Skip to main content

Long noncoding RNA Braveheart promotes cardiogenic differentiation of mesenchymal stem cells in vitro

Abstract

Background

Mesenchymal stem cells (MSCs) have limited potential of cardiogenic differentiation. In this study, we investigated the influence of long noncoding RNA Braveheart (lncRNA-Bvht) on cardiogenic differentiation of MSCs in vitro.

Methods

MSCs were obtained from C57BL/6 mice and cultured in vitro. Cells were divided into three groups: blank control, null vector control, and lncRNA-Bvht. All three groups experienced exposure to hypoxia (1% O2) and serum deprivation for 24 h, and 24 h of reoxygenation (20% O2). Cardiogenic differentiation was induced using 5-AZA for another 24 h. Normoxia (20% O2) was applied as a negative control during the whole process. Cardiogenic differentiation was assessed, and expressions of cardiac-specific transcription factors and epithelial-mesenchymal transition (EMT)-associated biomarkers were detected. Anti-mesoderm posterior1 (Mesp1) siRNA was transfected in order to block its expression, and relevant downstream molecules were examined.

Results

Compared with the blank control and null vector control groups, the lncRNA-Bvht group presented a higher percentage of differentiated cells of the cardiogenic phenotype in vitro both under the normal condition and after hypoxia/re-oxygenation. There was an increased level of cTnT and α-SA, and cardiac-specific transcription factors including Nkx2.5, Gata4, Gata6, and Isl-1 were significantly upregulated (P < 0.01). Expressions of EMT-associated genes including Snail, Twist and N-cadherin were much higher (P < 0.01). Mesp1 exhibited a distinct augmentation following lncRNA-Bvht transfection. Expressions of relevant cardiac-specific transcription factors and EMT-associated genes all presented a converse alteration in the condition of Mesp1 inhibition prior to lncRNA-Bvht transfection.

Conclusion

lncRNA-Bvht could efficiently promote MSCs transdifferentation into cells with the cardiogenic phenotype in vitro. It might function via enhancing the expressions of cardiac-specific transcription factors and EMT-associated genes. Mesp1 could be a pivotal intermediary in the procedure.

Background

Cardiovascular disease remains a major cause of morbidity and mortality worldwide [1]. The current treatment options for end-stage heart failure fail to regenerate myocardium that has gone through necrosis or apoptosis. Induction of cardiac regeneration to replace the lost cardiomyocytes in the injured heart represents a promising therapeutic approach in this context [2]. Stem cell therapy has emerged as a novel strategy for the treatment of ischemic heart disease during the past decade. Various stem cell types have been used for the repair of the damaged heart [2–4]. Noteworthy benefits are revealed in the regeneration of cardiomocytes following the transplantation of the precursor cells [2–4]. However, the underlying molecular mechanisms that lead to cardiomyocyte regeneration after cell therapy have not been fully elucidated.

Bone marrow-derived mesenchymal stem cells (BMMSCs) have a great potential of proliferation and differentiation, and they have been considered as a suitable source for cell therapy [5, 6]. Mesenchymal stem cells (MSCs) are capable of differentiating into cardiomyocytes under appropriate conditions both in vitro and in vivo [6]. In spite of this, the transdifferentiation efficiency of these cells is extremely low. Currently, several measures have been developed to promote the differentiation of MSCs into cardiomyocytes [7, 8]. However, most of these methods are inefficient and only a small percentage of differentiated cells can be produced. How to gain a high rate of cardiogenic differentiation from MSCs has become an issue that needs to be addressed.

Stem cell transdifferentiation into cardiomyocytes fundamentally relies on elaborate cellular and molecular mechanisms [9]. Recent discoveries demonstrate that the non-coding portion of the genome plays a crucial role in controlling cellular fate, phenotype and behavior [10]. A large number of noncoding RNAs (ncRNAs) that function as central orchestrators of cell-specific gene networks have been identified [10, 11]. An important subclass of these ncRNAs is the long noncoding RNAs (lncRNAs) that are broadly defined as regulatory noncoding transcripts more than 200 nucleotides in length. Although their biological roles and mechanisms of function remain largely elusive, accumulating evidence shows that lncRNAs participate in a wide spectrum of biological processes including cellular development, disease etiology, stem cell pluripotency and lineage specification [12]. There are already a handful reports indicating that lncRNAs can modulate cardiac differentiation during heart development [13, 14]. The long noncoding RNA Braveheart (lncRNA-Bvht) is a heart-associated lncRNA that has been identified as a pivotal regulator of cardiac lineage specification and differentiation [14]. It mediates cardiac commitment epigenetically and performs critical roles during cardiac differentiation in mouse embryonic stem cells (ESCs).

Epithelial-mesenchymal transition (EMT) is a biological process that is implicated in the developmental stage, organogenesis, tissue repair and pathological conditions [15]. Emerging evidence indicates that EMT might result in transformation of stem cell phenotypes. EMT accompanies transitions between stem-like cells and their more differentiated progeny, which perform critical functions in tissue repair and regeneration [16]. It has been revealed that EMT is involved in cardiac differentiation of ESCs and pluripotent stem cells (PSCs) [17, 18].

