header advert
Bone & Joint Open Logo

Receive monthly Table of Contents alerts from Bone & Joint Open

Comprehensive article alerts can be set up and managed through your account settings

View my account settings

Visit Bone & Joint Open at:

Loading...

Loading...

Open Access

Hip

MicroRNA expression analysis in peripheral blood and soft-tissue of patients with periprosthetic hip infection

a prospective controlled pilot study



Download PDF

Abstract

Aims

Current diagnostic tools are not always able to effectively identify periprosthetic joint infections (PJIs). Recent studies suggest that circulating microRNAs (miRNAs) undergo changes under pathological conditions such as infection. The aim of this study was to analyze miRNA expression in hip arthroplasty PJI patients.

Methods

This was a prospective pilot study, including 24 patients divided into three groups, with eight patients each undergoing revision of their hip arthroplasty due to aseptic reasons, and low- and high-grade PJI, respectively. The number of intraoperative samples and the incidence of positive cultures were recorded for each patient. Additionally, venous blood samples and periarticular tissue samples were collected from each patient to determine miRNA expressions between the groups. MiRNA screening was performed by small RNA-sequencing using the miRNA next generation sequencing (NGS) discovery (miND) pipeline.

Results

Overall, several miRNAs in plasma and tissue were identified to be progressively deregulated according to ongoing PJI. When comparing the plasma samples, patients with a high-grade infection showed significantly higher expression levels for hsa-miR-21-3p, hsa-miR-1290, and hsa-miR-4488, and lower expression levels for hsa-miR-130a-3p and hsa-miR-451a compared to the aseptic group. Furthermore, the high-grade group showed a significantly higher regulated expression level of hsa-miR-1260a and lower expression levels for hsa-miR-26a-5p, hsa-miR-26b-5p, hsa-miR-148b-5p, hsa-miR-301a-3p, hsa-miR-451a, and hsa-miR-454-3p compared to the low-grade group. No significant differences were found between the low-grade and aseptic groups. When comparing the tissue samples, the high-grade group showed significantly higher expression levels for 23 different miRNAs and lower expression levels for hsa-miR-2110 and hsa-miR-3200-3p compared to the aseptic group. No significant differences were found in miRNA expression between the high- and low-grade groups, as well as between the low-grade and aseptic groups.

Conclusion

With this prospective pilot study, we were able to identify a circulating miRNA signature correlating with high-grade PJI compared to aseptic patients undergoing hip arthroplasty revision. Our data contribute to establishing miRNA signatures as potential novel diagnostic and prognostic biomarkers for PJI.

Cite this article: Bone Jt Open 2024;5(6):479–488.

Take home message

With this prospective pilot study, we were able to identify a circulating microRNA (miRNA) signature correlating with high-grade periprosthetic joint infection (PJI) compared to aseptic patients undergoing hip arthroplasty revision.

Our data contribute to establishing miRNA signatures as potential novel diagnostic and prognostic biomarkers for PJI.

Introduction

Periprosthetic joint infection (PJI) is one of the most devastating complications after hip arthroplasty,1 and is commonly treated surgically by debridement, antibiotics and implant retention (DAIR), and single- or multistage revision.2,3 With rising numbers of arthroplasties being performed, and as a result of the growing elderly population, a subsequent increase of PJI has to be expected,4-6 which urges the need to diagnose PJI accurately and promptly for a successful treatment in these patients with a lasting infection-free survival.

Despite the eminent increase in research dealing with the accurate and sensitive diagnosis of hip PJI in recent years, PJI remains a diagnostic challenge.7-9 Widely used blood-based tests with serum biomarkers, such as CRP or ESR, may be very limited in detecting low-grade infections in particular, misdiagnosing more than one-third of patients.7,10-13 To address these challenges, researchers have explored alternative diagnostic methods such as advanced imaging techniques, molecular assays, and biomarker profiling.14,15 Moreover, investigations into the host immune response and identification of specific biomarkers associated with hip PJIs have gained attention as potential diagnostic tools.16

Numerous studies have explored the role of microRNA (miRNA) regulation in the immune response against bacterial pathogens.17-24 miRNAs are small non-coding RNAs, typically 20 to 22 nucleotides in length, that regulate biological processes including cellular differentiation, development, apoptosis, tumorigenesis, and targeting of foreign pathogens.24-29 Due to their stability within biofluids, especially in blood serum,30 and their characteristic profiles in various infectious diseases, miRNAs are promising PJI biomarker candidates with extensive diagnostic potential.31,32 Currently, there is no study in the literature investigating the diagnostic potential role of miRNA profiling for PJI caused by different virulent microorganisms. The purpose of this pilot study was to analyze different miRNA expressions from blood serum and periprosthetic tissue of patients with PJI of hip arthroplasty, and compare these with patients who had an aseptic revision arthroplasty.

Methods

Study design and cohort

The study protocol was reviewed and approved by the institutional ethics committee prior to commencement (EA1/283/19). In this prospective single-centre controlled pilot study, consecutive patients who underwent septic or aseptic revision surgery of hip arthroplasty in our institution (Charité University Hospital, Center for Musculoskeletal Surgery, Berlin, Germany) between November 2021 and July 2022 were enrolled to create three groups (aseptic revision, septic revision due to low-grade PJI, and septic revision due to high-grade infection), each consisting of eight patients. Exclusion criteria comprised concomitant infections of organs other than the ipsilateral hip joint, aged under 18 years, active tumour disease, and antibiotic treatment in the last four weeks preoperatively.

