Skip to main content

Open Access 14.05.2024 | Original Article

Correlation of serum DKK1 level with skeletal phenotype in children with osteogenesis imperfecta

verfasst von: Y. Wang, J. Hu, L. Sun, B. Zhou, X. Lin, Q. Zhang, O. Wang, Y. Jiang, W. Xia, X. Xing, M. Li

Erschienen in: Journal of Endocrinological Investigation

Abstract

Purpose

We aim to detect serum DKK1 level of pediatric patients with OI and to analyze its relationship with the genotype and phenotype of OI patients.

Methods

A cohort of pediatric OI patients and age-matched healthy children were enrolled. Serum levels of DKK1 and bone turnover biomarkers were measured by enzyme-linked immunosorbent assay. Bone mineral density (BMD) was measured by Dual-energy X-ray absorptiometry. Pathogenic mutations of OI were detected by next-generation sequencing and confirmed by Sanger sequencing.

Results

A total of 62 OI children with mean age of 9.50 (4.86, 12.00) years and 29 healthy children were included in this study. The serum DKK1 concentration in OI children was significantly higher than that in healthy children [5.20 (4.54, 6.32) and 4.08 (3.59, 4.92) ng/mL, P < 0.001]. The serum DKK1 concentration in OI children was negatively correlated with height (r = − 0.282), height Z score (r = − 0.292), ALP concentration (r = − 0.304), lumbar BMD (r = − 0.276), BMD Z score of the lumbar spine and femoral neck (r = − 0.32; r = − 0.27) (all P < 0.05). No significant difference in serum DKK1 concentration was found between OI patients with and without vertebral compression fractures. In patients with spinal deformity (22/62), serum DKK1 concentration was positively correlated with SDI (r = 0.480, P < 0.05). No significant correlation was observed between serum DKK1 concentration and the annual incidence of peripheral fractures, genotype and types of collagen changes in OI children.

Conclusion

The serum DKK1 level was not only significantly elevated in OI children, but also closely correlated to their skeletal phenotype, suggesting that DKK1 may become a new biomarker and a potential therapeutic target of OI.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s40618-024-02380-9.

Publisher's Note

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

Introduction

Osteogenesis imperfecta (OI) is a rare monogenic hereditary osteopathy with an incidence of 1 in 15,000 to 20,000 live births, characterized by bone fragility and multiple bone fractures [1]. OI is caused by alteration in synthesis and post-translational modification of type I collagen due to multiple genetic mutations [2, 3]. Recent studies have shown that the severity of OI phenotype is not only related to abnormal type I collagen metabolism, but also closely linked to changes in osteoblast activity [4, 5], of which the mechanism is not yet clear.
As is well known, WNT/β-catenin pathway plays an important role in regulation of differentiation and activity of osteoblasts. WNT1 mutation can lead to severe OI through leading to inactivation of WNT pathway and osteoblasts, suggesting that WNT pathway may be involved in the pathological processes of OI [6, 7]. Previous studies indicated that the natural inhibitors of the WNT pathway, including sclerostin, frizzled protein, and Dickkopf-1 (DKK1), produced marked effects on the differentiation, maturation and activity of osteoblasts [8]. DKK1 can block the binding of WNT to the specific cell surface receptors low-density lipoprotein receptor-related protein 5/6 (LRP5/6), and reduce the level of β-catenin in osteoblasts, then reducing the differentiation and maturation of osteoblasts [9]. However, it is unclear whether there is a change in serum DKK1 level in OI patients and whether it is related to OI phenotype.
Therefore, we detect the serum DKK1 level in pediatric patients with OI and analyze its relationship with BMD, fracture incidence, bone turnover makers, and the pathogenic genotype of OI, and to determine whether DKK1 has value as a novel biomarker and therapeutic target of OI.

Methods

Subjects

Patients less than 18 years old with OI were recruited from the Department of Endocrinology, Peking Union Medical College Hospital (PUMCH), from April 2017 to October 2023. The inclusion criteria of OI patients were as follows: (1) a history of at least one fracture under minor trauma during childhood and an age- and gender-adjusted BMD Z-score less than -2.0 at lumbar spine (LS) or proximal femur before any anti-osteoporosis therapy; (2) presence of blue sclera or dentinogenesis imperfecta and a family history of OI [10, 11]. The exclusion criteria were as follows: with other genetic or metabolic bone diseases, with other disease that could affect bone metabolism, ongoing treatment with glucocorticoids, anti-epileptic drugs, bisphosphonates, denosumab, teriparatide, etc., and with liver or kidney dysfunction.
Age-matched healthy children who underwent physical examinations at PUMCH were included as control. This study was approved by the Scientific Ethics Committee of PUMCH (JS-3545D), and informed consents were obtained from legal guardian of each OI patient and healthy children.

Phenotype assessment

The following phenotypic information of OI patients was collected: age of OI onset, age of confirmed OI, frequency and sites of bone fracture, skeletal malformations and extra-skeletal manifestations, including blue sclera, dentinogenesis imperfecta, hearing loss, joint ligament laxity, and muscle atrophy. Height and weight were measured using a Harpenden measuring instrument (Seritex, Inc., East Rutherford, NJ, USA). For patient who was unable to stand, body length in the supine position was measured. Height and weight Z scores for OI patients at different ages and gender were calculated according to the normal reference values for Chinese children [12].
Clinical fractures included nonvertebral fractures and symptomatic vertebral fractures, which were reported by the patients or their legal guardians and confirmed by X-ray films. A semiquantitative assessment of vertebral compression fracture (VCFs) was performed by radiologists at PUMCH using Genant classification [13]. The semiquantitative Spinal Deformity Index (SDI) provides a comprehensive evaluation of spinal fracture status, considering both the number and severity of fractures. Each vertebra is visually graded on a scale of 0 to 3, representing no fracture, mild, moderate, or severe fracture, and SDI is calculated by summing these grades across all vertebrae from T4 to L4 [14]. Scoliosis was confirmed by posterior-anterior radiographs and defined as a Cobb angle higher than 10° [15]. The annual incidence of peripheral fractures was calculated by dividing the total number of peripheral fractures by the duration of disease. Areal BMD at the lumbar spine (LS) 1–4, the femoral neck (FN) and the total hip was measured using dual-energy X-ray absorptiometry (DXA, GE Lunar Prodigy Advance, USA) and analyzed by software compatible with pediatric data. Calibration and quality checks were completed daily using the DXA equipment. Patients with vertebral compression fractures or significantly deformation were excluded from the lumbar BMD analysis. The BMD Z scores of the LS and FN of children and adolescents were calculated according to the normal BMD reference values for Asian children [16, 17].
The disease phenotype exhibits significant heterogeneity, including the mildest form (type I), the most severe form among surviving patients (type III), an intermediate form between type I and type III (type IV), and the unique type with interosseous membrane calcification of the forearm and/or hypertrophic callus (type V) [18, 19]. The perinatal lethality (type II) OI was not included in this study.