Mesoderm posterior 1 (Mesp1) is an essential transcription factor that marks a common multipotent cardiovascular progenitor [14]. Its expression can induce cardiovascular progenitor cells [19]. lncRNA-Bvht functions via Mesp1 to modulate the expression of cardiac transcription factors and further promote cardiogenic differentiation of ESCs [14]. Previous data show that Mesp1 is capable of initiating the EMT process by regulating EMT-associated genes [20].

In this study, lncRNA-Bvht was transfected into MSCs of C57BL/6 mice in order to investigate its implication on cardiogenic differentiation of these cells, and the underlying mechanism involved were explored in the procedure.

Methods

Ethics statement

Three-week-old C57BL/6 mice were obtained from the Animal Experimental Center of the Sun Yat-sen University. All animal handling and procedures were performed in accordance with protocols approved by the Animal Ethics Committee of Sun Yat-sen University (201210016).

Isolation and culture of bone marrow-derived mesenchymal stem cells

All experiment protocols described were approved by the Institutional Animal Care & Use Committee (IACUC) at Sun Yat-sen University. Bone marrow cells were collected from 3 to 4 weeks old C57BL/6 mice by flushing femurs and tibias under aseptic conditions. Cells were cultured (37 °C, 5% CO2) in 25 cm2 culture flasks with complete culture medium supplemented with 10% fetal bovine serum, l-glutamine (4.0 mM), penicillin (100 IU/mL) and streptomycin (100 μg/mL). On the third day of culture, the medium was replaced and non-adherent cells were removed. The adherent cells were washed two times gently with phosphate-buffered saline (PBS) to reduce the degree of hematopoietic lineage cell contamination. The cells were cultured in complete culture medium and the medium was changed every 3 to 4 days for 3–4 weeks. Adherent cells gaining 90% confluence were trypsinized with 0.25% trypsin–ethylenediamine tetraacetic acid (Invitrogen) and passaged into new flasks for further expansion. Characteristics of MSCs were identified by fluorescence-activated cell sorting as previously reported [21].

lncRNA-Bvht vector construction

The pre-lncRNA-Bvht oligonucleotides were chemically synthesized by Jinweizhi Co. Ltd. (Jiangsu, China). The primers were as follows: XhoI forward: 5′ccgCTCGAGGATCTCTGCCCCTCAGAGTCC3′, BamHI reverse: 5′cgcGGATCCAACATTTATTTTTAAAGTTTA 3′. The recovered polymerase chain reaction (PCR) products with the precursor sequence for lncRNA-Bvht were inserted into pLVX-IRES-ZsGreen1 vector. After the pre-lncRNA-Bvht viral-based vector was transformed to DH5α cells, antibiotic-resistant colonies were selected on LB-ampicillin (100 μg/mL) agar plates. The plasmid containing the target gene was verified by PCR, double digestion and DNA sequencing.

lncRNA-Bvht transfection

The monolayer of MSCs of uniform growth attaining 90% confluence were passaged. Culture medium was removed and cells were trypsinized with 0.25% trypsin–ethylenediamine tetraacetic acid (Invitrogen). The cells were re-seeded at a density of 1 × 106 cells per cell culture flask with complete medium for 24 h. Cells gaining 70–80% confluence were applied for transfection. The pLVX-IRES-ZsGreen1 vector encoding lncRNA-Bvht was transfected into MSCs with lipofectamine 2000 (Invitrogen) according to the manufacturer’s instructions. The medium was changed with fresh complete DMEM 8 h after transfection. The expression of ZsGreen was checked after 48 h of transfection.

siRNAs experiments

MSCs were incubated at 1 × 106 cells per well in six-well plates at day 0 with siRNAs against Mesp1 (Sigma) or control siRNAs (negative control, NC; Sigma). Transfection of siRNAs was performed using lipofectamine 2000 (Invitrogen) according to the manufacturer’s instructions. Mesp1 knockdown was determined by quantitative real-time PCR.

Hypoxia/reoxygenation treatment of MSCs

MSCs in the blank control, null vector control and lncRNA-Bvht groups all experienced hypoxia/reoxygenation treatment. Cells in the different groups were incubated in serum-free media with 1% O2 in a Galaxy® 48 R incubator (Eppendorf/Galaxy Corporation, USA) at 37 °C for 24 h and exposed to normoxic condition (20% O2) for another 24 h. Normoxia was used as a negative control during the experiments for the three groups.

Cardiogenic differentiation of MSCs

Differentiation of MSCs to cardiogenic cells was accomplished afterwards. MSCs of the three groups were seeded into six-well plates at a concentration of 1 × 106 cells per well. To induce cell differentiation, the cells were incubated in a medium containing 5-AZA (10uM; Sigma–Aldrich) for 24 h at 37 °C in a humidified atmosphere with 5% CO2. The cells were then washed twice and the medium was replaced with normal DMEM. The medium was changed every 3 days and this procedure was terminated at 2 weeks. The morphological changes in MSCs were observed under a microscope (Olympus, CX41).