The following data were recorded prospectively for each patient: sex and age, involved joint, clinical symptoms, surgical history of the involved joint, time interval between primary arthroplasty and revision surgery, radiological assessment, and type of revision surgery.

Standard diagnostic protocol and definition of PJI

The standard diagnostic protocol was established using institutional criteria according to European Bone and Joint Infection Society (EBJIS) to identify PJI.33 Infection was classified as low-grade if no signs of acute inflammatory symptoms such as fever, erythema, and warmth were present. The presence of acute signs or symptoms of local inflammation and/or sinus tract with evidence of communication to the joint or visualization of the prosthesis was considered as high-grade infection. The routine PJI protocol at our institution included laboratory values such as serum CRP levels, leucocyte counts, and the proportion of polymorphonuclear leucocytes in aspiration, as well as microbiological and histopathological findings at the time of revision surgery. At least five periprosthetic tissue cultures from various suspicious surgical sites, and at least one specimen for histopathological analysis, in addition to a sonication analysis of retrieved implants, were obtained intraoperatively for each patient. The histopathological analysis was based on the consensus classification of the periprosthetic membrane and neosynovium (formerly called the “synovial-like interface membrane”) presented at the national level in 2004 and at the international level in 2006.34-37

Specimens for microbiological analysis were collected with a different, sterile instrument each time and placed immediately into sterile containers without being touched, and analyzed within one hour postoperatively. The microbiological specimens, as well as sonication fluid cultures, were plated onto aerobic and anaerobic sheep blood agar plates and incubated for 14 days. Sonication was performed as previously described.9 The number of intraoperative samples and the incidence of positive cultures were documented for each patient. Moreover, radiological and intraoperative evaluation regarding component loosening was recorded. Furthermore, venous blood samples and intraoperative samples from periprosthetic tissue (in patients with PJI, from tissue with punctum maximum of infection) were collected, and plasma and tissue expression levels of different miRNAs were analyzed by small RNA-sequencing (Supplementary Material).38

MiRNA analysis

MiRNA screening and the statistical analysis of preprocessed next-generation sequencing (NGS) data were conducted by TAmiRNA (Austria) using the miRNA NGS discovery (miND) pipeline.39 Total RNA was extracted from both plasma and tissue samples using the miRNeasy mini kit (Qiagen, Germany). For plasma samples, the total RNA yield was enhanced by addition of glycogen to the aqueous phase prior to isopropanol precipitation. Total RNAs were eluded in 30 µl nuclease-free water. For tissue samples, RNA yield and integrity was checked using the Bioanalyzer RNA 6000 nano chips (Agilent, USA). For plasma, total RNA concentrations are low, therefore a fixed volume of 8.5 µl was used for library preparation using the RealSeq Biosciences kit. Libraries were quality-controlled and pooled at equimolar rates prior to sequencing on an Illumina Novaseq 6000 SP1 flow cell with 100 cycles (single-end). Fastq files were used for data analysis: following quality filtering, annotation, and filtering of low-abundant miRNAs using the “independent filtering” method provided through DESeq2, differential expression analysis between selected groups was performed using EdgeR with p-value adjustment using the Benjamini-Hochberg method to obtain false-discovery rates (FDRs). A FDR cut-off of 0.05 was applied to identify differentially regulated miRNAs. To visualize miRNA abundance and analyze correlation of miRNAs to clinical data, reads per million (RPM) normalization was applied to correct for variation in sequencing depth.

Statistical analysis

Patients were trichotomized into three groups depending on the positive culture and grade of the PJI (aseptic vs low-grade vs high-grade). Chi-squared test and Fisher’s exact test were used to find significant differences between categorical variables. For continuous data, Shapiro-Wilk test was performed to assess normality. Analysis of variance (ANOVA) was used to determine if there was a significant difference between the three groups in terms of mean CRP level, mean leucocyte count in aspiration, and mean time interval between primary arthroplasty and revision surgery. The identification of the differences in detail was realized by the Fisher’s least significant difference test. For non-normally distributed values, the Kruskal-Wallis test was applied. Correlation between clinical data (CRP) and microRNA expression values (reads per million) was performed using Spearman correlation and Spearman rank sum test together with Benjamini-Hochberg adjustment, which was applied to assess statistical significance. For statistical analysis, SPSS Statistics v. 29.0 software (IBM, USA) was employed. The results were given as mean and standard deviation (SD) or as number and percentage. A p-value < 0.05 was considered significant in each case.

Results

Patient characteristics

The mean age of all 24 patients was 70.0 years (SD 10.2), with 13 being female (54%). PJI characteristics and additional patient demographics are illustrated in Table I. The mean CRP level was significantly higher in the high-grade group compared to the low-grade and aseptic group (147 mg/l (SD 74) vs 11.4 mg/l (SD 12) vs 4.3 mg/l (SD 6), respectively; p < 0.001), as well as the mean leucocyte count in aspiration (150,876 per µl (SD 138,868) vs 15,370 per µl (SD 6,615) vs 766 per µl (SD 770), respectively; p = 0.004). The mean interval between primary arthroplasty and revision surgery did not change significantly between groups (aseptic: 10.1 years (SD 7.4); low-grade: 4.0 years (SD 5.2); high-grade: 8.4 years (SD 8.8); p = 0.251).

Table I.

Patient demographics and periprosthetic joint infection characteristics.