Determination of the serum DKK1 and biochemical marker concentration

Fasting blood samples of OI patients and healthy controls were obtained at 8:00–10:00 in the morning. The serum DKK1 concentration was measured by enzyme-linked immunosorbent assay (ELISA) (Cat. No. DKK100B, R&D systems, USA), which was completed by Key Laboratory of Endocrinology, National Health and Family Planning Commission, PUMCH. The minimum detection value was 0.948 pg/mL, the intra-assay coefficients of variation (CV) was 1.8–2.9%, and the inter-assay CV was 7.7–8.7%.
The serum concentrations of osteoprotegerin (OPG) and sclerostin were measured by enzyme-linked immunosorbent assay (ELISA) (Cat. No. SEA108Hu, Cloud-Clone Corp, China and Cat. No. BI-20472, BIOMEDICA, Austria). The minimum detection value of OPG and sclerostin were 0.059 ng/mL and 1.3 pmol/L, respectively. The intra-assay CV were ≤ 10%and ≤ 1% for OPG and sclerostin measurement, respectively. The inter-assay CVs were ≤ 12% and ≤ 5% for OPG and sclerostin detection, respectively.
The serum levels of calcium (Ca), phosphorus (P) and alkaline phosphatase (ALP, a bone formation marker) were measured using an automatic analyser (ADVIA 1800, Siemens, Germany). The serum levels of β-isomerized carboxy-telopeptide of type I collagen (β-CTX, a bone resorption marker), procollagen I N-terminal peptide (P1NP, a bone formation marker), 25-hydroxyvitamin D (25OHD), and intact parathyroid hormone (PTH) were assessed using an automated electrochemiluminescence system (E170, Roche Diagnostics, Switzerland). All the biochemical indicators were detected by clinical central laboratory of PUMCH.

Detection of pathogenic mutations in OI patients

Genomic DNA was extracted from peripheral leukocytes of OI patients using a DNA extraction kit (QIAamp DNA, Qiagen, Frankfurt, Germany), which was sequenced using targeted next-generation sequencing (NGS) (Illumina HiSeq2000 platform, Illumina, Inc., San Diego, CA, USA) [20]. The targeted NGS panel included all known candidate genes of OI, including COL1A1, COL1A2, IFITM5, SERPINF1, CRTAP, P3H1, PPIB, SERPINH1, FKBP10, PLOD2, BMP1, SP7, TMEM38B, WNT1, CREB3L1, SPARC, MBTPS2, P4HB, SEC24D and PLS3, and 708 other skeletal disease-associated candidate genes [21]. The pathogenicity of the detected variants was classified according to the 2015 guidelines of the American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) [22]. The pathogenic mutations identified by NGS were validated by polymerase chain reaction (PCR) and Sanger sequence (3730 DNA Analyser, Applied Biosystems, Foster City, CA, USA).
According to genetic patterns, OI patients were divided into autosomal dominant inheritance (AD) and non-AD groups. The AD group included patients carrying COL1A1, COL1A2, IFITM5, and P4HB mutations, and patients with other gene mutations were classified into the non-AD group. Based on different effects of pathogenic mutations on type I collagen metabolism, the mutations causing amino acid substitutions in the triple helix domain of COL1A1 or COL1A2 were classified as collagen structural defects, and nonsense mutations or frame-shift mutations in COL1A1 or COL1A2 that led to an early stop codon were classified as collagen protein reducers [20, 23]. Other mutations, such as splicing mutations, were not included because of the difficulty in predicting their effects on type I collagen metabolism.

Statistical analysis

The Shapiro‒Wilk test and Kolmogorov‒Smirnov test were used to determine whether the data fit a normal distribution. Normally distributed data were expressed as the mean ± standard deviation, abnormally distributed data were expressed as the median (quartiles), and count data were expressed as numbers. Normally distributed data were compared between two groups and among different subgroups with independent sample t-tests and analysis of variance (ANOVA), respectively. Abnormally distributed data were compared between two groups and among more than two groups using the Mann‒Whitney U test and a nonparametric test (Kruskal‒Wallis test), respectively. The chi-square test and Fisher’s exact test were used to compare categorical variables. To explore the correlation, Pearson correlation analysis was applied for normally distributed data, while Spearman correlation analysis was used for abnormally distributed data.
A two-tailed P value less than 0.05 was considered statistically significant. Statistical analysis was performed using SPSS software version 25.0 (SPSS, Inc., Chicago, IL, USA). Graphics were drawn using GraphPad Prism software 10.0 (GraphPad, San Diego, CA, USA).

Results

Basic characteristics of OI children

A total of 62 children with OI, with an average age of 9.50 (4.86, 12.00) years, were enrolled in this study. 29 age matched healthy children [8.00 (5.50, 10.00) years] were included in the study as normal controls (Table 1). There were 46 boys and 16 girls in OI group, and 13 boys and 16 girls in control group. There was a difference in gender ratio between the two groups (P < 0.01) (Table 1). The height of OI children was 128.78 ± 23.92 cm, similar to that of healthy children (129.98 ± 18.61 cm). The height Z score for OI children was -0.49 ± 1.38, which was lower than that for the healthy controls (0.48 ± 1.23, P < 0.01) (Table 1).
Table 1
Basic characteristics and serum DKK1 level of OI children and healthy controls
 
OI (n = 62)
Healthy control (n = 34)
P value
Reference range
Gender (male/female)
46/16
13/16
0.006
Age, years
9.50 (4.86, 12.00)
8.00 (5.50, 10.00)
0.397
Height, cm
128.78 ± 23.92
129.98 ± 18.61
0.832
Height Z score
− 0.49 ± 1.38
0.48 ± 1.23
0.005
Weight, kg
30.50 (16.75, 45.25)
25.00 (19.50, 31.00)
0.368
Weight Z score
− 0.06 (− 0.74, 1.01)
− 0.14 (− 0.80, 0.79)
0.700
Ca, mmol/L
2.48 ± 0.08
2.45 ± 0.08
0.206
2.13–2.70
P, mmol/L
1.69 ± 0.20
1.66 ± 0.16
0.550
0.95–2.65 [49]
ALP, U/L
319.15 ± 95.78
229.58 ± 66.30
0.000
42–390 [49]
ALT, U/L
13.50 (10.00, 20.00)
15.00 (10.00, 17.00)
0.823
7–40
Cr, μmol/L
36.50 (30.75, 40.00)
45.00 (33.00, 46.00)
0.003
45–84
PTH, pg/mL
24.15 (18.75, 35.60)
17.30 (15.00, 22.30)
0.000
15.0–65.0
P1NP, ng/mL
380.50 (273.75, 571.50)
423.00 (343.00, 531.00)
0.507
30.0–3000.0 [50]
β-CTX, ng/mL
1.14 (0.83, 1.43)
1.16 (0.97, 1.41)
0.655
0.40–3.30 [50]
25OHD, ng/mL
22.00 (16.45, 33.15)
23.50 (19.50, 28.65)
0.789
 > 30
OPG, ng/mL
0.64 (0.44, 1.05)
0.74 (0.52, 1.35)
0.254
Sclerostin, pmol/L
20.87 (14.94, 27.36)
30.41 (22.51,34.00)
0.001
DKK1, ng/mL
5.20 (4.54, 6.32)
4.08 (3.59, 4.92)
0.000
The results for normally distributed data were presented as the mean ± SD
Nonnormally distributed data were presented as medians (quartiles)
Bold numbers represent P < 0.05
OI osteogenesis imperfecta, Ca calcium, P phosphorus, ALP alkaline phosphatase, ALT glutamic-pyruvic transaminase, Cr creatinine, PTH parathyroid hormone, P1NP procollagen type 1 N-peptide, β-CTX β-C-terminal telopeptide of type 1 collagen, 25OHD 25-hydroxyvitamin D, OPG Osteoprotegerin
According to the Sillence classification, the children with OI were divided into the following groups: type I (34 patients, 55%), type III (12 patients, 19%), and type IV (16 patients, 26%) (Table 2). The annual incidence of peripheral fractures of children with types III and IV OI was higher than those of type I OI, with specific rates being 1.00 (1.00, 2.88) and 1.16 (0.85, 2.30) fractures per year in types III and IV OI versus 0.64(0.30, 1.00) fractures per year in type I OI (all P < 0.05) (Table 2). Additionally, the proportion of children with long bone malformations of types III and IV OI was significantly higher than that of type I OI children, accounting for 75% and 31% in types III and IV OI versus 3% in type I OI (all P < 0.05) (Table 2). Furthermore, the prevalence of scoliosis of type IV OI children (19%) was obviously higher than type I OI children (0%) (P < 0.05) (Table 2).
Table 2
The characteristics and serum DKK1 level of different clinical types of OI patients and healthy controls
 