Immunofluorescence staining

Slides with the treated cell samples taken from dishes were used directly. After drying at room temperature for a few minutes, they were permeabilized in 2% formaldehyde/PBS for 10 min. Antigen retrieval was followed by microwaving sections in sodium citrate buffer (1 M, pH 6.1). Sections were blocked with 5% bovine serum albumin (BSA) at room temperature before incubating with primary antibodies at 4 °C overnight (dilution cTnT, 1:100; α-SA, 1:100). After washing, sections were incubated with appropriate secondary antibodies and slides were counterstained with 4-6-diamidino-2-phenylindole (DAPI). Images were taken by fluorescent microscopy (Leica, Germany) with a CCD camera (Tokyo, Japan). The percentage of cTnT-positive cells was used to evaluate the efficiency of MSCs transdifferentiated into cells with the cardiogenic phenotype.

Western blot analysis

Protein levels were measured by western blot. Cells were washed several times with PBS before collection and lysed with modified RIPA buffer. Cells were completely lysed after repeated vortexing, and supernatants were acquired though centrifugation at 14,000 × g for 20 min. Proteins were resolved by sodium dodecyl sulfatepolyacrylamide gel (SDS-PAGE) and transferred to a polyvinylidenedifluoride (PVDF) membrane (IPVH00010, Millpore, Boston, USA) before incubation with the primary antibodies overnight at 4 °C. The membranes were subjected to three 5-min washes with TBST and incubated with anti-IgG horseradish peroxidase–conjugated secondary antibody (Southern biotech, Birmingham, USA) for 60 min at room temperature. After extensive washing, bands were detected by enhanced chemiluminescence. The band intensities were quantified by using image software (image J 2×, version 2.1.4.7).

Quantitative real-time PCR

Total RNA was isolated from cells using a Trizol reagent (Invitrogen) followed by digestion with RNase-free DNase (Promega). Concentration and integrity of total RNA were estimated and the real-time PCR was conducted on an ABI PRISM® 7500 Sequence Detection System using SYBR Green qPCR SuperMix (Invitrogen). The primers are described in Table 1. Specific products were amplified and detected at 95 °C for 10 min, followed by 40 cycles at 95 °C for 15 s and at 60 °C for 30 s, at which point data were acquired. The relative level of mRNA was calculated using the 2−ΔΔCt method. For the assays of the molecules examined, the results were quantified as the threshold cycle of each target gene and normalized into ΔCt value. Quantifications of fold-change in gene expressions were also performed using the 2−ΔΔCt method.

Table 1 List of primers for quantitative real-time polymerase chain reaction

Statistical analysis

All quantitative data are described as mean ± SD. The significance of differences among groups was determined by the analysis of variance and Scheffe’s multiple-comparison techniques. Comparisons between time-based measurements within each group were performed with analysis of variance for repeated measurements. A P value <0.05 was considered to be statistically significant.

Results

PCR amplification and sequencing of lncRNA-Bvht

DNA fragments of lncRNA-Bvht were successfully amplified by PCR. Electrophoresis revealed the specific band of lncRNA-Bvht at 500 bp (Fig. 1A). The sequence of lncRNA-Bvht was analyzed (Fig. 1B).

Fig. 1
figure 1

Electrophoresis of PCR products and sequence analysis of lncRNA-Bvht. A showed the specific band of lncRNA-Bvht at 500 bp by electrophoresis; (B) showed that the lncRNA-Bvht sequence was correctly constructed

lncRNA-Bvht transfection efficiency

ZsGreen was expressed after MSCs were transduced with the pLVX-IRES-ZsGreen1 vector. All the MSCs with ZsGreen expression were observed under the microscope (Fig. 2A). After lncRNA-Bvht tranfection, its expression in different cell groups was detected by quantitative real-time PCR. The mRNA level was significantly higher in the lncRNA-Bvht group compared with the blank control and null vector control groups (Fig. 2B; P < 0.01).

Fig. 2
figure 2

Detection of lncRNA-Bvht transfection efficiency. lncRNA-Bvht transfection efficiency was detected by the expression of ZsGreen and mRNA level of lncRNA-Bvht. A MSCs expressing ZsGreen after lncRNA-Bvht transfection were shown by fluorescent microscopy (×400); a represented MSCs transfected with lncRNA-Bvht and b showed that all the cells with ZsGreen expression were obtained. B The expression of lncRNA-Bvht in different cell groups was detected by quantitative real-time PCR

Cardiogenic differentiation in different cell groups

Cardiogenic differentiation in different cell groups was examined by immunofluorescence staining (Fig. 3). Morphology changes could be observed in different cells groups after 14 days of induction. The differentiated MSCs expressed cardiomyocyte-specific cell markers including cTnT (green colour; Fig. 3A1–A6, images b) and α-SA (red colour; Fig. 3A1–A6, images c). The lncRNA-Bvht group (Fig. 3A3 and A6) showed an obviously higher percentage of cTnT-positive cells than the blank control (Fig. 3A1 and A4) and null vector control groups (Fig. 3A2 and A5) both under the normal condition and after the hypoxia/reoxygenation treatment (P < 0.01; Fig. 3A1–A6 and B); mRNA and protein levels of cTnT and α-SA were also elevated after lncRNA-Bvht transfection (Fig. 3C).