Sex Age at revision surgery, yrs Years between PA and RS Side Smoking Tissue cultures at the time of RS Sonification microbiology CRP, mg/l Histopathology Surgery for PJI Aspiration/leucocytes, 1 /nl Aspiration/PMN, % Aspiration/microbiology
Aseptic
Female 85 22.8 R No Negative Negative 5.5 Type 1 Stem exchange 3,146 55 Negative
Female 57 12.8 R No Negative Negative 0.7 Type 1 One-stage exchange N/A N/A Negative
Female 68 16.8 R Yes Negative Negative 1.0 Type 1 Head/inlay exchange 807 10 Negative
Female 88 8.8 L No Negative Negative 18.7 Type 1 Acetabular component exchange 457 55 Negative
Female 81 6.9 R No Negative Negative 3.8 Type 1 Acetabular component exchange 67 28 Negative
Female 61 1.0 L No Negative Negative 0.6 N/A Stem exchange 1,683 57 Negative
Female 74 9.9 R No Negative Negative 2.2 Type 1 Stem exchange 394 21 Negative
Male 72 1.7 L No Negative Negative 1.9 Type 1 One-stage exchange 1,957 20 Negative
Low-grade
Female 70 1.4 L Yes Staph. lugdonensis, Staph. hominis Staph. lugdunensis, Staph. hominis 6.5 Type 3 One-stage exchange 15,767 86 Staph. lugdonensis
Male 59 1.1 L Yes MRSE MRSE 8.2 Type 4 One-stage exchange 16,662 95 MSSE
Male 73 3.4 L No Negative Finegoldia magna 14.0 Type 4 Explantation 11,680 91 Negative
Male 66 16.51 L Yes Negative Negative 40.1 Type 3 Explantation 7,817 82 Negative
Female 62 0.7 R No MSSE, Staph. hominis MSSE 5.7 Type 1 One-stage exchange N/A N/A MSSE
Male 61 4.5 R No MRSE MRSE 13.2 N/A Explantation 22,020 83 MRSE
Male 63 2.3 L No Negative MSSE 0.6 Type 4 One-stage exchange 25,240 24 Negative
Male 63 2.4 L No Negative Staph. capitis 3.4 Type 2 One-stage exchange 8,406 70 Staph. capitis
High-grade
Female 65 13.9 R No Negative Negative 143.5 Type 3 DAIR 36,580 97 Haemophilus parainfluenzae
Male 54 1.9 L No Strept. dysgalactiae Strept. dysgalactiae 150.8 Type 2 Explantation N/A N/A N/A
Male 66 1.2 R No Negative Negative 274.0 Type 2 Explantation 9,930 90 Negative
Female 75 0.9 L No Candida auris Negative 66.2 Type 2 DAIR N/A N/A Candida auris
Female 62 24.3 R No Serratia marcescens Serratia marcescens 55.0 Type 3 Explantation 147,020 73 Serratia marcescens
Male 87 0.1 L No Proteus mirabilis Proteus mirabilis 194.4 Type 2 DAIR 207,530 86 Proteus mirabilis
Male 89 14.6 R No Strept. angiosus Negative 95.4 Type 1 Explantation 353,320 N/A Strept. angiosus
Female 75 10.6 L No MRSE MRSE 198.6 Type 2 DAIR N/A N/A N/A
  1. Strept; streptococcus; DAIR, debridement, antibiotics and implant retention; MRSA, methicillin-resistant Staphylococcus aureus; MRSE, methicillin-resistant Staphylococcus epidermidis; MSSE, methicillin-susceptible Staphylococcus epidermidis; N/A, not available; PA, primary arthroplasty; PJI, periprosthetic joint infection; PMN, polymorphonuclear leucocyte; RS, revision surgery; Staph, staphylococcus.

Circulating miRNA expression profiles

Hsa-miR-21-3p, hsa-miR-1290, and hsa-miR-4488 expression levels were significantly higher in high-grade group compared to the aseptic group (FDR < 0.05) (Figure 1a, Table II). However, results for hsa-miR-130a-3p and hsa-miR-451a also indicated repression of specific miRNA species with high-grade infections compared to aseptic controls (FDR < 0.05) (Figure 1b, Table II). Comparing the high-grade to the low-grade group, we observed lower expression levels of hsa-miR-26a-5p, hsa-miR-26b-5p, hsa-miR-148b-5p, hsa-miR-301a-3p, hsa-miR-451a, and hsa-miR-454-3p in comparison to the low-grade group, whereas a significant upregulation of miRNA could only be observed in hsa-miR-1260a ( p ≤ 0.001) (Figures 2a and 2b). No significant differences were discovered between the low-grade and aseptic groups.

Fig. 1 
            Scatterplots depicting the levels of a) upregulated and b) downregulated micro RNAs (miRNAs) in plasma samples of high-grade (green) vs aseptic (red) group. CPM, counts per million.

Fig. 1

Scatterplots depicting the levels of a) upregulated and b) downregulated micro RNAs (miRNAs) in plasma samples of high-grade (green) vs aseptic (red) group. CPM, counts per million.

Table II.

Up- and downregulated micro RNAs in the plasma samples of the high-grade group in comparison to the aseptic group.

miRNA log2FC logCPM F p-value FDR
hsa-miR-1290 2.6 9.5 18 < 0.001 0.043
hsa-miR-451a -0.88 19 15 < 0.001 0.043
hsa-miR-21-3p 0.9 7.1 14 0.001 0.043
hsa-miR-130a-3p -0.81 8.9 14 0.001 0.043
hsa-miR-4488 2.6 7 13 0.002 0.049
  1. CPM, counts per million; FDR, false discovery rate.