OI type I (n = 34)
OI type III (n = 12)
OI type IV (n = 16)
Control (n = 29)
P value
Gender (male/female)
22/12
11/1j
13/3
13/16
0.013
Age, years
10.00 (7.00, 12.00)
4.75 (3.00, 12.50)
8.50 (4.25, 11.00)
8.00 (5.50, 10.00)
0.272
Height, cm
135.63 ± 20.90
117.96 ± 26.34
122.34 ± 24.73
129.98 ± 18.61
0.061
Height Z score
− 0.12 ± 1.00
− 0.91 ± 1.62j
− 0.95 ± 1.69f
0.48 ± 1.23
0.003
Weight, kg
35.00 (22.25, 46.25)
17.00 (13.63, 48.75)
26.00 (15.25, 40.63)
25.00 (19.50, 31.00)
0.205
Weight Z score
0.16 (-0.59, 1.04)
− 0.33 (− 0.75, 0.81)
0.06 (− 1.16, 1.18)
− 0.14 (− 0.80, 0.79)
0.756
Ca, mmol/L
2.48 ± 0.07
2.45 ± 0.06
2.50 ± 0.10
2.45 ± 0.08
0.221
P, mmol/L
1.72 ± 0.18
1.65 ± 0.23
1.65 ± 0.21
1.66 ± 0.16
0.586
ALP, U/L
319.32 ± 87.25e
295.42 ± 69.88
336.56 ± 127.30f
229.58 ± 66.30
0.000
ALT, U/L
13.50 (10.75, 195.00)
11.50 (10.00, 21.50)
19.00 (11.25, 20.00)
15.00 (10.00, 17.00)
0.445
Cr, μmol/L
37.50 (33.00, 43.00)
33.00 (27.50, 37.75)j
33.50 (27.50, 39.75)k
45.00 (33.00, 46.00)
0.007
PTH, pg/mL
27.95 (20.25, 37.10)b
20.20 (17.83, 34.18)
20.85 (17.75, 25.85)
17.30 (15.00, 22.30)
0.000
P1NP, ng/mL
403.00 (299.00, 577.00)
410.00 (295.00, 758.00)
280.00 (228.70, 506.93)
423.00 (343.00, 531.00)
0.248
β-CTX, ng/mL
1.23 (1.05, 1.62)a
1.11 (0.90, 1.30)
0.80 (0.66, 0.87)k
1.16 (0.97, 1.41)
0.000
25OHD, ng/mL
21.90 (17.25, 28.00)
26.20 (16.25, 58.70)
21.95 (15.70, 42.05)
23.50 (19.50, 28.65)
0.700
LS BMD, g/cm2
0.55 (0.48, 0.73)g, h
0.41 (0.33, 0.50)
0.37 (0.33, 0.56)
0.001
LS BMD Z-score
− 0.93 ± 1.26 g, h
− 2.31 ± 1.54
− 2.31 ± 1.95
0.003
FN BMD, g/cm2
0.54 ± 0.13 g,h
0.42 ± 0.12
0.43 ± 0.18
0.013
FN BMD Z-score
− 2.16 (− 3.45, -1.51)g, h
− 4.01 (− 5.02, − 2.06)
− 3.45 (− 5.25, − 2.25)
0.027
Troch BMD, g/cm2
0.41 ± 0.10
0.35 ± 0.12
0.34 ± 0.16
0.104
TH BMD, g/cm2
0.54 ± 0.12g,h
0.45 ± 0.12
0.46 ± 0.18
0.048
OPG, ng/mL
0.71 (0.46, 1.14)
0.61 (0.48, 1.09)
0.55 (0.29, 0.99)
0.74 (0.52, 1.35)
0.314
Sclerostin, pmol/L
25.12 (19.65, 32.46)c
17.74 (14.00, 22.61)l
18.49 (14.46, 20.52)m
30.41 (22.51, 34.00)
0.000
DKK1, ng/mL
5.20 (4.61, 6.19) e
5.67 (4.29, 6.42)j
4.80 (4.06, 6.48)
4.08 (3.59, 4.92)
0.003
Blue sclera, n
28
12
13
0.340
Dentinogenesis imperfecta, n
3
2
2
0.656
Ligament laxity, n
14
5
6
0.965
Hearing abnormality, n
0
0
1
0.452
Age at first fracture, years
3.00 (1.00, 5.00)
1.75 (1.00, 4.50)
2.00 (1.05, 5.00)
0.534
Peripheral fracture, n
30
12
16
0.298
Number of peripheral fracture
3.00 (1.00, 4.50)
4.50 (3.00, 6.00)
3 (2.25, 5.00)
0.090
Frequency of peripheral fracture per year
0.64 (0.30, 1.00)c, d
1.00 (1.00, 2.88)
1.16 (0.85, 2.30)
0.000
VCF, n
11
3
8
0.334
Wheelchair dependence, n
7
4
4
0.619
Long bone deformity, n
1g, h
9
5
0.000
Scoliosis, n
0h
0
3
0.021
Ribcage deformity, n
2
2
2
0.449
The results for normally distributed data were presented as the mean ± SD
Nonnormally distributed data were presented as medians (quartiles)
Categorical data were presented as numbers
Bold values indicated significant differences among 3 or 4 groups
OI osteogenesis imperfecta, Ca calcium, P phosphorus, ALP alkaline phosphatase, ALT glutamic-pyruvic transaminase, Cr creatinine, PTH parathyroid hormone, P1NP procollagen type 1 N-peptide, β-CTX β-C-terminal telopeptide of type 1 collagen, 25OHD 25-hydroxyvitamin D, LS lumbar spine, FN femoral neck, TH total hip, BMD bone mineral density, OPG Osteoprotegerin, VCF vertebral compression fracture
aP < 0.001 for OI-I vs. OI-IV
bP < 0.001 for OI-I vs. control
cP < 0.01 for OI-I vs. OI-III
dP < 0.01 for OI-I vs. OI-IV
eP < 0.01 for OI-I vs. control
fP < 0.01 for OI-IV vs. control
gP < 0.05 for OI-I vs. OI-III
hP < 0.05 for OI-I vs. OI-IV
iP < 0.05 for OI-I vs. control
jP < 0.05 for OI-III vs. control
kP < 0.05 for OI-IV vs. Control
lP < 0.01 for OI-II vs. Control
mP < 0.001 for OI-III vs. OI-IV
The pathogenic mutation spectrum of OI patients in this study was as follows: COL1A1 (38/62, 61%), COL1A2 (17/62, 27%), FKBP10 (3/62, 5%), PLS3 (2/62, 3%), P4HB (1/62, 2%), and PLOD2 (1/62, 2%) (Fig. 1).