Fig. 3
figure 3

Cardiogenic differentiation in the different cell groups. Cardiogenic differentiation of MSCs in the different cell groups was evaluated by the expressions of cTnT and α-SA. A Confocal microscopy of immunofluorescent staining of DAPI-labeled MSCs induced by 5-AZA after 14 days (200×). Cells stained with antibody to cTnT appeared green, and cells stained with antibody to α-SA appeared red. A1–A3 represented the expressions of cTnT and α-SA under normoxic condition, and A4–A6 represented the expressions of cTnT and α-SA under the hypoxia/reoxygenation (HR) condition; A1 and A4, A2 and A5, and A3 and A6 represented the blank control group, null vector control group, and lncRNA-Bvht group respectively. a Cells derived from DAPI-labeled MSCs induced by 5-AZA displayed blue nuclei; (b) Cells stained with antibody to cTnT appeared green in A1-A6; (c) Cells stained with antibody to α-SA appeared red in A1-A6; (d) Merged image of a, b, and c. B Comparison of the percentage of cTnT-positive cells among the different groups under normoxic condition and after hypoxia/reoxygenation respectively. C Western blot (a) and quantitative real-time PCR (b) analysis of the expressions of cTnT and α-SA in the different cell groups. **P <0.01 , versus blank control; ## P <0.01 , versus null vector control

Expressions of cardiac-specific transcription factors and EMT-associated genes in different cell groups after the induction of MSCs differentiation

The expressions of cardiac-specific transcription factors including Mesp1, Nkx2.5, Gata4, Gata6, and Isl1 were examined at different time points after the induction of cardiogenic differentiation. They all showed remarkably higher expressions in the lncRNA-Bvht group than the blank control and null vector control groups both under the normal condition and after hypoxia/reoxygenation (P < 0.01; Fig. 4). Expression levels of EMT-associated genes including Snail, Twist and N-cadherin were also upregulated in lncRNA-Bvht group compared with the other two groups (P < 0.01; Fig. 5).

Fig. 4
figure 4

Expressions of cardiac-specific transcription factors in different cell groups after the induction of cardiogenic differentiation. A Expressions of cardiac-specific transcription factors in different cell groups under normoxic condition. B Expressions of cardiac-specific transcription factors in different cell groups after hypoxia/reoxygenation (HR). * P<0.05, ** P<0.01, vesus blank control; # P<0.05, ## P<0.01, vesus null vector control

Fig. 5
figure 5

Expressions of EMT-associated genes in different cell groups after the induction of cardiogenic differentiation. A Expressions of EMT-associated genes in different cell groups under normoxic condition. B Expressions of EMT-associated genes in different cell groups after hypoxia/reoxygenation (HR). * P<0.05,** P<0.01, vesus blank control; # P<0.05,## P<0.01, vesus null vector control

Inhibition of Mesp1 interfered with MSCs transdifferentiation into cells with the cardiogenic phenotype induced by lncRNA-Bvht

Anti-Mesp1 siRNAs and control siRNAs (NC) were transiently transfected into undifferentiated MSCs before lncRNA-Bvht transfection and further induction of cardiogenic differentiation. Expressions of Mesp1 and relevant downstream molecules were analyzed 72 h later. A significant reduction in Mesp1 expression was observed in the anti-Mesp1 siRNA group. Expressions of cardiac differentiation-associated genes including Nkx2.5, Gata4, Gata6, and Isl1 were all decreased, and EMT-associated genes including Snail, Twist and N-cadherin were downregulated under the condition of Mesp1 inhibition in the lncRNA-Bvht transfection group both under normoxia and after hypoxia/reoxygenation (P < 0.01; Fig. 6).

Fig. 6
figure 6

Expressions of cardiac-specific transcription factors and EMT-associated genes in different cell groups after the inhibition of Mesp1. Western blot (A) and quantitative real-time PCR (B) analysis of cardiac-specific transcription factors and EMT-associated genes in different cell groups after the inhibition of Mesp1. HR hypoxia/reoxygenation. *P<0.05,**P<0.01, vesus blank control; #P<0.05, ##P<0.01, vesus null vector control; ☨P<0.05, ☨☨P<0.01, vesus lncRNA-Bvht

Discussion

This study demonstrated that lncRNA-Bvht tranfection could efficiently promote MSCs transdifferentiation into cells with the cardiogenic phenotype in vitro. MSCs-derived cells expressed cardiac-specific markers including cTnT and α-SA. Cardiac-specific transcription factors and EMT-associated genes were upregulated following lncRNA-Bvht transfection both under normal condition and after hypoxia/reoxygenation. However, the expressions of these molecules all presented a converse alteration under the condition of Mesp1 inhibition prior to lncRNA-Bvht transfection.

