Your privacy, your choice

We use essential cookies to make sure the site can function. We also use optional cookies for advertising, personalisation of content, usage analysis, and social media.

By accepting optional cookies, you consent to the processing of your personal data - including transfers to third parties. Some third parties are outside of the European Economic Area, with varying standards of data protection.

See our privacy policy for more information on the use of your personal data.

for further information and to change your choices.

Skip to main content

LINC01271 promotes fracture healing via regulating miR-19a-3p/PIK3CA axis

Abstract

Objective

Osteoporosis increases the risk of fragility fractures, impacting patients’ lives. This study aimed to investigate whether LINC01271 was involved in the process of fragility fractures and healing, providing a new perspective for its diagnosis and treatment.

Methods

This study included 94 healthy individuals, 82 patients with osteoporosis, and 85 patients with fragility fractures as subjects. RT-qPCR was used to measure the levels of LINC01271, miR-19a-3p, PIK3CA, and osteogenic differentiation markers in osteoblasts and subjects’ serum. Luciferase reporter assays, RIP experiments, and RNA pull-down assays were utilized to verify the target relationships between LINC01271 and miR-19a-3p, as well as between miR-19a-3p and PIK3CA. Cell proliferation and apoptosis were assessed using CCK-8 assays and flow cytometry.

Results

Compared to the healthy control group, the serum levels of LINC01271 were significantly reduced in patients with osteoporosis and fragility fractures. Furthermore, LINC01271 levels increased with time-dependent fracture healing. In vitro studies indicated that LINC01271 boosted osteoblast proliferation, inhibited apoptosis, and augmented osteogenic differences, whereas its inhibition reverses their effects. LINC01271 and PIK3CA were identified as targets of miR-19a-3p, and overexpression of miR-19a-3p could antagonize the effect of LINC01271 in promoting fracture healing.

Conclusion

The results indicated that LINC01271 may play a key role in osteoblast function and fracture healing through its interaction with miR-19a-3p and regulation of PIK3CA.

Introduction

Osteoporosis is a common skeletal disease, and its incidence increases with age [1]. With the loss of bone mass, pathological features such as defects in trabecular microstructure and elevated bone remodeling rates occur [2], lead to impaired bone strength and hence increased fragility [3, 4]. Fragility fractures refer to fractures that occur under normal or minimal trauma [5]. Due to the characteristics of osteoporosis, the incidence of fragility fractures in patients with osteoporosis is often higher thangeneral population [6]. Research suggests that fragility fractures often indicate the presence of osteoporosis [7]. Therefore, fragility fractures are also known as osteoporotic fractures. Fragility fractures can lead to severe consequences such as disability or death [8], and their recurrence rate is extremely high [9]. There is currently some evidence suggesting that medications such as calcitonin, as well as methods like Intermittent Pneumatic Compression (IPC) and Low-Intensity Pulsed Ultrasound (LIPUS), may promote fracture healing, but further clinical validation is still needed [10,11,12]. In addition, there is a need to explore more diagnostic and therapeutic strategies for fracture healing, and a deeper understanding of the mechanisms behind fracture healing is significant for bone regeneration.

Long non-coding RNAs (lncRNAs), which are RNA transcripts longer than 200nt [13]. LncRNAs exhibit tissue-specific expression, suggesting they play various roles in different cell types [14]. The skeleton, as a vital human tissue, also involves lncRNAs in its activities. The physiological activities of five types of bone cells—osteoblasts, osteoclasts, osteocytes, lining cells, and chondrocytes [15]—are all influenced by lncRNAs, and research primarily focusing on osteogenic differentiation and chondrogenesis [16]. lncRNAs such as H19 and MALAT1 are known to regulate osteogenic differentiation [17, 18]. However, the roles and mechanisms of lncRNAs in osteogenic differentiation vary; for instance, LINC00339 can inhibit fracture healing [19], while KCNQ1OT1 promotes osteogenic differentiation [20]. Recent evidence shows that the exosomal lncRNA-mRNA network in the tissue at the fracture site can promote the proliferation and migration of bone marrow stromal cells (BMSCs) [21]. lncRNAs can also regulate osteogenic differentiation through signaling pathways such as Wnt/β-catenin [22]. The therapeutic potential of non-coding RNAs in musculoskeletal-related diseases has been demonstrated [23, 24], thus the role and diagnostic and therapeutic potential of lncRNAs in fracture healing are also being emphasized. LINC01271 is also a type of lncRNAs, but research on LINC01271 is still in its early stages. The few in-depth studies available have mainly concentrated on cancer; for example, one study identified LINC01271 as a human ortholog of MaTAR25, involved in the development of human breast cancer [25]. Notably, a study by Fei et al. found that LINC01271 was differentially expressed in postmenopausal osteoporosis patients by RNA sequencing [26]. Furthermore, age-related reduction in bone density leads to fragility fractures, and bone mesenchymal stem cells (BMSCs) play a crucial role in bone mineralization. Yin et al. analyzed differential expression datasets of lncRNAs in BMSCs from middle-aged and elderly individuals obtained from the GEO database and found that LINC01271 was significantly reduced in the BMSCs of older adults [18]. However, there have been no reports on whether LINC01271 is involved in fragility fractures and their healing. Although various lncRNAs associated with delayed fracture healing have been identified, many studies still lack an in-depth understanding of their molecular mechanisms, and most research has focused solely on fragility fractures or osteoporosis, neglecting the connections between them. Our study includes patients with osteoporosis and fragility fractures and explores the downstream signaling pathways of LINC01271, aiming to investigate the role and clinical value of LINC01271 in fragility fractures. This study also helps to supplement the knowledge system regarding the functional research of specific lncRNAs in fracture healing. Our study may provide new insights into the management of fractures to promote healing.