Serum DKK1 levels and bone turnover marker levels in children with OI

The serum DKK1 concentration was 5.20 (4.54, 6.32) ng/mL in children with OI, which was significantly higher than that of healthy children [4.08 (3.59, 4.92)] ng/mL (P < 0.001) (Fig. 2a). No difference was found in the serum DKK1 concentrations between boys and girls with OI (Fig. 2b). The serum DKK1 concentration was 5.20 (4.61, 6.19) ng/mL, 5.67 (4.29, 6.42) ng/mL and 4.80 (4.06, 6.48) ng/mL in children with type I, type III OI and type IV OI, respectively (Table 2). No significant difference in the serum DKK1 levels was observed among different clinical types of OI children (Fig. 2c). The serum DKK1 concentration in type I and type III OI children was significantly higher than that in normal children (all P < 0.05) (Fig. 2c).
The serum ALP level in children with OI was 319.15 ± 95.78 U/L, which was significantly higher than that in healthy children (229.58 ± 66.30 U/L) (P < 0.001). There were no significant differences in the serum 25-hydroxyvitamin D, calcium, phosphorus, β-CTX and P1NP levels between children with OI and healthy controls (Table 1). The serum sclerostin level in children with OI was 20.87 (14.94, 27.36) pmol/L, which was significantly lower than that in healthy children [30.41 (22.51, 34.00) pmol/L] (P < 0.01). There were no significant differences in the OPG level between children with OI and healthy controls (Table 1).

Correlation between serum DKK1 level and skeletal phenotype in OI children

After adjusting for age and gender, the serum DKK1 concentration in OI children exhibited a significantly negative correlation with lumbar spine BMD (r = − 0.276, P < 0.05) (Fig. 3a). However, no significant correlations were observed between DKK1 concentration and BMD at the femoral neck, greater trochanter, or total hip (Fig. 3b–d). Furthermore, there was a negative correlation between serum DKK1 concentration and BMD Z scores at LS and FN (r = − 0.315, P < 0.05; r = − 0.266, P < 0.05) (Fig. 3e, f).
The serum DKK1 concentration in OI children was negatively correlated with the ALP concentration (r = − 0.304, P < 0.05) (Fig. 4a) and positively correlated with serum calcium concentration (r = 0.257, P < 0.05) (Fig. 4d) and negatively correlated with PTH concentration (r = − 0.269, P < 0.05) (Fig. 4e). The serum DKK1 concentration in children with OI was negatively correlated with height (r = − 0.282, P < 0.05) and the height Z score (r = − 0.292, P < 0.05) (Fig. 5a, b). No significant correlations were found between serum DKK1 level and age, weight, weight Z score, serum levels of P1NP, β-CTX, P, 25-hydroxyvitamin D, sclerostin and OPG or liver and kidney function of children with OI (Fig. 4b, c, f, g, Fig. 5c, d and Supplementary Fig. 1a–b).
There was no significant correlation between the serum DKK1 concentration and annual peripheral fractures incidence of children with OI (Supplementary Fig. 1c). The serum DKK1 concentrations were 5.67 ± 1.12 ng/mL and 5.19 ± 1.12 ng/mL in OI patients with and without vertebral compression fractures, respectively, which had no significant difference (Table 3). In patients with spinal deformity (22/62), serum DKK1 concentration was positively correlated with SDI (r = 0.480, P < 0.05) (Fig. 3g).
Table 3
Serum DKK1 level in OI patients with different genetic mode, abnormal collagen metabolism, and spinal fracture or not
 
DKK1, ng/mL
P value
Different genders
 Boys
5.44 ± 1.10
0.342
 Girls
5.12 ± 1.26
Different genetic modes
 AD (n = 56)
5.35 ± 1.18
0.799
 Non-AD (n = 6)
5.47 ± 0.65
Abnormal collagen metabolism
 Structural defect (n = 25)
5.57 ± 1.27
0.359
 Quantitative reduction (n = 25)
5.26 ± 1.14
Vertebral compression fractures
 VCF (n = 22)
5.67 ± 1.12
0.113
 Non-VCF (n = 40)
5.19 ± 1.12
The results for normally distributed data were presented as the mean ± SD
Nonnormally distributed data were presented as medians (quartiles)
AD autosomal dominant inheritance, VCF vertebral compression fracture

Relationship between serum DKK1 level and genotype in OI children

According to the pathogenic gene mutation profile, 56 patients consisted with AD inheritance, 4 with AR inheritance, and 2 with X-linked inheritance. The serum DKK1 concentrations of OI children had no significant difference between the AD group and the non-AD group. Based on abnormal metabolic patterns of type I collagen, there were 25 patients with collagen structural defects (8 patients with α1 chain defects and 17 patients with α2 chain defects) and 25 patients with reduced collagen quantity. No significant difference was found between OI children with structural defects and insufficient quantity of type I collagen (Table 3).