Fig. 2 
            Scatterplots depicting the levels of a) upregulated and b) downregulated micro RNAs (miRNAs) in plasma samples of high- (red) vs low-grade (green) group. CPM, counts per million.

Fig. 2

Scatterplots depicting the levels of a) upregulated and b) downregulated micro RNAs (miRNAs) in plasma samples of high- (red) vs low-grade (green) group. CPM, counts per million.

Circulating miRNAs in correlation with serum CRP levels

A significant correlation existed between serum CRP levels and circulating miRNA dysregulation (Spearman rank sum test adjusted for multiple testing, p.adj < 0.05). In total, 76 different miRNAs in plasma showed a significant correlation with serum CRP levels, of which 75 showed a positive association while only hsa-miR-451a showed a negative association (p.adj = 0.03, r = −0.58), indicating that patients with higher CRP levels had more upregulation of these circulating miRNAs. Circulating miRNAs with significant positive association to CRP are illustrated in Table III.

Table III.

Top ten circulating micro RNAs with significant association to CRP levels.

miRNA Estimate (r) p-value p.adj.
hsa-miR-331-5 p 0.7675 < 0.001 0.002
hsa-miR-589-5 p 0.7299 < 0.001 0.004
hsa-miR-584-5p 0.7286 < 0.001 0.004
hsa-miR-502-3p 0.7182 < 0.001 0.004
hsa-miR-1306-5p 0.7091 < 0.001 0.005
hsa-miR-362-3p 0.6909 < 0.001 0.007
hsa-miR-3605-3p 0.6831 < 0.001 0.008
hsa-miR-454-5p 0.6792 0.001 0.009
hsa-miR-542-3p 0.6792 0.001 0.009
hsa-miR-424-3p 0.6779 0.001 0.009
  1. miRNA, micro RNA.

miRNA in human tissue samples

In comparison to the aseptic group, 23 different miRNAs were significantly higher in high-grade group (hsa-miR-154-3p, hsa-miR-224-5p, hsa-miR-299-5p, hsa-miR-329-3p, hsa-miR-370-5p, hsa-miR-376a-2-5p, hsa-miR-376a-5p, hsa-miR-376c-3p, hsa-miR-337-3p, hsa-miR-369-3p, hsa-miR-376a-3p, hsa-miR-377-3p, hsa-miR-382-3p, hsa-miR-424-5p, hsa-miR-485-3p, hsa-miR-487a-3p, hsa-miR-493-5p, hsa-miR-495-3p, hsa-miR-542-3p, hsa-miR-542-5p, hsa-miR-543, hsa-miR-656-3p and hsa-miR-1185-5p; p < 0.05). Only the top 12 miRNA upregulations by log2FC are illustrated in Figure 3a. For hsa-miR-2110 and hsa-miR-3200-3p, expression levels were significantly lower (p ≤ 0.001) (Figure 3b). The dysregulated miRNAs in the tissue samples of the high-grade group compared to the aseptic group are illustrated in Table IV. No significant differences were demonstrated in miRNA expression between high- and low-grade groups, nor between low-grade and aseptic groups.

Fig. 3 
            Scatterplots depicting the levels of a) upregulated (top 12 by logFC) and b) downregulated micro RNAs (miRNAs) in tissue samples of high-grade (green) vs aseptic (red) group. CPM, counts per million.

Fig. 3

Scatterplots depicting the levels of a) upregulated (top 12 by logFC) and b) downregulated micro RNAs (miRNAs) in tissue samples of high-grade (green) vs aseptic (red) group. CPM, counts per million.

Table IV.

Top ten dysregulated micro RNAs in the tissue samples of the high-grade group in comparison to the aseptic group.

miRNA log2FC logCPM F p-value FDR
hsa-miR-376a-2-5p 2 2.9 29 < 0.001 0.014
hsa-miR-1185-5p 1.4 4.4 28 < 0.001 0.014
hsa-miR-337-3p 1.6 4.5 26 < 0.001 0.014
hsa-miR-299-5p 1.9 4.6 25 < 0.001 0.014
hsa-miR-377-3p 1.5 6.3 21 < 0.001 0.017
hsa-miR-224-5p 2.1 8.3 21 < 0.001 0.017
hsa-miR-2110 -1.7 4.9 20 < 0.001 0.017
hsa-miR-329-3p 1.3 3 19 < 0.001 0.017
hsa-miR-542-5p 2.1 4.1 19 < 0.001 0.017
hsa-miR-376a-5p 1.4 4.6 19 < 0.001 0.017
  1. CPM, counts per million; FDR, false discovery rate; miRNA, micro RNA.

Different miRNA expression in plasma and tissues of patients

The present study identified specific miRNAs that were differently expressed in both plasma and tissue samples. The first heatmap (Figure 4a) shows only the top 50 miRNAs. Overall, 341 miRNAs were detected in plasma and tissues (Figure 4b).

Fig. 4 
            a) This heatmap is for only the top 50 micro RNAs (miRNAs) (based on coefficient of variation (CV%)). An additional filter was introduced to increase the robustness: only miRNAs that showed reads assigned per million (RPM) in at least 1 /n (groups) percent of samples (e.g. with four groups, the miRNA has to have an RPM value above five in at least 25% of the samples). This removed miRNAs that had a high CV, but were only expressed in a too small amount of samples to have any statistical significance or biological relevance. b) A total of 341 miRNAs were shown in the other heatmap, based on the same filters described for the top 50 miRNAs. On the x-axis, plasma and tissue samples are depicted, whereas on the y-axis, the detected miRNAs are illustrated. On the top of the heatmaps, scale bar with plasma (red) and tissue (green) sections is demonstrated.

