Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.160
Filter
1.
JCI Insight ; 9(11)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38855864

ABSTRACT

The transcription factor SRY-related HMG box 9 (Sox9) is essential for chondrogenesis. Mutations in and around SOX9 cause campomelic dysplasia (CD) characterized by skeletal malformations. Although the function of Sox9 in this context is well studied, the mechanisms that regulate Sox9 expression in chondrocytes remain to be elucidated. Here, we have used genome-wide profiling to identify 2 Sox9 enhancers located in a proximal breakpoint cluster responsible for CD. Enhancer activity of E308 (located 308 kb 5' upstream) and E160 (located 160 kb 5' upstream) correlated with Sox9 expression levels, and both enhancers showed a synergistic effect in vitro. While single deletions in mice had no apparent effect, simultaneous deletion of both E308 and E160 caused a dwarf phenotype, concomitant with a reduction of Sox9 expression in chondrocytes. Moreover, bone morphogenetic protein 2-dependent chondrocyte differentiation of limb bud mesenchymal cells was severely attenuated in E308/E160 deletion mice. Finally, we found that an open chromatin region upstream of the Sox9 gene was reorganized in the E308/E160 deletion mice to partially compensate for the loss of E308 and E160. In conclusion, our findings reveal a mechanism of Sox9 gene regulation in chondrocytes that might aid in our understanding of the pathophysiology of skeletal disorders.


Subject(s)
Campomelic Dysplasia , Cell Differentiation , Chondrocytes , Chondrogenesis , SOX9 Transcription Factor , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Animals , Chondrocytes/metabolism , Mice , Campomelic Dysplasia/genetics , Campomelic Dysplasia/pathology , Campomelic Dysplasia/metabolism , Chondrogenesis/genetics , Cell Differentiation/genetics , Enhancer Elements, Genetic/genetics , Chromatin/metabolism , Chromatin/genetics , Gene Expression Regulation, Developmental , Mice, Knockout , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Humans , Bone Development/genetics
2.
Nat Commun ; 15(1): 4820, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844479

ABSTRACT

Chondrocyte differentiation controls skeleton development and stature. Here we provide a comprehensive map of chondrocyte-specific enhancers and show that they provide a mechanistic framework through which non-coding genetic variants can influence skeletal development and human stature. Working with fetal chondrocytes isolated from mice bearing a Col2a1 fluorescent regulatory sensor, we identify 780 genes and 2'704 putative enhancers specifically active in chondrocytes using a combination of RNA-seq, ATAC-seq and H3K27ac ChIP-seq. Most of these enhancers (74%) show pan-chondrogenic activity, with smaller populations being restricted to limb (18%) or trunk (8%) chondrocytes only. Notably, genetic variations overlapping these enhancers better explain height differences than those overlapping non-chondrogenic enhancers. Finally, targeted deletions of identified enhancers at the Fgfr3, Col2a1, Hhip and, Nkx3-2 loci confirm their role in regulating cognate genes. This enhancer map provides a framework for understanding how genes and non-coding variations influence bone development and diseases.


Subject(s)
Chondrocytes , Chondrogenesis , Enhancer Elements, Genetic , Receptor, Fibroblast Growth Factor, Type 3 , Animals , Enhancer Elements, Genetic/genetics , Humans , Chondrocytes/metabolism , Chondrocytes/cytology , Mice , Chondrogenesis/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Gene Expression Regulation, Developmental , Bone Development/genetics , Extremities/embryology , Male , Cell Differentiation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Female
3.
BMJ Open ; 14(5): e075417, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760054

ABSTRACT

PURPOSE: The Bone And MicroBiOme Onset (BAMBOO) study is an ongoing prospective observational cohort study conducted in Tianjin, China, aiming to determine age-appropriate trajectories for microbiome maturation and bone development and to identify the influence of dietary factors in the process. PARTICIPANTS: The recruitment started in September 2021 and was completed in February 2023. A total of 1380 subjects were recruited, 690 at birth (group 1) and 690 at 6 months of age (group 2). Groups 1 and 2 will be followed up for 12 months and 36 months, respectively. FINDINGS TO DATE: The age of the mothers was 31.1±3.7 (mean±SD), and the birth weight of infants was 3.3±0.5 kg with an incidence of caesarean section 50.4%. Food diary information of the first 100 subjects showed that 64 food items were introduced by 6 months. A pilot microbiome analysis revealed that at the species level, bacterial communities were composed of mostly Bacteroides dorei, Bacteroides vulgatus and Escherichia coli, which were consistent with that of previous reports. Feasibility assessments of breast milk vitamin D and human milk oligosaccharides were validated through certified reference measurements. The early data assessment showed a high reliability of the data generated from this study. FUTURE PLANS: Data collection will be completed in August 2025. Four stage-statistical analyses will be performed as the cohort reaches certain age thresholds before the final report. Analysis of BAMBOO data will be used to develop age-appropriate trajectories for microbiome maturation and bone development for children aged 0-3 years and investigate the contribution of dietary factors in the process. TRIAL REGISTRATION NUMBER: ChiCTR2100049972.