Discussion

This study detected serum DKK1 level in a relatively large cohort of children with OI and assessed the correlation for the first time between DKK1 level and the skeletal phenotype and genotype in OI children. We found that serum DKK1 level of OI children was significantly higher than that of age-matched normal children. Interestingly, the serum DKK1 concentration was negatively correlated with the bone formation biomarker ALP, lumbar spine BMD and BMD Z scores at lumbar spine and femoral neck, and serum DKK1 concentration was positively correlated with SDI in OI children with spinal deformities. No significant correlation was found between DKK1 level and fracture incidence or OI pathogenic genotype. These findings indicated DKK1 may be a useful novel biomarker for OI.
A series of studies have shown that DKK1 plays important roles in regulating bone formation as it is the natural inhibitor of the WNT signaling pathway. DKK1 had been found to be associated with many skeletal diseases [2429]. In a 3-year follow-up study of liver transplant recipients, serum DKK1 level was significantly increased, and liver transplant recipients who experienced fractures had significantly higher DKK1 level than patients without fractures [25]. In patients with multiple myeloma, the level of DKK1 was significantly increased, and the severity of skeletal lesions was significantly positively correlated with the DKK1 level [26]. The mechanism involved that DKK1 could be secreted by multiple myeloma cells and bone marrow mesenchymal stem cells, which inhibited the classical WNT pathway, impeding osteoblast maturation and bone matrix mineralization, leading to osteolytic lesions of multiple myeloma [27]. In addition, a murine model of breast cancer had unveiled that DKK1 overexpression markedly enhanced bone metastasis and osteolysis, concurrently upregulated tumor proliferation within metastatic sites, and the knockdown of DKK1 could mitigate bone metastasis [28]. Moreover, patients with disuse osteoporosis due to long-term bed rest had elevated serum DKK1 level and reduced expression of β-catenin, resulting in decreased bone formation and increased bone resorption [29]. The above studies indicated that DKK1 is an important factor regulating bone turnover, and its abnormal secretion is closely related to various metabolic and tumorigenic bone diseases.
There were also studies on DKK1 in small sample of patients with OI and animal model of OI. In a study involving 18 OI children, the serum DKK1 level was higher in OI group than in the age-matched normal control group [30]. Furthermore, the study revealed that the serum from OI children could inhibit differentiation of osteoblasts, and this effect could be countered by an anti-DKK1 antibody [30]. In addition, animal studies revealed that the expression of DKK1 was higher and β-catenin was lower in bone tissues of OI model (OIM) mice [31]. DKK1 expression could be significantly inhibited after treatment with antisense oligonucleotides that antagonize microRNA-29a, and bone microarchitecture and BMD of OIM mice were improved [31]. Based on the findings of the above study and our study, DKK1 may regulate bone formation through affecting the WNT pathway, thereby participating in the pathological processes of OI.
As we know, the WNT pathway plays an essential role in osteoblast differentiation and maturation [32, 33], which is initiated when the WNT ligand binds to the Frizzled and LRP5/6 receptors simultaneously. The activation of co-receptors leads to the inhibition of glycogen synthase kinase 3 (Gsk3) activity and the stabilization of the β-catenin protein [34]. Stable β-catenin subsequently undergoes nuclear translocation and interacts with T-cell factor and lymphoid enhancer factor (TCF/LEF), a transcription factor, to promote gene expression in osteoblasts [34, 35]. DKK1 is a natural antagonist of the WNT pathway. Our studies demonstrated that serum DKK1 concentration was significantly higher in OI patients, which could inhibit the activity of the WNT pathway, leading to a decrease in the expression of multiple genes in osteoblasts, reduce differentiation and maturation of osteoblasts, and inhibit bone formation. Therefore, we found that DKK1 level was negatively correlated with bone formation biomarker of ALP and lumbar BMD, Z scores of BMD at lumbar spine and femoral neck, indicating that DKK1 had the potential as a novel biochemical marker for OI patients.
Moreover, the natural WNT antagonist, sclerostin, has become a important target of anti-osteoporosis drugs. Romosozumab, a monoclonal antibody of sclerostin, is effective in increasing BMD and reducing vertebral, nonvertebral, and hip fractures [36, 37]. It has shown potential therapeutic benefits in two patients with OI [38, 39]. Moreover, setrusumab, another monoclonal antibody of sclerostin, has exhibited promising treatment potential in a randomized Phase IIb study in adults with OI and in a phase 2/3 ORBIT study involving children and adolescents with OI [40, 41]. Furthermore, sclerostin nucleic acid aptamers targeting the Loop3 domain of sclerostin, can promote bone formation, increase BMD, and improve bone microarchitecture of OI animal model [42]. Notably, sclerostin antibodies have shown promising outcomes in various mouse models mimicking OI [4345]. These findings indicate that natural antagonists of the WNT pathway may hold immense potential in the treatment of OI.
It is worth noting that DKK1 is also natural antagonist of WNT pathway, which is expected to be a novel therapy target for OI. Studies showed that a reduction in DKK1 level through heterozygous gene knockout could lead to an increase in bone formation, vertebral trabecular bone volume and trabecular thickness in ovariectomized (OVX) mice [46]. The deletion of DKK1 could increase bone formation through resulting in an elevated skeletal expression of WNT target genes, including Lef1 and Axin2 [46]. In a murine model of multiple myelom, anti-DKK1 antibody could prevent bone loss in both trabecular and cortical compartments [47]. Moreover, animal experiments demonstrated that DKK1-Ab at a dose of 25 mg/kg twice a week for 28 days could enhance callus formation and bone mechanical strength in a tibial fracture mouse model [48]. These studies suggested that DKK1 may also be one of the potential targets for treatment of OI.
This study indicated that serum DKK1 level was higher in OI children than that in healthy children. Serum DKK1 level was found for the first time to be negatively correlated with serum ALP level, lumbar BMD, BMD Z scores at lumbar spine and femoral neck in children with OI. DKK1 level was also positively correlated with SDI in OI children with spinal deformities. However, this study had a series of limitations: only serum DKK1 concentration was measured, and we did not measure the DKK1 level in bone tissue, which is more important for regulating bone formation. In addition, this study was a cross-sectional design, which only indicated some correlations, but could not confirm their causal relationships. The sample size of this study was relatively small, and it was difficult to reveal the correlation between DKK1 level and fracture incidence or genotype of OI patients.

Conclusion

The serum DKK1 level was not only significantly elevated in OI children, but also closely correlated to their skeletal phenotype, suggesting that DKK1 may become a novel biomarker and a potential therapeutic target of OI.

Acknowledgements

We appreciate the patients and healthy controls for participation in this study. This study is supported by National Key R&D Program of China (2021YFC2501700), National Natural Science Foundation of China (82070908, 82370894), CAMS Innovation Fund for Medical Sciences (CIFMS) (2021-I2M-1-051), and National High-Level Hospital Clinical Research Funding (2022-PUMCH-B-014).

Declarations

Conflict of interest

The authors declare that they have no conflicts of interest. The authors have no disclosure.