Fig. 4

a) This heatmap is for only the top 50 micro RNAs (miRNAs) (based on coefficient of variation (CV%)). An additional filter was introduced to increase the robustness: only miRNAs that showed reads assigned per million (RPM) in at least 1 /n (groups) percent of samples (e.g. with four groups, the miRNA has to have an RPM value above five in at least 25% of the samples). This removed miRNAs that had a high CV, but were only expressed in a too small amount of samples to have any statistical significance or biological relevance. b) A total of 341 miRNAs were shown in the other heatmap, based on the same filters described for the top 50 miRNAs. On the x-axis, plasma and tissue samples are depicted, whereas on the y-axis, the detected miRNAs are illustrated. On the top of the heatmaps, scale bar with plasma (red) and tissue (green) sections is demonstrated.

Discussion

To our knowledge, this study represents the first evaluation of miRNA profiling in PJI diagnostics. Currently, approximately 2,600 different miRNAs have been discovered in the human genome, with approximately 2,000 of them identified in the bloodstream circulation.40 From this pool of miRNAs, we identified the upregulation of hsa-miR-21-3p, hsa-miR-1260a, hsa-miR-1290, and hsa-miR-4488 in the high-grade group, along with downregulation of hsa-miR-26a-5p, hsa-miR-26b-5p, hsa-miR-130a-3p, hsa-miR-148b-5p, hsa-miR-301a-3p, hsa-miR-451a, and hsa-miR-454-3p compared to the aseptic and low-grade groups. Some of these miRNA dysregulations have been described in other contexts in recent literature. For instance, miR-21, well established as an oncogenic miRNA,41 regulates phagocytosis during infection, potentially limiting the availability of the intracellular niche of pathogens like Listeria monocytogenes.42 Hsa-miR-130a is relevant in the colonization of intestinal mucosa in Crohn’s disease patients, facilitating the intracellular replication of adherent–invasive Escherichia coli.43,44 Hsa-miR-451, one of the miRNAs controlling the NF-kB pathway, is upregulated during chlamydia infection.45 However, hsa-miR-451-upregulation has also been associated with haemolysis, a potential confounder of circulating miRNA data.46,47 Other miRNA dysregulations identified in this study were demonstrated for the first time in the literature in the context of a bacterial infection. This suggests a crucial step in establishing miRNA signatures as potential novel diagnostical tools and prognostic serum biomarkers for hip PJI.

In the present study, high-grade PJI showed significantly higher expression levels for 23 different miRNAs and lower expression levels for hsa-miR-2110 and hsa-miR-3200-3p in tissue samples compared to patients with no infection. To our knowledge, these miRNA signatures were described for the first time in accordance with bacterial infection, which is a crucial first step for tissue miRNA profiling in the diagnosis of hip PJI.

The present study revealed that miRNA profiles expressed in tissue vary from those in plasma. This incongruence between the miRNA expression profiles suggests that miRNAs do not simply “leak” from infected tissue, as the plasma profile does not simply reflect the tissue profile. This observation is highly indicative of differential pathological processes occurring between plasma and tissue cells. The characteristic distribution of extracellular miRNAs suggests that they may constitute a specific biological response. This unexpected level of complexity needs to be considered with regard to the use of serum biomarkers with miRNA profiling of plasma and tissues for diagnosing hip PJI.

Among the miRNAs circulating in the bloodstream and correlating with CRP serum levels, the let-7 family is one of the most extensively researched miRNAs in relation to infection.24 It is associated with cell differentiation and development,48 and functions as an actor of the acute innate immune response.22 The present study detected an upregulation of the let-7 family in correlation to CRP, except for hsa-let-7d-5p, which was downregulated upon CRP increase. Moreover, some of the detected upregulated miRNAs in correlation with CRP increase such as hsa-miR-127, hsa-miR-193, hsa-miR-212-3p, hsa-miR-365, and hsa-miR-432-5p were already described in literature in the context of bacterial infection.49-51

MiRNAs function as important regulators in bacterial infection, suggesting their great application potential as novel therapeutic targets to cure bacterial infections. Although there are no bacterial infection-related miRNA-based drugs investigated in clinical research to date, miRNAs still represent a promising approach for future therapies or as immune modulators against invading pathogens. It is important to note that detected regulations of miRNAs in the high-grade group compared to the aseptic group were irrespective of bacterial pathogens. For this reason, it can be concluded that miRNA expression during infection is not specific to the bacterial pathogen but rather to the reaction of the host against it. Moreover, individual genes are usually not controlled by a single miRNA and the ramifications of the identified miRNA species on the severity of PJI need to be further evaluated. The regulation of the miRNAs illustrated in this study may influence different molecular biological processes in the development and progression of PJI, representing potential targets in therapy.