Materials and methods

Inclusion of clinical subjects

The study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethics Committee of Affiliated Hospital of Zunyi Medical University before the study began. We provided all patients and volunteers with detailed explanations of the study’s content, potential risks, significance, and privacy protection methods. All participants agreed to take part in the study and signed informed consent forms. Before the study, sample size estimation was conducted using G*Power software (http://www.gpower.hhu.de/) with parameters: F test; ANOVA: Fixed effect, omnibus, one-way Statistical test. A minimum of 207 samples was required to achieve α = 0.05, power (1-β) = 0.90, and medium effect size (F = 0.25). Considering a 10% dropout rate, a total of 228 samples (76 per group) were needed. Therefore, the sample size included in our study is larger than this estimate.

A total of 261 subjects were included in this study, consisting of 94 healthy individuals, 82 patients with osteoporosis, and 85 patients with fragility fractures. All patients were treated at Affiliated Hospital of Zunyi Medical University. To reduce errors, we followed the inclusion and exclusion criteria based on previous studies for patient selection: Inclusion criteria for patients with osteoporosis: 1). Patients aged over 50, aligning with the common age of onset for osteoporosis; 2). diagnosed with osteoporosis; 3). Complete medical records available. Exclusion criteria: 1). Patients who have taken osteoporosis prescription drugs for more than three months; 2). Patients with malignant tumors, heart or liver failure, immune system deficiencies, or coagulation disorders; 3). Patients with other diseases affecting bone metabolis. Inclusion criteria for fragility fractures: 1). Patients aged over 45; 2). Meeting the diagnostic criteria for fragility fractures, which are fractures caused by low-energy injuries (e.g., fractures of the vertebral, hip, or forearm/wrist or others); 3). Complete medical records available. Exclusion criteria: 1). Recent use of medications that may interfere with bone healing; 2). Multiple fractures; 3). History of previous fractures; 4). Coexisting severe comorbidities that lead to poor healing, such as diabetes.

Cell culture and transfection

The human osteoblastic cell line hFOB1.19 was cultured in RPMI-1640 medium containing 10% horse serum and 5% fetal bovine serum. The incubation environment was set at 37 °C with a CO2 concentration of 5%.

hFOB1.19 cells were seeded in 6-well plates, and overexpression vectors for LINC01271 (oe-LINC01271) along with its negative controls (oe-NC), miR-19a-3p mimic and its control (mimic NC), as well as miR-19a-3p inhibitor and its control (inhibitor NC), were constructed. The aforementioned viral vectors were mixed with Lipofectamine 3000 and added to the 6-well plates. Cells in the control group did not undergo any transfection with these reagents.

RT-qPCR

Total RNA was extracted using the miRNeasy Mini Kit according to the manufacturer’s instructions. Subsequently, LINC01271 and PIK3CA were reverse transcribed using the PrimpScript RT reagent kit. miR-19a-3p was reverse transcribed using the Mir-X miRNA First-Strand Synthesis Kit. Finally, RT-qPCR was conducted using TB Green Premix Ex Taq II. GAPDH served as the internal control for LINC01271 and PIK3CA, while U6 served as the internal control for miR-19a-3p. The levels of LINC01271, miR-19a-3p, PIK3CA, and the osteogenic differentiation markers osteocalcin (OCN), Runt-related transcription factor 2 (RUNX2) and bone sialoprotein (BSP)mRNA was quantified using the 2−ΔΔCt method.

Cell proliferation

Proliferation of hFOB1.19 cells was evaluated using the Cell Counting Kit-8 (CCK-8) assay. In brief, cells were seeded in 96-well plates, and a certain volume of Cell Counting Kit-8 reaction solution was added at 0, 24, 48, and 72 h of incubation. Following this, cells were further incubated in the dark for 2 h. Finally, absorbance was measured at 450 nm.