Ethical approval

This study was approved by the Scientific Ethics Committee of PUMCH.
Informed consents were obtained from legal guardian of each OI patient and healthy children.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/​4.​0/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Tournis S, Dede AD (2018) Osteogenesis imperfect—a clinical update. Metab Clin Exp 80:27–37PubMedCrossRef Tournis S, Dede AD (2018) Osteogenesis imperfect—a clinical update. Metab Clin Exp 80:27–37PubMedCrossRef
3.
Zurück zum Zitat Gnoli M, Brizola E, Tremosini M, Di Cecco A, Sangiorgi L (2023) Vitamin d and bone fragility in individuals with osteogenesis imperfecta: a scoping review. Int J Mol Sci 24:9416PubMedPubMedCentralCrossRef Gnoli M, Brizola E, Tremosini M, Di Cecco A, Sangiorgi L (2023) Vitamin d and bone fragility in individuals with osteogenesis imperfecta: a scoping review. Int J Mol Sci 24:9416PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Jovanovic M, Mitra A, Besio R, Contento BM, Wong KW, Derkyi A et al (2023) Absence of tric-b from type xiv osteogenesis imperfecta osteoblasts alters cell adhesion and mitochondrial function—a multi-omics study. Matrix Biol J Int Soc Matrix Biol 121:127–148CrossRef Jovanovic M, Mitra A, Besio R, Contento BM, Wong KW, Derkyi A et al (2023) Absence of tric-b from type xiv osteogenesis imperfecta osteoblasts alters cell adhesion and mitochondrial function—a multi-omics study. Matrix Biol J Int Soc Matrix Biol 121:127–148CrossRef
5.
Zurück zum Zitat Daponte V, Tonelli F, Masiero C, Syx D, Exbrayat-Héritier C, Biggiogera M et al (2023) Cell differentiation and matrix organization are differentially affected during bone formation in osteogenesis imperfecta zebrafish models with different genetic defects impacting collagen type i structure. Matrix biol J Int Soc Matrix Biol 121:105–126CrossRef Daponte V, Tonelli F, Masiero C, Syx D, Exbrayat-Héritier C, Biggiogera M et al (2023) Cell differentiation and matrix organization are differentially affected during bone formation in osteogenesis imperfecta zebrafish models with different genetic defects impacting collagen type i structure. Matrix biol J Int Soc Matrix Biol 121:105–126CrossRef
6.
Zurück zum Zitat Hu J, Lin X, Gao P, Zhang Q, Zhou B, Wang O et al (2023) Genotypic and phenotypic spectrum and pathogenesis of wnt1 variants in a large cohort of patients with oi/osteoporosis. J Clin Endocrinol Metab 108:1776–1786PubMedPubMedCentralCrossRef Hu J, Lin X, Gao P, Zhang Q, Zhou B, Wang O et al (2023) Genotypic and phenotypic spectrum and pathogenesis of wnt1 variants in a large cohort of patients with oi/osteoporosis. J Clin Endocrinol Metab 108:1776–1786PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X et al (2022) Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther 7:3PubMedPubMedCentralCrossRef Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X et al (2022) Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther 7:3PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Wong SK, Mohamad NV, Jayusman PA, Ibrahim N (2023) A review on the crosstalk between insulin and wnt/β-catenin signalling for bone health. Int J Mol Sci 24:12441PubMedPubMedCentralCrossRef Wong SK, Mohamad NV, Jayusman PA, Ibrahim N (2023) A review on the crosstalk between insulin and wnt/β-catenin signalling for bone health. Int J Mol Sci 24:12441PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Dincel AS, Jørgensen NR (2023) New emerging biomarkers for bone disease: sclerostin and dickkopf-1 (dkk1). Calcif Tissue Int 112:243–257PubMedCrossRef Dincel AS, Jørgensen NR (2023) New emerging biomarkers for bone disease: sclerostin and dickkopf-1 (dkk1). Calcif Tissue Int 112:243–257PubMedCrossRef
10.
Zurück zum Zitat Bishop N, Adami S, Ahmed SF, Antón J, Arundel P, Burren CP et al (2013) Risedronate in children with osteogenesis imperfecta: a randomised, double-blind, placebo-controlled trial. Lancet 382:1424–1432PubMedCrossRef Bishop N, Adami S, Ahmed SF, Antón J, Arundel P, Burren CP et al (2013) Risedronate in children with osteogenesis imperfecta: a randomised, double-blind, placebo-controlled trial. Lancet 382:1424–1432PubMedCrossRef
11.
Zurück zum Zitat Li LJ, Zheng WB, Zhao DC, Yu W, Wang O, Jiang Y et al (2019) Effects of zoledronic acid on vertebral shape of children and adolescents with osteogenesis imperfecta. Bone 127:164–171PubMedCrossRef Li LJ, Zheng WB, Zhao DC, Yu W, Wang O, Jiang Y et al (2019) Effects of zoledronic acid on vertebral shape of children and adolescents with osteogenesis imperfecta. Bone 127:164–171PubMedCrossRef
12.
Zurück zum Zitat Li H, Ji CY, Zong XN, Zhang YQ (2009) height and weight standardized growth charts for chinese children and adolescents aged 0 to 18 years. Zhonghua er ke za zhi Chin J Pediatr 47:487–492 Li H, Ji CY, Zong XN, Zhang YQ (2009) height and weight standardized growth charts for chinese children and adolescents aged 0 to 18 years. Zhonghua er ke za zhi Chin J Pediatr 47:487–492
13.
Zurück zum Zitat Genant HK, Wu CY, van Kuijk C, Nevitt MC (1993) Vertebral fracture assessment using a semiquantitative technique. J Bone Miner Res 8:1137–1148PubMedCrossRef Genant HK, Wu CY, van Kuijk C, Nevitt MC (1993) Vertebral fracture assessment using a semiquantitative technique. J Bone Miner Res 8:1137–1148PubMedCrossRef
14.
Zurück zum Zitat Kerkeni S, Kolta S, Fechtenbaum J, Roux C (2009) Spinal deformity index (sdi) is a good predictor of incident vertebral fractures. Osteoporos Int 20:1547–1552PubMedCrossRef Kerkeni S, Kolta S, Fechtenbaum J, Roux C (2009) Spinal deformity index (sdi) is a good predictor of incident vertebral fractures. Osteoporos Int 20:1547–1552PubMedCrossRef
15.
Zurück zum Zitat Kuznia AL, Hernandez AK, Lee LU (2020) Adolescent idiopathic scoliosis: common questions and answers. Am Fam Physician 101:19–23PubMed Kuznia AL, Hernandez AK, Lee LU (2020) Adolescent idiopathic scoliosis: common questions and answers. Am Fam Physician 101:19–23PubMed
16.
Zurück zum Zitat Khadilkar AV, Sanwalka NJ, Chiplonkar SA, Khadilkar VV, Mughal MZ (2011) Normative data and percentile curves for dual energy x-ray absorptiometry in healthy indian girls and boys aged 5–17 years. Bone 48:810–819PubMedCrossRef Khadilkar AV, Sanwalka NJ, Chiplonkar SA, Khadilkar VV, Mughal MZ (2011) Normative data and percentile curves for dual energy x-ray absorptiometry in healthy indian girls and boys aged 5–17 years. Bone 48:810–819PubMedCrossRef
17.