To derive cardiomyocytes from stem cell precursors has been adopted as a pivotal therapeutic strategy for the repair of the injured heart. MSCs provide a valuable platform for the treatment of heart disease based on regenerative medicine [22]. Nevertheless, they show limited cardiomyogenic potential in spite of functional benefits resulting from their transplantation. Arduous efforts have been made to escalate the efficiency of cardiogenic differentiation of these cells. However, the efficacy of cellular cardiomyoplasty with MSCs remains frustrating, raising the need for alternative induction methods.

lncRNAs have been shown to be implicated in the modulation of stem cell pluripotency and cardiac differentiation [23]. It is revealed that lncRNAs are integral components of stem cell transcriptional networks [24, 25]. The knockdown or overexpression of relevant lncRNAs reciprocally influences the pluripotent transcription factors, dominating stem cell pluripotent state and lineage specificity [26–28]. Other studies have exhibited that lncRNAs regulate the cellular reprogramming process and play a pivotal role during the reprogramming of somatic cells [29, 30]. lncRNAs performing as competitive endogenous RNAs (ceRNAs) have been found to be differentially expressed in differentiating human cardiac progenitor cells (CPCs). These ceRNAs exert regulatory roles in cardiac lineage specification and differentiation [31].

Much attention has been drawn to the role of lncRNAs in heart development and cardiac differentiation [32]. Several lncRNAs have been uncovered as critical players in the development of the early cardiovascular system and cardiac differentiation [13, 14]. Some enhancer-associated lncRNAs have also been reported to take control of cardiac specification, differentiation and homeostasis [33]. lncRNA-Bvht is a newly discovered cardiac-specific lncRNA in the mouse. It promotes cardiogenic differentiation of ESCs and retains the cardiac phenotype in neonatal cardiomyocytes [14]. In this study, lncRNA-Bvht was transfected into MSCs in order to investigate its effect on cardiogenic differentiation of these cells. We discovered that a larger proportion of cells with the cardiogenic phenotype were induced after lncRNA-Bvht transfection. lncRNA-Bvht transfected MSCs displayed evenly distributed and regularly organized myofibrils after 14 days of stimuli in culture. The differentiated cells were shown to have a mature cardiogenic phenotype as evidenced by a much higher expression of cTnT. There was an enhanced level of cardiac-specific transcription factors incuding Nkx2.5, Gata4, Gata6, and Isl-1, suggesting that lncRNA-Bvht could efficiently induce the transdifferentiation of MSCs into cells with the cardiogenic phenotype in vitro by promoting the expressions of cardiac differentiation-associated genes.

Studies have demonstrated that EMT exerts a crucial role during heart development and cardiac repair. It facilitates CPCs formation during embryonic development and leads to the generation of CPCs in the adult heart [34]. EMT positively regulates cardiac differentiation of ESCs and PSCs [17, 18]. Further investigations into the EMT-based regenerative program might contribute to a more efficient strategy for cardiomyogenesis of stem cells and cardiac repair. In this study, genes implicated in EMT such as Snail and Twist were upregulated during lncRNA-Bvht induced transdifferentiation of MSCs. Twist is a crucial transcription factor that participates in the regulation of multi-progenitor states and lineage specificity of MSCs [35, 36]. It has recently been reported that Twist can modulate MSCs lineage commitment by effecting MSCs fate switching between osteogenesis and adipogenesis [36]. In this study, Twist was upregulated after lncRNA-Bvht transfaection, indicating that it might facilitate the cardiac lineage differentiation of MSCs. N-cadherin is identified as another indicator of EMT. The EMT process accompanies an elevated expression of N-cadherin. It has been revealed that N-cadherin could be a novel prospective cell surface marker of MSCs that present an apparently higher ability for cardiomyocyte differentiation [37]. N-cadherin has also been shown to be the only cell-surface gene that is highly expressed in the optimal human umbilical cord blood-derived MSCs (hUCB-MSCs). hUCB-MSCs with a notable expression of N-cadherin exhibit an enhanced cell survival rate and interact with host cardiomyocytes more effectively [38]. In this study, we also found that N-cadherin expression level was significantly enhanced after lncRNA-Bvht transfection. The aforementioned data suggest that EMT-associated genes were involved in lncRNA-Bvht mediated cardiogenic differentiation of MSCs.