Cell apoptosis

The apoptosis of hFOB1.19 cells was assessed using the Annexin V-FITC/PI apoptosis detection kit. Briefly, collected hFOB1.19 cells were washed 3 times with pre-cold PBS and then re-suspended. Next, 5 µL of Annexin V-FITC and 5 µL of PI were added to the cells and incubated for 15 min. Finally, the apoptotic status was analyzed using a flow cytometer. The overall apoptosis rate was calculated as the sum of early and late apoptotic rates.

Bioinformatics analysis

The LncRNASNP2 database predicted the target miRNA for LINC01271. TargetScan, ENCORI, miRNet, miRDB, and miRWalk were conducted to predict targets for miR-19a-3p, and the overlapping targets were examined. In addition, the STRING database was used for protein-protein interaction (PPI) network analysis of overlapping targets. Genes with the top 10 highest degrees in the PPI network were viewed as hub genes.

Luciferase reporter gene assay

hFOB1.19 cells were seeded in 24-well plates. Wild-type (WT) or mutated (MUT) 3’ UTR luciferase reporter plasmids of LINC01271 and PIK3CA were constructed. Lipofectamine 3000 was used to co-transfect miR-19a-3p mimic, mimic NC, miR-19a-3p inhibitor, or inhibitor NC with the aforementioned vectors into the cells. After 48 h of transfection, relative luciferase activity was assessed using a dual-luciferase reporting detection system.

Alkaline phosphatase (ALP)measurement

On days 0, 7, and 14 of osteogenic differentiation, as well as 48 h post-transfection, ALP activity was measured. The cells were seeded into 96-well plate. Then, beta-glycerophosphate and ascorbic acid were added, and after 48 h of incubation, ALP activity was measured using p-nitrophenyl phosphate as the substrate, with absorbance assessed at a wavelength of 405 nm.

Nuclear and cytoplasmic fractionation

Nuclear and cytoplasmic RNA were isolated using the SurePrep Nuclear or Cytoplasmic RNA Purification Kit. Lysis buffer was utilized to lyse hFOB1.19 cells, followed by RNA separation using a cell disruption and cell fractionation device. Finally, LINC01271 levels in the fractions were evaluated using RT-qPCR.

RNA immunoprecipitation (RIP) assay

The binding relationship between miR-19a-3p and LINC01271, as well as between miR-19a-3p and PIK3CA, was verified using the RIP assay kit. First, cells were resuspended in RIP lysis buffer, then centrifuged and incubated overnight with Anti-Ago2 or Anti-IgG. The RNA from the obtained immunoprecipitates was extracted using the GenElute™ Total RNA Purification Kit. Finally, the enrichment of LINC01271, miR-19a-3p, and PIK3CA in the cells was analyzed using RT-qPCR.

RNA pull-down assay

An RNA Pull-Down Assay was conducted using an RNA Pull Down Assay Kit. In brief, hFOB1.19 cells were first lysed and then incubated with biotinylated miR-19a-3p probes and negative control. Subsequently, streptavidin-coated magnetic beads were added. After incubating overnight, LINC01271 levels were measured using RT-qPCR.

Statistical analysis

Data analysis was performed using GraphPad Prism 9.0 and SPSS 23.0. Each experiment was replicated at least three times biologically. Results were expressed as the mean ± standard deviation (SD). Group differences were analyzed using a two-tailed Student’s t-test or one-way ANOVA. P < 0.05 was considered statistically significant.

Results

Expression levels of LINC01271 in osteoporosis and fragility fracture patients

The study first assessed the clinical information of the three groups of subjects. As shown in Table 1, there were no statistically significant differences in age and BMI index among the three groups (P > 0.05). Additionally, the T scores and 25-(OH) Vitamin D levels of osteoporosis and fragility fracture patients were obviously lower than healthy group (P < 0.001), with fragility fracture patients having the lowest values. Among the 85 fragility fracture patients, there were 45 cases of hip fractures, 27 cases of forearm/wrist fractures, 4 cases of vertebral fractures, and 9 cases of fractures at other sites.

Table 1 Clinical baseline characteristics of participants

By analyzing serum samples from the subjects, it was found that LINC01271 levels in the serum of osteoporosis were notably decreased compared to healthy individuals (P < 0.001, Fig. 1A). Notably, as shown in Fig. 1B, the ROC curve indicated that LINC01271 could differentiate between healthy individuals and osteoporotic patients, with an AUC of 0.886 (95% CI, 0.832–0.940), and sensitivities and specificities of 87.23% and 87.71%, respectively. Furthermore, serum LINC01271 levels were noticeably upregulated in patients with fragility fracture than the osteoporosis (P < 0.001, Fig. 1C). And LINC01271 levels increased with the healing time of fracture patients (P < 0.05, Fig. 1D).