This study has several limitations. First, our study contains a small sample size, which is possibly why we could not identify any significant difference in miRNA dysregulation between aseptic and low-grade groups in plasma and tissue samples, and between low- and high-grade groups in tissue samples. Second, the present study is neither randomized nor blinded, inevitably introducing selection and evaluation biases. Additional double-blind randomized studies, including a larger cohort number, are required to further substantiate these findings. Moreover, low-grade infections may not have the virulence to induce significant differences in miRNA expression. Nevertheless, we were able to identify circulating miRNA signatures and local expression profiles in tissue and blood samples, correlating with the severity of hip PJI for the first time. Third, including another group of patients with primary septic arthritis of the hip would have been beneficial to demonstrate the specificity of the identified miRNAs for implant-related infections. Lastly, there is no consensus on the the PJI definition criteria. None of the existing definition criteria are uniformly accepted and may largely influence the clinical course of the patients as well as the outcomes. However, the EBJIS criteria demonstrate a high sensitivity for low-grade PJIs as reported in literature.12,33

High-grade hip PJI correlates with individual plasma and tissue miRNA signatures compared to aseptic patients with the revision of hip arthroplasty. The present study illustrates different miRNA expressions, contributing to the pool of miRNA signatures as potential novel diagnostic and prognostic biomarkers for hip PJI.


Correspondence should be sent to Alp Paksoy. E-mail:

References

1. Kamath AF , Ong KL , Lau E , et al. Quantifying the burden of revision total joint arthroplasty for periprosthetic infection . J Arthroplasty . 2015 ; 30 ( 9 ): 1492 1497 . Crossref PubMed Google Scholar

2. Engesæter LB , Dale H , Schrama JC , Hallan G , Lie SA . Surgical procedures in the treatment of 784 infected THAs reported to the Norwegian Arthroplasty Register . Acta Orthop . 2011 ; 82 ( 5 ): 530 537 . Crossref PubMed Google Scholar

3. Fink B . Revision of late periprosthetic infections of total hip endoprostheses: pros and cons of different concepts . Int J Med Sci . 2009 ; 6 ( 5 ): 287 295 . Crossref PubMed Google Scholar

4. Kurtz S , Ong K , Lau E , Mowat F , Halpern M . Projections of primary and revision hip and knee arthroplasty in the United States from 2005 to 2030 . J Bone Joint Surg Am . 2007 ; 89-A ( 4 ): 780 785 . Crossref PubMed Google Scholar

5. Sukeik M , Patel S , Haddad FS . Aggressive early débridement for treatment of acutely infected cemented total hip arthroplasty . Clin Orthop Relat Res . 2012 ; 470 ( 11 ): 3164 3170 . Crossref PubMed Google Scholar

6. No authors listed . NIH consensus conference: Total hip replacement. NIH Consensus Development Panel on Total Hip Replacement . JAMA . 1995 ; 273 ( 24 ): 1950 1956 . Crossref PubMed Google Scholar

7. Akgün D , Müller M , Perka C , Winkler T . The serum level of C-reactive protein alone cannot be used for the diagnosis of prosthetic joint infections, especially in those caused by organisms of low virulence . Bone Joint J . 2018 ; 100-B ( 11 ): 1482 1486 . Crossref PubMed Google Scholar

8. Zimmerli W , Trampuz A , Ochsner PE . Prosthetic-joint infections . N Engl J Med . 2004 ; 351 ( 16 ): 1645 1654 . Crossref PubMed Google Scholar

9. Renz N , Mudrovcic S , Perka C , Trampuz A . Orthopedic implant-associated infections caused by Cutibacterium spp. - a remaining diagnostic challenge . PLoS One . 2018 ; 13 ( 8 ): e0202639 . Crossref PubMed Google Scholar

10. Akgün D , Bürger J , Pumberger M , Putzier M . C-reactive protein misdiagnoses delayed postoperative spinal implant infections in patients with low-virulent microorganisms . Eur Spine J . 2019 ; 28 ( 12 ): 2990 2995 . Crossref PubMed Google Scholar

11. Piper KE , Fernandez-Sampedro M , Steckelberg KE , et al. C-reactive protein, erythrocyte sedimentation rate and orthopedic implant infection . PLoS One . 2010 ; 5 ( 2 ): e9358 . Crossref PubMed Google Scholar

12. Pérez-Prieto D , Portillo ME , Puig-Verdié L , et al. C-reactive protein may misdiagnose prosthetic joint infections, particularly chronic and low-grade infections . Int Orthop . 2017 ; 41 ( 7 ): 1315 1319 . Crossref PubMed Google Scholar

13. Kheir MM , Tan TL , Shohat N , Foltz C , Parvizi J . Routine diagnostic tests for periprosthetic joint infection demonstrate a high false-negative rate and are influenced by the infecting organism . J Bone Joint Surg Am . 2018 ; 100-A ( 23 ): 2057 2065 . Crossref PubMed Google Scholar

14. Hao L , Wen P , Song W , et al. Direct detection and identification of periprosthetic joint infection pathogens by metagenomic next-generation sequencing . Sci Rep . 2023 ; 13 ( 1 ): 7897 . Crossref PubMed Google Scholar

15. Tang Y , Zhao D , Wang S , Yi Q , Xia Y , Geng B . Diagnostic value of next-generation sequencing in periprosthetic joint infection: a systematic review . Orthop Surg . 2022 ; 14 ( 2 ): 190 198 . Crossref PubMed Google Scholar

16. Gehrke T , Lausmann C , Citak M , Bonanzinga T , Frommelt L , Zahar A . The accuracy of the alpha defensin lateral flow device for diagnosis of periprosthetic joint infection . J Bone Joint Surg Am . 2018 ; 100-A ( 1 ): 42 48 . Crossref PubMed Google Scholar