Mesp1 is a basic helix-loop-helix transcription factor that symbolizes the earliest progenitors of the cardiovascular system [19]. Mesp1 positive cells have been reported to show an increased expression of mesodermal and EMT markers and preferential cardiac differentiation, implying a critical role of Mesp1 in promoting EMT and cardiac differentiation [20]. Evidence shows that Mesp1 regulates EMT and cardiovascular differentiation in ESCs and CPCs [20, 39]. Several pharmacological agents have been introduced to enhance the cardiomyogenic ability of MSCs for cardiac repair [40]. MSCs treated with these agents show an appreciably elevated expression of Mesp1, with a later widespread upregulation of the structural genes of cardiomyocytes. Other data display that a recombinant method using various growth factors and cell cytokines guides MSCs into a cardiac progenitor phenotype with a significantly increased expression of Mesp1, conferring cardiac competency on these cells to regenerate new cardiomyocytes [41]. In this study, Mesp1 was remarkably increased at different time points after the induction of cardiogenic differentiation both under the normal condition and after hypoxia/reoxygenation following the lncRNA-Bvht transfection. There was a distinct upregualtion of cardiac differentiation-associated genes and biomarkers of the EMT process. Mesp1 repression before lncRNA-Bvht transfection significantly downregulated the expressions of cardiac differentiation-associated genes and biomarkers of EMT, indicating that lncRNA-Bvht mediated upregulation of Mesp1 might contribute to the expression of cardiac differentiation-associated genes and biomarkers of the EMT process, which subsequently leads to the transdifferentiation of MSCs into cells with the cardiogenic phenotype.

Limitations of the study

This study was performed in vitro, in vivo studies will be conducted in the future to investigate the effects of lncRNA-Bvht on cardiomyocyte differentiation of MSCs in vivo. In this work, we mainly demonstrated that lncRNA-Bvht could induce the transdifferentiation of MSCs into cells with cardiogenenic phenotypes. Further studies will be focused on functional properties of these differentiated cells, including their contraction and electrophysiological characteristics and whether they possess the ability to form functional syncytium with host cardiomyocytes. Specific signaling pathways involved in the functional mechanisms of lncRNA-Bvht will also be explored later on.

Conclusions

lncRNA-Bvht could efficiently promote MSCs transdifferentiation into cells with the cardiogenic phenotype in vitro. It might function via enhancing the expressions of cardiac-specific transcription factors and EMT-associated genes. Mesp1 could be a crucial intermediary in the procedure. This study provides new insight for the application of lncRNAs in stem cell-mediated cardiomyocyte regeneration. Further work on the functional mechanisms of these novel molecules might be conducive for improving the therapeutic efficiency of MSCs transplantation.

Abbreviations

ceRNA:

Competitive endogenous RNA

CPC:

Cardiac progenitor cell

EMT:

Epithelial-mesenchymal transition

ESC:

Embryonic stem cell

hUCB-MSC:

Human umbilical cord blood-derived mesenchymal stem cell

lncRNA-Bvht:

long noncoding RNA Braveheart

lncRNA:

long noncoding RNA

Mesp1:

Mesoderm posterior1

MSC:

Mesenchymal stem cell

ncRNA:

noncoding RNA

PBS:

Phosphate-buffered saline

PSC:

Pluripotent stem cell

References

  1. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, et al. Heart disease and stroke statistics—2015 update: a report from the American Heart Association. Circulation. 2015;131(4):e29–322.

    Article  PubMed  Google Scholar 

  2. Hou J, Wang L, Jiang J, Zhou C, Guo T, Zheng S, et al. Cardiac stem cells and their roles in myocardial infarction. Stem Cell Rev. 2013;9(3):326–38.

    Article  CAS  PubMed  Google Scholar 

  3. Fox IJ, Daley GQ, Goldman SA, Huard J, Kamp TJ, Trucco M. Stem cell therapy. Use of differentiated pluripotent stem cells as replacement therapy for treating disease. Science. 2014;345:1247391.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Garbern JC, Lee RT. Cardiac stem cell therapy and the promise of heart regeneration. Cell Stem Cell. 2013;12:689–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hou J, Wang L, Hou J, Guo T, Xing Y, Zheng S, et al. Peroxisome proliferator-activated receptor gamma promotes mesenchymal stem cells to express connexin43 via the inhibition of TGF-β1/Smads signaling in a rat model of myocardial infarction. Stem Cell Rev. 2015;11(6):885–99.

    Article  CAS  PubMed  Google Scholar 

  6. Piryaei A, Soleimani M, Heidari MH, Saheli M, Rohani R, Almasieh M. Ultrastructural maturation of human bone marrow mesenchymal stem cells-derived cardiomyocytes under alternative induction of 5-azacytidine. Cell Biol Int. 2015;39(5):519–30.

    Article  CAS  PubMed  Google Scholar 

  7. Xing Y, Lv A, Wang L, Yan X. The combination of angiotensin II and 5-azacytidine promotes cardiomyocyte differentiation of rat bone marrow mesenchymal stem cells. Mol Cell Biochem. 2012;360(1–2):279–87.

    Article  CAS  PubMed  Google Scholar 

  8. Perán M, López-Ruiz E, González-Herrera L, Bustamante M, Valenzuela A, Marchal JA. Cellular extracts from post-mortem human cardiac tissue direct cardiomyogenic differentiation of human adipose tissue-derived stem cells. Cytotherapy. 2013;15(12):1541–8.

    Article  PubMed  Google Scholar 

  9. Wamstad JA, Wang X, Demuren OO, Boyer LA. Distal enhancers: new insights into heart development and disease. Trends Cell Biol. 2014;24:294–302.