Fig. 1
figure 1

Expression Levels of LINC01271 in Osteoporosis and Fragility Fracture Patients. (A) Reduced levels of LINC01271 in patients with osteoporosis. (B) ROC curves of LINC01271 in osteoporosis. (C) Reduced levels of LINC01271 in patients with fragility fractures. (D) Increased levels of LINC01271 in patients with fractures with healing time. * P < 0.05, **** P < 0.0001

Expression levels of LINC01271 in osteogenic differentiation of osteoblasts

As shown in Fig. 2A and B, the levels of osteogenic differentiation markers OCN, BSP, RUNX2-related mRNA, and ALP activity in hFOB1.19 cells significantly increased with the duration of osteogenic differentiation (P < 0.01). Meanwhile, LINC01271 levels in the cells exhibited a similar increasing trend with the duration of osteogenic differentiation (P < 0.001, Fig. 2C).

Fig. 2
figure 2

Expression levels of LINC01271 in osteoblast osteogenic differentiation. ALP activity(A), markers of osteogenic differentiation (OCN, BSP, RUNX2) (B), and LINC01271 levels(C) increased with the duration of osteogenic differentiation. ** P < 0.01, *** P < 0.001, **** P < 0.0001

Effects of LINC01271 on osteoblast proliferation and differentiation

To explore the effects of LINC01271 on the osteogenic process, functional assays were conducted to modulate LINC01271 levels in osteoblasts. Transfection of oe-LINC01271 resulted in increased levels of LINC01271, whereas transfection of si-LINC01271 resulted in decreased levels (P < 0.001, Fig. 3A). Moreover, increasing LINC01271 significantly enhanced cell proliferation, as well as the levels of osteogenic differentiation markers and ALP activity, while the apoptosis rate decreased significantly with the rise in LINC01271 levels (P < 0.01, Fig. 3B-E). Conversely, inhibiting LINC01271 expression resulted in decreased cell proliferation, increased apoptosis, and significantly reduced levels of osteogenic differentiation markers and ALP activity (P < 0.01, Fig. 3B-E).

Fig. 3
figure 3

Effect of LINC01271 on osteoblast proliferation and differentiation. Effects of modulating LINC01271 on LINC01271 levels(A), cell proliferation(B), apoptosis(C), ALP activity(D), and markers of osteogenic differentiation(E). ** P < 0.01, *** P < 0.001, **** P < 0.0001

LINC01271 sponged miR-19a-3p

LINC01271 was primarily enriched in the cytoplasm (Fig. 4A), indicating that it met the prerequisites for being a competing endogenous RNA (ceRNA). Subsequently, we predicted the target of LINC01271 using the lncRNASNP2 online database and identified the binding site of LINC01271 to miR-19a-3p (Fig. 4B). Furthermore, we validated their targeted binding relationship through luciferase reporter assays, RIP experiments, and RNA pull-down assays (Fig. 4C-E). Clinically, miR-19a-3p levels in osteoporosis and fragility fracture patients were found to be significantly increased compared to healthy volunteers (P < 0.001, Fig. 4F). Correlation analysis revealed a significant negative correlation between LINC01271 and miR-19a-3p levels in fragility fracture patients (r = -0.757, P < 0.001, Fig. 4G). Based on this, we further analyzed the changes in miR-19a-3p levels both clinically and in cells. The results indicated that as the duration of osteogenic differentiation or fracture healing increased, the levels of miR-19a-3p significantly decreased (P < 0.05, Fig. 4H and I). Additionally, we found thatmiR-19a-3p levels decreased as LINC01271 levels increased (P < 0.01), while inhibiting LINC01271 led to a statistical increase in miR-19a-3p levels (P < 0.001, Fig. 4J).

Fig. 4
figure 4

LINC01271 sponged miR-19a-3p. (A) Sublocalization analysis of LINC01271. (B) Binding site of LINC01271 and miR-19a-3p. (C) Luciferase reporter gene assay. (D) RIP assay. (E) RNA pull-down assay. (F) Correlation analysis of miR-19a-3p. (G) LINC01271 and miR-19a-3p in serum from osteoporosis patients and fragility fracture patients. Both miR-19a-3p levels decreased significantly with increasing osteogenic differentiation(H) or fracture healing time(I). J. Effect of modulating LINC01271 on miR-19a-3p. * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001

Upregulation of miR-19a-3p reversed the effects of LINC01271 on osteogenic activity

To further investigate the impact of the LINC01271/miR-19a-3p axis on fracture healing, we co-transfected miR-19a-3p mimic while overexpressing LINC01271. The results showed that overexpression of LINC01271 inhibited the expression of miR-19a-3p, while transfection with the miR-19a-3p mimic reversed the inhibitory effect of LINC01271 on miR-19a-3p (P < 0.001, Fig. 5A). Overexpression of LINC01271 promoted osteoblast proliferation, inhibited apoptosis, and enhanced osteogenic differentiation markers levels and ALP activity (P < 0.05). Importantly, overexpression of miR-19a-3p greatly antagonized the promotion of osteogenic differentiation by LINC01271 (P < 0.01, Fig. 5B-E).