17. Eulalio A , Schulte L , Vogel J . The mammalian microRNA response to bacterial infections . RNA Biol . 2012 ; 9 ( 6 ): 742 750 . Crossref PubMed Google Scholar

18. Das K , Garnica O , Dhandayuthapani S . Modulation of host miRNAs by intracellular bacterial pathogens . Front Cell Infect Microbiol . 2016 ; 6 : 79 . Crossref PubMed Google Scholar

19. Silwal P , Kim YS , Basu J , Jo E-K . The roles of microRNAs in regulation of autophagy during bacterial infection . Semin Cell Dev Biol . 2020 ; 101 : 51 58 . Crossref PubMed Google Scholar

20. Zhou X , Li X , Wu M . miRNAs reshape immunity and inflammatory responses in bacterial infection . Signal Transduct Target Ther . 2018 ; 3 ( 1 ): 14 . Crossref PubMed Google Scholar

21. Fang Y , Chen H , Hu Y , et al. Burkholderia pseudomallei-derived miR-3473 enhances NF-κB via targeting TRAF3 and is associated with different inflammatory responses compared to Burkholderia thailandensis in murine macrophages . BMC Microbiol . 2016 ; 16 ( 1 ): 283 . Crossref PubMed Google Scholar

22. Staedel C , Darfeuille F . MicroRNAs and bacterial infection . Cell Microbiol . 2013 ; 15 ( 9 ): 1496 1507 . Crossref PubMed Google Scholar

23. Maudet C , Mano M , Eulalio A . MicroRNAs in the interaction between host and bacterial pathogens . FEBS Lett . 2014 ; 588 ( 22 ): 4140 4147 . Crossref PubMed Google Scholar

24. Aguilar C , Mano M , Eulalio A . MicroRNAs at the host-bacteria interface: host defense or bacterial offense . Trends Microbiol . 2019 ; 27 ( 3 ): 206 218 . Crossref PubMed Google Scholar

25. Ouellet DL , Plante I , Barat C , Tremblay MJ , Provost P . Emergence of a complex relationship between HIV-1 and the microRNA pathway . Methods Mol Biol . 2009 ; 487 : 415 433 . Crossref PubMed Google Scholar

26. Berkhout B , Liu YP . Towards improved shRNA and miRNA reagents as inhibitors of HIV-1 replication . Future Microbiol . 2014 ; 9 ( 4 ): 561 571 . Crossref PubMed Google Scholar

27. Bueno MJ , Pérez de Castro I , Malumbres M . Control of cell proliferation pathways by microRNAs . Cell Cycle . 2008 ; 7 ( 20 ): 3143 3148 . Crossref PubMed Google Scholar

28. Shenoy A , Blelloch RH . Regulation of microRNA function in somatic stem cell proliferation and differentiation . Nat Rev Mol Cell Biol . 2014 ; 15 ( 9 ): 565 576 . Crossref PubMed Google Scholar

29. Jovanovic M , Hengartner MO . miRNAs and apoptosis: RNAs to die for . Oncogene . 2006 ; 25 ( 46 ): 6176 6187 . Crossref PubMed Google Scholar

30. Benz F , Roy S , Trautwein C , Roderburg C , Luedde T . Circulating microRNAs as biomarkers for sepsis . Int J Mol Sci . 2016 ; 17 ( 1 ): 78 . Crossref PubMed Google Scholar

31. Tribolet L , Kerr E , Cowled C , et al. MicroRNA biomarkers for infectious diseases: from basic research to biosensing . Front Microbiol . 2020 ; 11 : 1197 . Crossref PubMed Google Scholar

32. Agosile O , Benson U , Ugochukwu O , Okei N , Fajobi S , Akachukwu S , et al. MicroRNA and Immune Response to Viral, Bacterial and Fungal Infections . 2022 . Google Scholar

33. McNally M , Sousa R , Wouthuyzen-Bakker M , et al. The EBJIS definition of periprosthetic joint infection . Bone Joint J . 2021 ; 103-B ( 1 ): 18 25 . Crossref PubMed Google Scholar

34. Krenn V , Morawietz L , Perino G , et al. Revised histopathological consensus classification of joint implant related pathology . Pathol Res Pract . 2014 ; 210 ( 12 ): 779 786 . Crossref PubMed Google Scholar

35. Morawietz L , Classen R-A , Schröder JH , et al. Proposal for a histopathological consensus classification of the periprosthetic interface membrane . J Clin Pathol . 2006 ; 59 ( 6 ): 591 597 . Crossref PubMed Google Scholar

36. Morawietz L , Gehrke T , Classen R , Barden B , Otto M , Hansen T , et al. Vorschlag für die Konsensus-Klassifikation der periprothetischen Membran gelockerter Hüft- und Knieprothesen . Der Pathologe . 2004 ; 25 : 375 - 84 . Google Scholar

37. Klein MJ , Bonar SF , Freemont T , et al. Non-Neoplastic Diseases of Bones and Joints . American Registry of Pathology , 2011 . Crossref Google Scholar

38. Khamina K , Diendorfer AB , Skalicky S , et al. A microRNA next-generation-sequencing discovery assay (miND) for genome-scale analysis and absolute quantitation of circulating microRNA biomarkers . Int J Mol Sci . 2022 ; 23 ( 3 ): 1226 . Crossref PubMed Google Scholar

39. Diendorfer A , Khamina K , Pultar M , Hackl M . MiND (mirna NGS discovery pipeline): a small RNA-seq analysis pipeline and report generator for microrna biomarker discovery studies . F1000Research . 2022 ; 11 : 233 . Google Scholar