Subject(s)
Bone Development , Humans , China , Infant , Female , Prospective Studies , Infant, Newborn , Male , Bone Development/physiology , Milk, Human/microbiology , Gastrointestinal Microbiome/physiology , Adult , Child, Preschool , Vitamin D , Diet , Cohort Studies
4.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38752331

ABSTRACT

C-type natriuretic peptide (CNP) plays a crucial role in enhancing endochondral bone growth and holds promise as a therapeutic agent for impaired skeletal growth. To overcome CNP's short half-life, we explored the potential of dampening its clearance system. Neprilysin (NEP) is an endopeptidase responsible for catalyzing the degradation of CNP. Thus, we investigated the effects of NEP inhibition on skeletal growth by administering sacubitril, a NEP inhibitor, to C57BL/6 mice. Remarkably, we observed a dose-dependent skeletal overgrowth phenotype in mice treated with sacubitril. Histological analysis of the growth plate revealed a thickening of the hypertrophic and proliferative zones, mirroring the changes induced by CNP administration. The promotion of skeletal growth observed in wild-type mice treated with sacubitril was nullified by the knockout of cartilage-specific natriuretic peptide receptor B (NPR-B). Notably, sacubitril promoted skeletal growth in mice only at 3 to 4 weeks of age, a period when endogenous CNP and NEP expression was higher in the lumbar vertebrae. Additionally, sacubitril facilitated endochondral bone growth in organ culture experiments using tibial explants from fetal mice. These findings suggest that NEP inhibition significantly promotes skeletal growth via the CNP/NPR-B pathway, warranting further investigations for potential applications in people with short stature.


Subject(s)
Biphenyl Compounds , Bone Development , Mice, Inbred C57BL , Natriuretic Peptide, C-Type , Neprilysin , Animals , Neprilysin/metabolism , Neprilysin/antagonists & inhibitors , Neprilysin/genetics , Natriuretic Peptide, C-Type/pharmacology , Natriuretic Peptide, C-Type/metabolism , Bone Development/drug effects , Mice , Biphenyl Compounds/pharmacology , Mice, Knockout , Aminobutyrates/pharmacology , Signal Transduction/drug effects , Male , Valsartan/pharmacology , Growth Plate/drug effects , Growth Plate/metabolism , Drug Combinations , Tetrazoles/pharmacology
5.
Cell Rep Med ; 5(5): 101574, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38776873

ABSTRACT

The existing suite of therapies for bone diseases largely act to prevent further bone loss but fail to stimulate healthy bone formation and repair. We describe an endogenous osteopeptide (PEPITEM) with anabolic osteogenic activity, regulating bone remodeling in health and disease. PEPITEM acts directly on osteoblasts through NCAM-1 signaling to promote their maturation and formation of new bone, leading to enhanced trabecular bone growth and strength. Simultaneously, PEPITEM stimulates an inhibitory paracrine loop: promoting osteoblast release of the decoy receptor osteoprotegerin, which sequesters RANKL, thereby limiting osteoclast activity and bone resorption. In disease models, PEPITEM therapy halts osteoporosis-induced bone loss and arthritis-induced bone damage in mice and stimulates new bone formation in osteoblasts derived from patient samples. Thus, PEPITEM offers an alternative therapeutic option in the management of diseases with excessive bone loss, promoting an endogenous anabolic pathway to induce bone remodeling and redress the imbalance in bone turnover.


Subject(s)
Bone Resorption , Osteoblasts , Osteogenesis , Animals , Humans , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Mice , Bone Resorption/pathology , Bone Resorption/metabolism , Anabolic Agents/pharmacology , Anabolic Agents/therapeutic use , Bone Remodeling/drug effects , Osteoporosis/pathology , Osteoporosis/metabolism , Osteoporosis/drug therapy , RANK Ligand/metabolism , Osteoclasts/metabolism , Osteoclasts/drug effects , Bone Development/drug effects , Osteoprotegerin/metabolism , Female , Signal Transduction/drug effects , Peptides/pharmacology , Male , Mice, Inbred C57BL , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology
6.
Ecotoxicol Environ Saf ; 279: 116501, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38805831

ABSTRACT

6:2 Chlorinated polyfluoroalkyl ether sulfonate (F-53B) is a new type of perfluorinated and polyfluoroalkyl substance (PFAS) that is used extensively in industry and manufacturing. F-53B causes damage to multiple mammalian organs. However, the impacts of F-53B on bone are unknown. Maternal exposure to F-53B is of particular concern because of the vulnerability of the developing fetus and newborn to contaminants from the mother. The goal of this study was to examine the impacts of maternal F-53B exposure on bone growth and development in offspring and to explore its underlying mechanisms. Herein, C57BL/6 J mice were given free access to deionized water containing 0, 0.57, or 5.7 mg/L F-53B during pregnancy and lactation. F-53B exposure resulted in impaired liver function, decreased IGF-1 secretion, dysregulation of bone metabolism and disruption of the dynamic balance between osteoblasts and osteoclasts in male offspring. F-53B inhibits longitudinal bone growth and development and causes osteoporosis in male offspring. F-53B may affect the growth and development of offspring bone via the IGF-1/OPG/RANKL/CTSK signaling pathway. This study provides new insights for the study of short stature and bone injury caused by F-53B.