Zurück zum Zitat Xu H, Zhao Z, Wang H, Ding M, Zhou A, Wang X et al (2013) Bone mineral density of the spine in 11,898 chinese infants and young children: a cross-sectional study. PLoS ONE 8:e82098PubMedPubMedCentralCrossRef Xu H, Zhao Z, Wang H, Ding M, Zhou A, Wang X et al (2013) Bone mineral density of the spine in 11,898 chinese infants and young children: a cross-sectional study. PLoS ONE 8:e82098PubMedPubMedCentralCrossRef
18.
19.
Zurück zum Zitat Whyte MP, Aronson J, McAlister WH, Weinstein RS, Wenkert D, Clements KL et al (2021) Coalescing expansile skeletal disease: Delineation of an extraordinary osteopathy involving the ifitm5 mutation of osteogenesis imperfecta type v. Bone 145:115835PubMedCrossRef Whyte MP, Aronson J, McAlister WH, Weinstein RS, Wenkert D, Clements KL et al (2021) Coalescing expansile skeletal disease: Delineation of an extraordinary osteopathy involving the ifitm5 mutation of osteogenesis imperfecta type v. Bone 145:115835PubMedCrossRef
20.
Zurück zum Zitat Liu Y, Asan MD, Lv F, Xu X, Wang J et al (2017) Gene mutation spectrum and genotype-phenotype correlation in a cohort of chinese osteogenesis imperfecta patients revealed by targeted next generation sequencing. Osteoporos Int 28:2985–2995PubMedCrossRef Liu Y, Asan MD, Lv F, Xu X, Wang J et al (2017) Gene mutation spectrum and genotype-phenotype correlation in a cohort of chinese osteogenesis imperfecta patients revealed by targeted next generation sequencing. Osteoporos Int 28:2985–2995PubMedCrossRef
21.
Zurück zum Zitat Claeys L, Storoni S, Eekhoff M, Elting M, Wisse L, Pals G et al (2021) Collagen transport and related pathways in osteogenesis imperfecta. Hum Genet 140:1121–1141PubMedPubMedCentralCrossRef Claeys L, Storoni S, Eekhoff M, Elting M, Wisse L, Pals G et al (2021) Collagen transport and related pathways in osteogenesis imperfecta. Hum Genet 140:1121–1141PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J et al (2015) Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the american college of medical genetics and genomics and the association for molecular pathology. Genet Med Off J Am Coll Med Genet 17:405–424 Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J et al (2015) Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the american college of medical genetics and genomics and the association for molecular pathology. Genet Med Off J Am Coll Med Genet 17:405–424
25.
Zurück zum Zitat Kuo SJ, Chen CL, Chen SH, Ko JY (2023) Changes in serum bone metabolism markers after living donor liver transplantation (ldlt) and their association with fracture occurrences. Life (Basel, Switzerland) 13:1438PubMed Kuo SJ, Chen CL, Chen SH, Ko JY (2023) Changes in serum bone metabolism markers after living donor liver transplantation (ldlt) and their association with fracture occurrences. Life (Basel, Switzerland) 13:1438PubMed
26.
Zurück zum Zitat Gerov V, Gerova D, Micheva I, Nikolova M, Mihaylova G, Galunska B (2023) Dynamics of bone disease biomarkers dickkopf-1 and sclerostin in patients with multiple myeloma. J Clin Med 12:4440PubMedPubMedCentralCrossRef Gerov V, Gerova D, Micheva I, Nikolova M, Mihaylova G, Galunska B (2023) Dynamics of bone disease biomarkers dickkopf-1 and sclerostin in patients with multiple myeloma. J Clin Med 12:4440PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Zhuang X, Zhang H, Li X, Li X, Cong M, Peng F et al (2017) Differential effects on lung and bone metastasis of breast cancer by wnt signalling inhibitor dkk1. Nat Cell Biol 19:1274–1285PubMedCrossRef Zhuang X, Zhang H, Li X, Li X, Cong M, Peng F et al (2017) Differential effects on lung and bone metastasis of breast cancer by wnt signalling inhibitor dkk1. Nat Cell Biol 19:1274–1285PubMedCrossRef
29.
Zurück zum Zitat Ma X, Zhu X, He X, Yi X, Jin A (2021) The wnt pathway regulator expression levels and their relationship to bone metabolism in thoracolumbar osteoporotic vertebral compression fracture patients. Am J Transl Res 13:4812–4818PubMedPubMedCentral Ma X, Zhu X, He X, Yi X, Jin A (2021) The wnt pathway regulator expression levels and their relationship to bone metabolism in thoracolumbar osteoporotic vertebral compression fracture patients. Am J Transl Res 13:4812–4818PubMedPubMedCentral
30.
Zurück zum Zitat Brunetti G, Papadia F, Tummolo A, Fischetto R, Nicastro F, Piacente L et al (2016) Impaired bone remodeling in children with osteogenesis imperfecta treated and untreated with bisphosphonates: The role of dkk1, rankl, and tnf-α. Osteoporos Int 27:2355–2365PubMedCrossRef Brunetti G, Papadia F, Tummolo A, Fischetto R, Nicastro F, Piacente L et al (2016) Impaired bone remodeling in children with osteogenesis imperfecta treated and untreated with bisphosphonates: The role of dkk1, rankl, and tnf-α. Osteoporos Int 27:2355–2365PubMedCrossRef
31.
Zurück zum Zitat Ko JY, Wang FS, Chen SH, Kuo SJ (2023) Micro ribonucleic acid-29a (mir-29a) antagonist normalizes bone metabolism in osteogenesis imperfecta (oi) mice model. Biomedicines 11:465PubMedPubMedCentralCrossRef Ko JY, Wang FS, Chen SH, Kuo SJ (2023) Micro ribonucleic acid-29a (mir-29a) antagonist normalizes bone metabolism in osteogenesis imperfecta (oi) mice model. Biomedicines 11:465PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Brunetti G, D’Amato G, Chiarito M, Tullo A, Colaianni G, Colucci S et al (2019) An update on the role of rankl-rank/osteoprotegerin and wnt-ß-catenin signaling pathways in pediatric diseases. World J Pediatr WJP 15:4–11PubMedCrossRef Brunetti G, D’Amato G, Chiarito M, Tullo A, Colaianni G, Colucci S et al (2019) An update on the role of rankl-rank/osteoprotegerin and wnt-ß-catenin signaling pathways in pediatric diseases. World J Pediatr WJP 15:4–11PubMedCrossRef
34.
35.
Zurück zum Zitat Zhang B, Li R, Wang W, Zhou X, Luo B, Zhu Z et al (2020) The role of wnt1 mutant variant (wnt1(c.677c>t) ) in osteogenesis imperfecta. Ann Hum Genet 84:447–455PubMedPubMedCentralCrossRef Zhang B, Li R, Wang W, Zhou X, Luo B, Zhu Z et al (2020) The role of wnt1 mutant variant (wnt1(c.677c>t) ) in osteogenesis imperfecta. Ann Hum Genet 84:447–455PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S et al (2016) Romosozumab treatment in postmenopausal women with osteoporosis. N Engl J Med 375:1532–1543PubMedCrossRef Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S et al (2016) Romosozumab treatment in postmenopausal women with osteoporosis. N Engl J Med 375:1532–1543PubMedCrossRef
37.