Fig. 5
figure 5

Up-regulation of miR-19a-3p reverses the effect of LINC01271 on osteogenic activity. Effect of co-transfection of oe-LINC01271 and miR-19a-3p mimic on miR-19a-3p levels(A), cell proliferation(B), apoptosis (C), and ALP activity(D), as well as markers of osteogenic differentiation(E). * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001

Target relationship between PIK3CA and miR-19a-3p

Using TargetScan, ENCORI, miRWalk, miRDB and miRNet online databases for miR-19a-3p target gene prediction, we identified a total of 81 overlapping targets (Fig. 6A). We then analyzed these 81 target genes using the PPI network and determined the top 10 hub genes based on the Degree topological algorithm, among which PIK3CA caught our attention (Fig. 6B). The binding site between the 3’-UTR of the PIK3CA and miR-19a-3p was illustrated in Fig. 6C. We further validated the interaction between PIK3CA and miR-19a-3p through luciferase reporter assays, RIP experiments, and RNA pull-down assays (P < 0.01, Fig. 6D-F). In addition, in contrast to miR-19a-3p, PIK3CA levels in the serum of osteoporosis and fragility fracture patients were significantly decreased (P < 0.001, Fig. 6G). And in patients with fragility fractures, PIK3CA was significantly negatively correlated with miR-19a-3p levels (r = -0.830, P < 0.001, Fig. 6H), while PIK3CA levels showed a significant positive correlation with LINC01271 levels (r = 0.619, P < 0.001, Fig. 6I). Furthermore, PIK3CA levels significantly increased with the healing time of fracture patients (P < 0.05, Fig. 6J). After overexpression of LINC01271, PIK3CA levels significantly increased, while transfection with the miR-19a-3p mimic led to a notable decrease in PIK3CA levels (P < 0.001, Fig. 6K).

Fig. 6
figure 6

Target relationship between PIK3CA and miR-19a-3p. (A) miR-19a-3p target gene prediction Venn plots. (B) PPI network. (C) Binding sites of PIK3CA and miR-19a-3p. (D) Luciferase reporter gene assay. (E) RIP assay. (F) RNA pull-down assay. (G) PIK3CA levels in patients with osteoporosis and fragility fractures. Correlation analysis of PIK3CA with miR-19a-3p(H) and LINC01271(I). J. Changes in PIK3CA levels with time of osteogenic differentiation. K. Effects of modulating LINC01271 and miR-19a-3p on PIK3CA. * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001

Discussion

It is well-known that osteoporosis patients have a higher risk of fractures. In middle-aged and older individuals, particularly postmenopausal women, the incidence of osteoporosis increases due to changes in estrogen levels during menopause [27], indirectly leading to a rise in the occurrence of fragility fractures. Fragility fractures not only impose significant clinical and economic burdens on individuals and society, but patients often have a poor prognosis [28, 29]. Therefore, it is essential to explore the underlying mechanisms of osteoporosis and its associated fractures. With advancements in technology, research on the epigenetic regulation of osteoporosis and related fractures mediated by non-coding RNAs has been increasing [30], research on the roles and therapeutic potential of non-coding RNAs in skeletal-related diseases is also a hot topic [31,32,33]. It has been reported that many lncRNAs are associated with the occurrence, treatment, and prognosis of orthopedic diseases [34]. In our study, we detected decreased levels of LINC01271 in the serum of patients with osteoporosis and fragility fractures, and we found that LINC01271 levels increased with fracture healing time. ROC analysis also demonstrated that LINC01271 could significantly differentiate between healthy individuals and osteoporosis patients. Similarly, Li et al. reported a significant downregulation of LINC01271 in bone marrow mesenchymal stem cells from osteoporosis patients [35]. Based on this analysis, we conducted further studies using hFOB1.19 cells. The results indicated that overexpression of LINC01271 promoted osteoblast proliferation and differentiation, thereby participating in the fracture healing process.

LncRNAs can regulate downstream target genes by competitively binding to miRNAs [36]. Therefore, we further investigated the potential mechanism of LINC01271 in fracture healing. Our research revealed a specific binding relationship between LINC01271 and miR-19a-3p, with LINC01271 negatively regulating miR-19a-3p. miR-19a-3p wasrecognized as a crucial regulatory gene involved in the differentiation of bone cells [37]. Additionally, previous studies have demonstrated that miR-19a-3p promotes the development of osteoporosis by inhibiting osteogenic differentiation of bone marrow stem cells [38]. In our investigation, we found that in contrast to LINC01271, miR-19a-3p levels were significantly higher in patients with osteoporosis and fragility fractures. This observation aligned with the findings reported by Pan et al. [39]. Furthermore, we investigated the effect of introducing miR-19a-3p mimics on the promotion of fracture healing by LINC01271. We found that overexpression of miR-19a-3p could antagonize the effects of LINC01271 on fracture healing, specifically inhibiting osteogenic differentiation markers and promoting apoptosis. However, there is current evidence showing that LINC01271 is involved in the proliferation and migration of glioma cells [40], and miR-19a-3p has also been found to regulate various tumor processes [41, 42]. This seems to conflict with the molecular role of the LINC01271/miR-19a-3p axis in fracture healing observed in this study. This may be due to the fact that the same genes play different roles in different physiological processes and signaling networks [43].

