  • von Kossa staining
  • Immunoblotting analysis
  • Multiplex PCR of HOX genes
  • Real-time quantitative PCR analysis (qPCR)
  • Statistical analysis
  • Results
  • Characterization of hMSCs
  • Osteogenic differentiation
  • Analysis of HOX gene expression using multiplex PCR
  • Expression of HOXC13 and HOXD13
  • Discussion
  • Tables
  • p
  • p
  • Assistant Editor: Klaus Hartfelder
  • *
    These authores contributed equally to this work.
  • Publication Dates

    • Publication in this collection
      19 Nov 2008
    • Date of issue
      2008

    History

    • Accepted
      2201
    • Received
      2610
    Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
    E-mail: editor@gmb.org.br