    Article  CAS  PubMed  Google Scholar 

  10. Lam MT, Li W, Rosenfeld MG, Glass CK. Enhancer RNAs and regulated transcriptional programs. Trends Biochem Sci. 2014;39:170–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Beilharz TH. Understanding the regulation of coding and noncoding transcription in cell populations. Curr Genet. 2016;62(2):317–9.

    Article  CAS  PubMed  Google Scholar 

  12. Dey BK, Mueller AC, Dutta A. Long non-coding RNAs as emerging regulators of differentiation, development, and disease. Transcription. 2014;5(4), e944014.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Grote P, Wittler L, Hendrix D, Koch F, Währisch S, Beisaw A, et al. The tissue-specific lncRNA Fendrr is an essential regulator of heart and body wall development in the mouse. Dev Cell. 2013;24:206–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Klattenhoff CA, Scheuermann JC, Surface LE, Bradley RK, Fields PA, Steinhauser ML, et al. Braveheart, a long noncoding RNA required for cardiovascular lineage commitment. Cell. 2013;152:570–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Pasquier J, Abu-Kaoud NA, Thani H, Rafii A. Epithelial to mesenchymal transition in a clinical perspective. J Oncol. 2015;2015:792182.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Asli NS, Harvey RP. Epithelial to mesenchymal transition as a portal to stem cell characters embedded in gene networks. Bioessays. 2013;35(3):191–200.

    Article  CAS  PubMed  Google Scholar 

  17. Richter A, Valdimarsdottir L, Hrafnkelsdottir HE, Runarsson JF, Omarsdottir AR, Ward-van Oostwaard D, et al. BMP4 promotes EMT and mesodermal commitment in human embryonic stem cells via SLUG and MSX2. Stem Cells. 2014;32(3):636–48.

    Article  CAS  PubMed  Google Scholar 

  18. Zhang J, Klos M, Wilson GF, Herman AM, Lian X, Raval KK, et al. Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: the matrix sandwich method. Circ Res. 2012;111(9):1125–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. David R, Schwarz F, Rimmbach C, Nathan P, Jung J, Brenner C, et al. Selection of a common multipotent cardiovascular stem cell using the 3.4-kb Mesp1 promoter fragment. Basic Res Cardiol. 2013;108(1):312.

    Article  PubMed  Google Scholar 

  20. Den Hartogh SC, Schreurs C, Monshouwer-Kloots JJ, Davis RP, Elliott DA, Mummery CL, et al. Dual reporter MESP1 mCherry/w-NKX2-5 eGFP/w hESCs enable studying early human cardiac differentiation. Stem Cells. 2015;33(1):56–67.

    Article  Google Scholar 

  21. Wang T, Tang W, Sun S, Ristagno G, Huang Z, Weil MH. Intravenous infusion of bone marrow mesenchymal stem cells improves myocardial function in a rat model of myocardial ischemia. Crit Care Med. 2007;35:2587–93.

    Article  PubMed  Google Scholar 

  22. Xing Y, Hou J, Guo T, Zheng S, Zhou C, Huang H, et al. microRNA-378 promotes mesenchymal stem cell survival and vascularization under hypoxic-ischemic conditions in vitro. Stem Cell Res Ther. 2014;5(6):130.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Kim DH, Marinov GK, Pepke S, Singer ZS, He P, Williams B, et al. Single-cell transcriptome analysis reveals dynamic changes in lncRNA expression during reprogramming. Cell Stem Cell. 2015;16(1):88–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Savić N, Bär D, Leone S, Frommel SC, Weber FA, Vollenweider E, et al. lncRNA maturation to initiate heterochromatin formation in the nucleolus is required for exit from pluripotency in ESCs. Cell Stem Cell. 2014;15(6):720–34.

    Article  PubMed  Google Scholar 

  25. Li YP, Wang Y. Large noncoding RNAs are promising regulators in embryonic stem cells. J Genet Genomics. 2015;42(3):99–105.

    Article  CAS  PubMed  Google Scholar 

  26. Schonrock N, Harvey RP, Mattick JS. Long noncoding RNAs in cardiac development and pathophysiology. Circ Res. 2012;111:1349–62.

    Article  CAS  PubMed  Google Scholar 

  27. Flynn RA, Chang HY. Long noncoding RNAs in cell-fate programming and reprogramming. Cell Stem Cell. 2014;14(6):752–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ghosal S, Das S, Chakrabarti J. Long noncoding RNAs: new players in the molecular mechanism for maintenance and differentiation of pluripotent stem cells. Stem Cells Dev. 2013;22(16):2240–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Bao X, Wu H, Zhu X, Guo X, Hutchins AP, Luo Z, et al. The p53-induced lincRNA-p21 derails somatic cell reprogramming by sustaining H3K9me3 and CpG methylation at pluripotency gene promoters. Cell Res. 2015;25(1):80–92.