Based on the regulatory mechanism by which miRNAs can bind to target mRNA and degrade or inhibit its translation [44], we conducted an in-depth study of the downstream target genes of the LINC01271/miR-19a-3p axis. In our research, phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) was identified as a target mRNA of miR-19a-3p, and previous studies have also reported the targeting relationship between miR-19a-3p and PIK3CA [45]. Notably, upregulation of PIK3CA in osteomyelitis patients was found to alleviate the inhibitory effect of miR-320a on osteogenic differentiation [46]. And our study found that PIK3CA levels were markedly decreased in the serum of patients with osteoporosis and fragility fractures, which was consistent with the findings of Ma et al. [47]. Moreover, our research confirmed that PIK3CA was negatively correlated with miR-19a-3p, while positively correlated with LINC01271. In contrast to miR-19a-3p, PIK3CA levels increased with osteogenic differentiation and rose with increasing levels of LINC01271. Additionally, transfecting miR-19a-3p mimics inhibited PIK3CA expression. These results collectively indicated that the potential mechanism by which miR-19a-3p influences fracture healing involves targeting PIK3CA. Nevertheless, our study also has certain limitations. Although the study involved the mechanistic connections between LINC01271, miR-19a-3p, and PIK3CA, the downstream effects of PIK3CA activation on osteoblast function have yet to be demonstrated, which will be a focus of our future work.

In conclusion, our results supported the hypothesis that LINC01271 is associated with osteoporosis and fragility fractures. Overexpression of LINC01271 can stimulate osteoblast proliferation and differentiation by targeting miR-19a-3p, thereby promoting fracture healing, while PIK3CA may serve as a target in the potential mechanism of miR-19a-3p. This study provides new insights for developing diagnostic biomarkers for osteoporotic fractures and offers potential targets for the treatment and prognosis of osteoporosis and fragility fractures.

Data availability

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

References

  1. Kelsey JL. Risk factors for osteoporosis and associated fractures. Public Health Rep. 1989;104(SupplSuppl):14–20.

    PubMed  PubMed Central  Google Scholar 

  2. Armas LA, Recker RR. Pathophysiology of osteoporosis: new mechanistic insights. Endocrinol Metab Clin North Am. 2012;41(3):475–86.

    Article  CAS  PubMed  Google Scholar 

  3. Lane JM, Russell L, Khan SN, Osteoporosis. Clin Orthop Relat Res. 2000; (372): 139–50.

  4. Hubacek JA, Weichetova M. [Genetic determinants of osteoporosis]. Cas Lek Cesk. 2005;144(1):5–9. discussion – 10.

    CAS  PubMed  Google Scholar 

  5. Shoji MM, Ingall EM, Rozental TD. Upper Extremity Fragility fractures. J Hand Surg Am. 2021;46(2):126–32.

    Article  PubMed  Google Scholar 

  6. Prasad B, Ferguson T, Tangri N, Ng CY, Nickolas TL. Association of Bone Mineral Density with fractures across the spectrum of chronic kidney disease: the Regina CKD-MBD Study. Can J Kidney Health Dis. 2019; 62054358119870539.

  7. Walters S, Khan T, Ong T, Sahota O. Fracture liaison services: improving outcomes for patients with osteoporosis. Clin Interv Aging. 2017; 12117–27.

  8. Tan JT, Cheung CL, Cheung KS. Relationship between Helicobacter pylori infection, osteoporosis, and fracture. J Gastroenterol Hepatol. 2024;39(10):2006–17.

    Article  PubMed  Google Scholar 

  9. Yang Z, Feng L, Wang M, Li Y, Bai S, Lu X et al. Sesamin promotes osteoporotic fracture Healing by activating chondrogenesis and angiogenesis pathways. Nutrients. 2022; 14(10).

  10. Migliorini F, Cocconi F, Vecchio G, Schaefer L, Koettnitz J, Maffulli N. Pharmacological agents for bone fracture healing: talking points from recent clinical trials. Expert Opin Investig Drugs. 2023;32(9):855–65.

    Article  CAS  PubMed  Google Scholar 

  11. Khanna A, Gougoulias N, Maffulli N. Intermittent pneumatic compression in fracture and soft-tissue injuries healing. Br Med Bull. 2008;88(1):147–56.

    Article  PubMed  Google Scholar 

  12. Martinez de Albornoz P, Khanna A, Longo UG, Forriol F, Maffulli N. The evidence of low-intensity pulsed ultrasound for in vitro, animal and human fracture healing. Br Med Bull. 2011; 10039–57.