40. Juzenas S , Venkatesh G , Hübenthal M , et al. A comprehensive, cell specific microRNA catalogue of human peripheral blood . Nucleic Acids Res . 2017 ; 45 ( 16 ): 9290 9301 . Crossref PubMed Google Scholar

41. Kumarswamy R , Volkmann I , Thum T . Regulation and function of miRNA-21 in health and disease . RNA Biol . 2011 ; 8 ( 5 ): 706 713 . Crossref PubMed Google Scholar

42. Johnston DGW , Kearney J , Zasłona Z , Williams MA , O’Neill LAJ , Corr SC . MicroRNA-21 limits uptake of Listeria monocytogenes by macrophages to reduce the intracellular niche and control infection . Front Cell Infect Microbiol . 2017 ; 7 : 201 . Crossref PubMed Google Scholar

43. Larabi A , Dalmasso G , Delmas J , Barnich N , Nguyen HTT . Exosomes transfer miRNAs from cell-to-cell to inhibit autophagy during infection with Crohn’s disease-associated adherent-invasive E. coli Gut Microbes . 2020 ; 11 ( 6 ): 1677 1694 . Crossref PubMed Google Scholar

44. Martinez-Medina M , Aldeguer X , Lopez-Siles M , et al. Molecular diversity of Escherichia coli in the human gut: new ecological evidence supporting the role of adherent-invasive E. coli (AIEC) in Crohnʼs disease . Inflamm Bowel Dis . 2009 ; 15 ( 6 ): 872 882 . Crossref PubMed Google Scholar

45. Igietseme JU , Omosun Y , Stuchlik O , et al. Role of epithelial-mesenchyme transition in chlamydia pathogenesis . PLoS One . 2015 ; 10 ( 12 ): e0145198 . Crossref PubMed Google Scholar

46. Kirschner MB , Kao SC , Edelman JJ , et al. Haemolysis during sample preparation alters microRNA content of plasma . PLoS One . 2011 ; 6 ( 9 ): e24145 . Crossref PubMed Google Scholar

47. Shkurnikov MYu , Knyazev EN , Fomicheva KA , et al. Analysis of plasma microRNA associated with hemolysis . Bull Exp Biol Med . 2016 ; 160 ( 6 ): 748 750 . Crossref PubMed Google Scholar

48. Roush S , Slack FJ . The let-7 family of microRNAs . Trends Cell Biol . 2008 ; 18 ( 10 ): 505 516 . Crossref PubMed Google Scholar

49. Lin Y , Zhang Y , Yu H , Tian R , Wang G , Li F . Identification of unique key genes and miRNAs in latent tuberculosis infection by network analysis . Mol Immunol . 2019 ; 112 : 103 114 . Crossref PubMed Google Scholar

50. Miotto P , Mwangoka G , Valente IC , et al. miRNA signatures in sera of patients with active pulmonary tuberculosis . PLoS One . 2013 ; 8 ( 11 ): e80149 . Crossref PubMed Google Scholar

51. Liu X , Mao Y , Kang Y , et al. MicroRNA-127 promotes anti-microbial host defense through restricting A20-mediated de-ubiquitination of STAT3 . iScience . 2020 ; 23 ( 1 ): 100763 . Crossref PubMed Google Scholar

Author contributions

A. Paksoy: Conceptualization, Methodology, Software, Validation, Formal analysis, Investigation, Data curation, Writing – original draft, Writing – review & editing, Visualization

S. Meller: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Resources, Data curation, Writing – original draft, Writing – review & editing, Supervision, Project administration, Funding acquisition

F. Schwotzer: Conceptualization, Methodology, Software, Validation, Formal analysis, Data curation, Writing – original draft, Writing – review & editing, Visualization

P. Moroder: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Resources, Data curation, Writing –original draft, Writing – review & editing, Visualization, Supervision, Project administration, Funding acquisition

A. Trampuz: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Data curation, Writing – original draft, Supervision, Project administration

J-P. Imiolczyk: Methodology, Software, Validation, Investigation, Data curation, Writing – original draft, Writing – review & editing

C. Perka: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Resources, Data curation, Writing – original draft, Writing – review & editing, Supervision, Project administration, Funding acquisition

M. Hackl: Conceptualization, Methodology, Validation, Data curation, Writing – original draft

F. Plachel: Conceptualization, Methodology, Validation, Data curation, Writing – original draft

D. Akgün: Conceptualization, Methodology, Software, Validation, Formal analysis, Investigation, Resources, Data curation, Writing – original draft, Writing – review & editing, Visualization, Supervision, Project administration, Funding acquisition

Funding statement

The authors disclose receipt of the following financial or material support for the research, authorship, and/or publication of this article: Stiftung Endoprothetik (Deutsche Gesellschaft für Endoprothetik).

ICMJE COI statement

The authors have no conflicts of interest to disclose. D. Akgün reports that this study was supported by the Stiftung Endoprothetik.

Data sharing

All data generated or analyzed during this study are included in the published article and/or in the supplementary material.

Ethical review statement

The study protocol was reviewed and approved by the institutional ethics committee prior to commencement (EA1/283/19).

Open access funding

The open access fee for this article was funded by the Charité University Hospital.

Supplementary material

Detailed description of the data analysis.

© 2024 Paksoy et al. Open Access This article is distributed under the terms of the Creative Commons Attributions (CC BY 4.0) licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original author and source are credited.