Zurück zum Zitat Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T et al (2017) Romosozumab or alendronate for fracture prevention in women with osteoporosis. N Engl J Med 377:1417–1427PubMedCrossRef Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T et al (2017) Romosozumab or alendronate for fracture prevention in women with osteoporosis. N Engl J Med 377:1417–1427PubMedCrossRef
38.
Zurück zum Zitat Dattagupta A, Petak S (2023) Osteoporosis improved by romosozumab therapy in a patient with type i osteogenesis imperfecta. AACE clinical case reports 9:209–212PubMedPubMedCentralCrossRef Dattagupta A, Petak S (2023) Osteoporosis improved by romosozumab therapy in a patient with type i osteogenesis imperfecta. AACE clinical case reports 9:209–212PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Uehara M, Nakamura Y, Nakano M, Miyazaki A, Suzuki T, Takahashi J (2022) Efficacy of romosozumab for osteoporosis in a patient with osteogenesis imperfecta: a case report. Modern rheumatology case reports 6:128–133PubMedCrossRef Uehara M, Nakamura Y, Nakano M, Miyazaki A, Suzuki T, Takahashi J (2022) Efficacy of romosozumab for osteoporosis in a patient with osteogenesis imperfecta: a case report. Modern rheumatology case reports 6:128–133PubMedCrossRef
40.
Zurück zum Zitat Glorieux FH, Devogelaer JP, Durigova M, Goemaere S, Hemsley S, Jakob F et al (2017) Bps804 anti-sclerostin antibody in adults with moderate osteogenesis imperfecta: results of a randomized phase 2a trial. J Bone Miner Res 32:1496–1504PubMedCrossRef Glorieux FH, Devogelaer JP, Durigova M, Goemaere S, Hemsley S, Jakob F et al (2017) Bps804 anti-sclerostin antibody in adults with moderate osteogenesis imperfecta: results of a randomized phase 2a trial. J Bone Miner Res 32:1496–1504PubMedCrossRef
41.
Zurück zum Zitat Lewiecki EM (2023) Evaluating setrusumab for the treatment of osteogenesis imperfecta: phase 2 data from the phase 2/3 ORBIT study. J Bone Miner Res 38(2) Lewiecki EM (2023) Evaluating setrusumab for the treatment of osteogenesis imperfecta: phase 2 data from the phase 2/3 ORBIT study. J Bone Miner Res 38(2)
42.
Zurück zum Zitat Wang L, Yu Y, Ni S, Li D, Liu J, Xie D et al (2022) Therapeutic aptamer targeting sclerostin loop3 for promoting bone formation without increasing cardiovascular risk in osteogenesis imperfecta mice. Theranostics 12:5645–5674PubMedPubMedCentralCrossRef Wang L, Yu Y, Ni S, Li D, Liu J, Xie D et al (2022) Therapeutic aptamer targeting sclerostin loop3 for promoting bone formation without increasing cardiovascular risk in osteogenesis imperfecta mice. Theranostics 12:5645–5674PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Cardinal M, Dessain A, Roels T, Lafont S, Ominsky MS, Devogelaer JP et al (2020) Sclerostin-antibody treatment decreases fracture rates in axial skeleton and improves the skeletal phenotype in growing oim/oim mice. Calcif Tissue Int 106:494–508PubMedCrossRef Cardinal M, Dessain A, Roels T, Lafont S, Ominsky MS, Devogelaer JP et al (2020) Sclerostin-antibody treatment decreases fracture rates in axial skeleton and improves the skeletal phenotype in growing oim/oim mice. Calcif Tissue Int 106:494–508PubMedCrossRef
44.
Zurück zum Zitat Olvera D, Stolzenfeld R, Marini JC, Caird MS, Kozloff KM (2018) Low dose of bisphosphonate enhances sclerostin antibody-induced trabecular bone mass gains in brtl/+ osteogenesis imperfecta mouse model. J Bone Miner Res 33:1272–1282PubMedCrossRef Olvera D, Stolzenfeld R, Marini JC, Caird MS, Kozloff KM (2018) Low dose of bisphosphonate enhances sclerostin antibody-induced trabecular bone mass gains in brtl/+ osteogenesis imperfecta mouse model. J Bone Miner Res 33:1272–1282PubMedCrossRef
45.
Zurück zum Zitat Cardinal M, Tys J, Roels T, Lafont S, Ominsky MS, Devogelaer JP et al (2019) Sclerostin antibody reduces long bone fractures in the oim/oim model of osteogenesis imperfecta. Bone 124:137–147PubMedCrossRef Cardinal M, Tys J, Roels T, Lafont S, Ominsky MS, Devogelaer JP et al (2019) Sclerostin antibody reduces long bone fractures in the oim/oim model of osteogenesis imperfecta. Bone 124:137–147PubMedCrossRef
46.
Zurück zum Zitat Lehmann J, Thiele S, Baschant U, Rachner TD, Niehrs C, Hofbauer LC et al (2021) Mice lacking dkk1 in t cells exhibit high bone mass and are protected from estrogen-deficiency-induced bone loss. iScience 24:102224. Lehmann J, Thiele S, Baschant U, Rachner TD, Niehrs C, Hofbauer LC et al (2021) Mice lacking dkk1 in t cells exhibit high bone mass and are protected from estrogen-deficiency-induced bone loss. iScience 24:102224.
47.
Zurück zum Zitat Simic MK, Mohanty ST, Xiao Y, Cheng TL, Taylor VE, Charlat O et al (2023) Multi-targeting dkk1 and lrp6 prevents bone loss and improves fracture resistance in multiple myeloma. J Bone Miner Res 38:814–828PubMedCrossRef Simic MK, Mohanty ST, Xiao Y, Cheng TL, Taylor VE, Charlat O et al (2023) Multi-targeting dkk1 and lrp6 prevents bone loss and improves fracture resistance in multiple myeloma. J Bone Miner Res 38:814–828PubMedCrossRef
48.
Zurück zum Zitat Jin H, Wang B, Li J, Xie W, Mao Q, Li S et al (2015) Anti-dkk1 antibody promotes bone fracture healing through activation of β-catenin signaling. Bone 71:63–75PubMedCrossRef Jin H, Wang B, Li J, Xie W, Mao Q, Li S et al (2015) Anti-dkk1 antibody promotes bone fracture healing through activation of β-catenin signaling. Bone 71:63–75PubMedCrossRef
49.
Zurück zum Zitat Zhang C, Zhao Z, Sun Y, Xu L, JiaJue R, Cui L et al (2019) Clinical and genetic analysis in a large chinese cohort of patients with x-linked hypophosphatemia. Bone 121:212–220PubMedCrossRef Zhang C, Zhao Z, Sun Y, Xu L, JiaJue R, Cui L et al (2019) Clinical and genetic analysis in a large chinese cohort of patients with x-linked hypophosphatemia. Bone 121:212–220PubMedCrossRef
50.
Zurück zum Zitat Chubb SAP, Vasikaran SD, Gillett MJ (2023) Reference intervals for plasma β-ctx and p1np in children: A systematic review and pooled estimates. Clin Biochem 118:110582PubMedCrossRef Chubb SAP, Vasikaran SD, Gillett MJ (2023) Reference intervals for plasma β-ctx and p1np in children: A systematic review and pooled estimates. Clin Biochem 118:110582PubMedCrossRef
Metadaten
Titel
Correlation of serum DKK1 level with skeletal phenotype in children with osteogenesis imperfecta
verfasst von
Y. Wang
J. Hu
L. Sun
B. Zhou
X. Lin
Q. Zhang
O. Wang
Y. Jiang
W. Xia
X. Xing
M. Li
Publikationsdatum
14.05.2024
Verlag
Springer International Publishing
Erschienen in
Journal of Endocrinological Investigation
Elektronische ISSN: 1720-8386
DOI
https://doi.org/10.1007/s40618-024-02380-9

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.