  13. Bridges MC, Daulagala AC, Kourtidis A. LNCcation: lncRNA localization and function. J Cell Biol. 2021; 220(2).

  14. Zuckerman B, Ulitsky I. Predictive models of subcellular localization of long RNAs. RNA. 2019;25(5):557–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Balestri W, Hickman GJ, Morris RH, Hunt JA, Reinwald Y. Triphasic 3D in Vitro Model of bone-tendon-muscle interfaces to study their regeneration. Cells. 2023; 12(2).

  16. Li D, Yang C, Yin C, Zhao F, Chen Z, Tian Y, et al. LncRNA, important player in Bone Development and Disease. Endocr Metab Immune Disord Drug Targets. 2020;20(1):50–66.

    Article  PubMed  Google Scholar 

  17. Huang Y, Zheng Y, Jia L, Li W, Long Noncoding. RNA H19 promotes osteoblast differentiation Via TGF-beta1/Smad3/HDAC signaling pathway by deriving miR-675. Stem Cells. 2015;33(12):3481–92.

    Article  CAS  PubMed  Google Scholar 

  18. Yin J, Zheng Z, Zeng X, Zhao Y, Ai Z, Yu M et al. lncRNA MALAT1 mediates osteogenic differentiation of bone mesenchymal stem cells by sponging miR-129-5p. PeerJ. 2022; 10e13355.

  19. Li Y, Sun Y, Ma K, Wang S, Wang Z, Huang L. Functional mechanism and clinical implications of LINC00339 in delayed fracture healing. J Orthop Surg Res. 2024;19(1):511.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Chen L, Xiong Y, Yan C, Zhou W, Endo Y, Xue H, et al. LncRNA KCNQ1OT1 accelerates fracture healing via modulating miR-701-3p/FGFR3 axis. FASEB J. 2020;34(4):5208–22.

    Article  CAS  PubMed  Google Scholar 

  21. Wang D, Diao S, Zhou X, Zhou J, Liu Y. A new method regulates bone fracture tissue exosome lncRNA-mRNA to promote mesenchymal stem cell proliferation and migration. Injury. 2024;55(2):111210.

    Article  PubMed  Google Scholar 

  22. Mai YX, Li ZP, Pang FX, Zhou ST, Li N, Wang YY et al. Aucubin promotes osteogenic differentiation and facilitates bone formation through the lncRNA-H19 driven Wnt/beta-Catenin Signaling Regulatory Axis. Stem Cells Int. 2024; 20245388064.

  23. Giordano L, Porta GD, Peretti GM, Maffulli N. Therapeutic potential of microRNA in tendon injuries. Br Med Bull. 2020;133(1):79–94.

    Article  CAS  PubMed  Google Scholar 

  24. Oliviero A, Della Porta G, Peretti GM, Maffulli N. MicroRNA in osteoarthritis: physiopathology, diagnosis and therapeutic challenge. Br Med Bull. 2019;130(1):137–47.

    Article  CAS  PubMed  Google Scholar 

  25. Chang KC, Diermeier SD, Yu AT, Brine LD, Russo S, Bhatia S, et al. MaTAR25 lncRNA regulates the Tensin1 gene to impact breast cancer progression. Nat Commun. 2020;11(1):6438.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Fei Q, Bai X, Lin J, Meng H, Yang Y, Guo A. Identification of aberrantly expressed long non-coding RNAs in postmenopausal osteoporosis. Int J Mol Med. 2018;41(6):3537–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. LeBoff MS, Greenspan SL, Insogna KL, Lewiecki EM, Saag KG, Singer AJ, et al. The clinician’s guide to prevention and treatment of osteoporosis. Osteoporos Int. 2022;33(10):2049–102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Barron RL, Oster G, Grauer A, Crittenden DB, Weycker D. Determinants of imminent fracture risk in postmenopausal women with osteoporosis. Osteoporos Int. 2020;31(11):2103–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Burge R, Dawson-Hughes B, Solomon DH, Wong JB, King A, Tosteson A. Incidence and economic burden of osteoporosis-related fractures in the United States, 2005–2025. J Bone Min Res. 2007;22(3):465–75.

    Article  Google Scholar 

  30. Ghayor C, Weber FE. Epigenetic regulation of bone remodeling and its impacts in osteoporosis. Int J Mol Sci 2016; 17(9).

  31. Gargano G, Oliva F, Oliviero A, Maffulli N. Small interfering RNAs in the management of human rheumatoid arthritis. Br Med Bull. 2022;142(1):34–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gargano G, Asparago G, Spiezia F, Oliva F, Maffulli N. Small interfering RNAs in the management of human osteoporosis. Br Med Bull. 2023;148(1):58–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gargano G, Oliviero A, Oliva F, Maffulli N. Small interfering RNAs in tendon homeostasis. Br Med Bull. 2021;138(1):58–67.

    Article  CAS  PubMed  Google Scholar 

  34. Zhou Z, Chen J, Huang Y, Liu D, Chen S, Qin S. Long noncoding RNA GAS5: a new factor involved in Bone diseases. Front Cell Dev Biol. 2021; 9807419.