    Article  CAS  PubMed  Google Scholar 

  30. Sadahiro T, Yamanaka S, Ieda M. Direct cardiac reprogramming: progress and challenges in basic biology and clinical applications. Circ Res. 2015;116(8):1378–91.

    Article  CAS  PubMed  Google Scholar 

  31. Ounzain S, Pezzuto I, Micheletti R, Burdet F, Sheta R, Nemir M, et al. Functional importance of cardiac enhancer-associated noncoding RNAs in heart development and disease. J Mol Cell Cardiol. 2014;76:55–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ounzain S, Crippa S, Pedrazzini T. Small and long non-coding RNAs in cardiac homeostasis and regeneration. Biochim Biophys Acta. 2013;1833(4):923–33.

    Article  CAS  PubMed  Google Scholar 

  33. Ounzain S, Micheletti R, Arnan C, Plaisance I, Cecchi D, Schroen B, et al. CARMEN, a human super enhancer-associated long noncoding RNA controlling cardiac specification, differentiation and homeostasis. J Mol Cell Cardiol. 2015;89(Pt A):98–112.

    Article  CAS  PubMed  Google Scholar 

  34. Limana F, Bertolami C, Mangoni A, Di Carlo A, Avitabile D, Mocini D, et al. Myocardial infarction induces embryonic reprogramming of epicardial c-kit(+) cells: role of the pericardial fluid. J Mol Cell Cardiol. 2010;48:609–18.

    Article  CAS  PubMed  Google Scholar 

  35. Boregowda SV, Krishnappa V, Haga CL, Ortiz LA, Phinney DG. A clinical indications prediction scale based on TWIST1 for human mesenchymal stem cells. EBioMedicine. 2015;4:62–73.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Camp E, Anderson PJ, Zannettino AC, Gronthos S. Tyrosine kinase receptor c-ros-oncogene 1 mediates TWIST-1 regulation of human mesenchymal stem cell lineage commitment. Bone. 2016;94:98–107.

    Article  PubMed  Google Scholar 

  37. Ishimine H, Yamakawa N, Sasao M. N-Cadherin is a prospective cell surface marker of human mesenchymal stem cells that have high ability for cardiomyocyte differentiation. Biochem Biophys Res Commun. 2013;438(4):753–9.

    Article  CAS  PubMed  Google Scholar 

  38. Lee EJ, Choi EK, Kang SK, Kim GH, Park JY, Kang HJ, et al. N-cadherin determines individual variations in the therapeutic efficacy of human umbilical cord blood-derived mesenchymal stem cells in a rat model of myocardial infarction. Mol Ther. 2012;20(1):155–67.

    Article  CAS  PubMed  Google Scholar 

  39. Chiapparo G, Lin X, Lescroart F. Mesp1 controls the speed, polarity, and directionality of cardiovascular progenitor migration. J Cell Biol. 2016;213(4):463–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Yang J, Kaur K, Ong LL, Eisenberg CA, Eisenberg LM. Inhibition of G9a histone methyltransferase converts bone marrow mesenchymal stem cells to cardiac competent progenitors. Stem Cells Int. 2015;2015:270428.

    PubMed  PubMed Central  Google Scholar 

  41. Behfar A, Yamada S, Crespo-Diaz R, Nesbitt JJ, Rowe LA, Perez-Terzic C, et al. Guided cardiopoiesis enhances therapeutic benefit of bone marrow human mesenchymal stem cells in chronic myocardial infarction. J Am Coll Cardiol. 2010;56(9):721–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

This work was performed by the investigators of the Sun Yat-sen Memorial Hospital of Sun Yat-sen University. The authors thank Shaoxin Zheng for special technical assistance in the experiments. The authors take responsibility for all aspects of the work and have no differences with regard to the data presentation and interpretation.

Funding

This study was supported by National Natural Science Foundation of China (Nos. 81070125, 81270213, and 81670306), Science and Technology Foundation in Guangdong Province (Nos. 2010B031600032 and 2014A020211002), the Fundamental Research Funds for the Central Universities (No. 13ykzd16), and the Medical Science and Technology Research Fund of Guangdong Province (No. A2016264).

Availability of supporting data

All data generated or analyzed during this study are included in this published article.

Authors’ contributions

JH and HL carried out the cell culture, transfection, participated in the statistical analysis, and drafted the manuscript. CZ and HW carried out the molecular assay. TG participated in the vector construction. QW and TZ carried out cell staining and statistical analysis. SZ participated with technical assistance. TW conceived the study and participated in the study design. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethical approval and consent to participate

Animals were obtained from the Animal Experimental Center of the Sun Yat-sen University. All animal handling and procedures were performed in accordance with protocols approved by the Animal Ethics Committee of Sun Yat-sen University (201210016).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tong Wang.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hou, J., Long, H., Zhou, C. et al. Long noncoding RNA Braveheart promotes cardiogenic differentiation of mesenchymal stem cells in vitro. Stem Cell Res Ther 8, 4 (2017). https://0-doi-org.brum.beds.ac.uk/10.1186/s13287-016-0454-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13287-016-0454-5

Keywords