  35. Li B, Wang J, Xu F, Wang Q, Liu Q, Wang G, et al. LncRNA RAD51-AS1 regulates human bone marrow mesenchymal stem cells via Interaction with YBX1 to ameliorate osteoporosis. Stem Cell Rev Rep. 2023;19(1):170–87.

    Article  CAS  PubMed  Google Scholar 

  36. Guo X, Zhang J, Han X, Wang G. LncRNA SNHG1 delayed Fracture Healing via modulating miR-181a-5p/PTEN Axis. J Invest Surg. 2022;35(6):1304–12.

    Article  PubMed  Google Scholar 

  37. Zhu D, Fang H, Yu H, Liu P, Yang Q, Luo P, et al. Alcohol-induced inhibition of bone formation and neovascularization contributes to the failure of fracture healing via the miR-19a-3p/FOXF2 axis. Bone Joint Res. 2022;11(6):386–97.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Chen S, Li Y, Zhi S, Ding Z, Huang Y, Wang W, et al. lncRNA Xist regulates osteoblast differentiation by sponging miR-19a-3p in aging-induced osteoporosis. Aging Dis. 2020;11(5):1058–68.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Pan LX, Ding W. LncRNA HAGLR accelerates femoral neck fracture healing through negatively regulating miRNA-19a-3p. Eur Rev Med Pharmacol Sci. 2020;24(8):4080–7.

    PubMed  Google Scholar 

  40. Xia Z, Tu R, Liu F, Zhang H, Dai Z, Wang Z, et al. PD-L1-related IncRNAs are associated with malignant characteristics and immune microenvironment in glioma. Aging. 2023;15(19):10785–810.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Du L, Dou K, Zhang D, Xia H, Liang N, Wang N et al. MiR-19a-3p Promotes Aerobic Glycolysis in Ovarian Cancer Cells via IGFBP3/PI3K/AKT Pathway. Folia Biol (Praha). 2023; 69(5–6): 163 – 72.

  42. Yao ZX, Tu JH, Liu YL, Xue XF, Qin L. Long non-coding RNA LINC00342 promotes the Proliferation, Invasion, and Migration of Primary Hepatocellular Carcinoma Cells by regulating the expression of miRNA-19a-3p, miRNA-545-5p, and miRNA-203a-3p. Biochem Genet. 2024;62(2):675–97.

    Article  CAS  PubMed  Google Scholar 

  43. Ni J, Tian W, Liang S, Wang H, Ren Y. Promoter methylation-mediated silencing of the MiR-192-5p promotes Endometrial Cancer Progression by Targeting ALX1. Int J Med Sci. 2021;18(12):2510–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yu C, Chen B, Su H, Yang Y. Long non-coding RNA MIAT serves as a biomarker of fragility fracture and promotes fracture healing. J Orthop Surg Res. 2024;19(1):343.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Liu M, Yang P, Fu D, Gao T, Deng X, Shao M, et al. Allicin protects against myocardial I/R by accelerating angiogenesis via the miR-19a-3p/PI3K/AKT axis. Aging. 2021;13(19):22843–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gao D, Shi J, Lu S, Li J, Lv K, Xu Y, et al. METTL3 accelerates staphylococcal protein A (SpA)-induced osteomyelitis progression by regulating m6A methylation-modified miR-320a. J Orthop Surg Res. 2024;19(1):729.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Ma W, Li C. Enhancing postmenopausal osteoporosis: a study of KLF2 transcription factor secretion and PI3K-Akt signaling pathway activation by PIK3CA in bone marrow mesenchymal stem cells. Arch Med Sci. 2024;20(3):918–37.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, Q.C., L.H., W.J. and W.X., B.C.; Data curation, Q.C., L.H., W.J., M.H. and J.S.; Formal analysis, J.L. and H.S.; Funding acquisition, W.X., B.C.; Investigation, J.L. and H.S.; Methodology, Q.C., L.H., M.H. and J.S., B.C.; Project administration, W.X., B.C.; Resources, J.L. and H.S.; Software, J.L. and H.S.; Supervision, W.X., B.C.; Validation, W.J., M.H. and J.S.; Visualization, W.X.; Roles/Writing - original draft, W.J.; Writing - review & editing, Q.C., L.H., W.X., B.C.

Corresponding authors

Correspondence to Wei Xiong or Bin Chen.

Ethics declarations

Ethics approval and consent to participate

The study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethics Committee of Affiliated Hospital of Zunyi Medical University before the study began. The participants’ right to be informed about the study was ensured and agreed to participate in the study.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, Q., Huang, L., Ji, W. et al. LINC01271 promotes fracture healing via regulating miR-19a-3p/PIK3CA axis. J Orthop Surg Res 20, 33 (2025). https://doi.org/10.1186/s13018-024-05444-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13018-024-05444-x

Keywords