Subject(s)
Bone Development , Lactation , Maternal Exposure , Mice, Inbred C57BL , Prenatal Exposure Delayed Effects , Animals , Female , Male , Pregnancy , Mice , Maternal Exposure/adverse effects , Bone Development/drug effects , Insulin-Like Growth Factor I/metabolism , Fluorocarbons/toxicity , Osteoprotegerin/metabolism , Osteoclasts/drug effects , Bone and Bones/drug effects , Osteoblasts/drug effects , Sulfonic Acids/toxicity
7.
Cell Signal ; 120: 111222, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38729327

ABSTRACT

BACKGROUND: Bone development involves the rapid proliferation and differentiation of osteogenic lineage cells, which makes accurate chromosomal segregation crucial for ensuring cell proliferation and maintaining chromosomal stability. However, the mechanism underlying the maintenance of chromosome stability during the rapid proliferation and differentiation of Prx1-expressing limb bud mesenchymal cells into osteoblastic precursor cells remains unexplored. METHODS: A transgenic mouse model of RanGAP1 knockout of limb and head mesenchymal progenitor cells was constructed to explore the impact of RanGAP1 deletion on bone development by histomorphology and immunostaining. Subsequently, G-banding karyotyping analysis and immunofluorescence staining were used to examine the effects of RanGAP1 deficiency on chromosome instability. Finally, the effects of RanGAP1 deficiency on chromothripsis and bone development signaling pathways were elucidated by whole-genome sequencing, RNA-sequencing, and qPCR. RESULTS: The ablation of RanGAP1 in limb and head mesenchymal progenitor cells expressing Prx1 in mice resulted in embryonic lethality, severe cartilage and bone dysplasia, and complete loss of cranial vault formation. Moreover, RanGAP1 loss inhibited chondrogenic or osteogenic differentiation of mesenchymal stem cells (MSCs). Most importantly, we found that RanGAP1 loss in limb bud mesenchymal cells triggered missegregation of chromosomes, resulting in chromothripsis of chromosomes 1q and 14q, further inhibiting the expression of key genes involved in multiple bone development signaling pathways such as WNT, Hedgehog, TGF-ß/BMP, and PI3K/AKT in the chromothripsis regions, ultimately disrupting skeletal development. CONCLUSIONS: Our results establish RanGAP1 as a critical regulator of bone development, as it supports this process by preserving chromosome stability in Prx1-expressing limb bud mesenchymal cells.


Subject(s)
Cell Differentiation , Chromosomal Instability , Limb Buds , Mesenchymal Stem Cells , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Limb Buds/metabolism , Bone Development , Osteogenesis/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Signal Transduction , Mice, Knockout , Chondrogenesis/genetics
8.
Gene ; 923: 148561, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-38754570

ABSTRACT

Intermuscular bones, which are present in numerous economically significant fish species, have a negative impact on the development of aquaculture. The Asb15b gene, primarily expressed in skeletal muscle, plays a crucial role in regulating protein turnover and the development of muscle fibers. It stimulates protein synthesis and controls the differentiation of muscle fibers. In this study, we employed CRISPR/Cas9 technology to generate homozygous zebrafish strains with 7 bp and 49 bp deletions in the Asb15b gene. Subsequent analyses using skeleton staining demonstrated a substantial reduction in the number of intermuscular bones in adult Asb15b-/- -7 bp and Asb15b-/- -49 bp mutants compared to the wild-type zebrafish, with decreases of 30 % (P < 0.001) and 40 % (P < 0.0001), respectively. Histological experiments further revealed that the diameter and number of muscle fibers in adult Asb15b-/- mutants did not exhibit significant changes when compared to wild-type zebrafish. Moreover, qRT-PCR experiments demonstrated significant differences in the expression of bmp6 and runx2b genes, which are key regulators of intermuscular bone development, during different stages of intermuscular bone development in Asb15b-/- mutants. This study strongly suggests that the Asb15b gene plays a crucial role in regulating intermuscular bone development in fish and lays the groundwork for further exploration of the role of the Asb15b gene in zebrafish intermuscular bone development.


Subject(s)
Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , CRISPR-Cas Systems , Gene Deletion , Muscle, Skeletal/metabolism , Bone Development/genetics , Bone and Bones/metabolism , Gene Expression Regulation, Developmental
9.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 256-262, 2024 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-38645858

ABSTRACT

Runt-related transcription factor (RUNX1) is a transcription factor closely involved in hematopoiesis. RUNX1 gene mutation plays an essential pathogenic role in the initiation and development of hematological tumors, especially in acute myeloid leukemia. Recent studies have shown that RUNX1 is also involved in the regulation of bone development and the pathological progression of bone-related diseases. RUNX1 promotes the differentiation of mesenchymal stem cells into chondrocytes and osteoblasts and modulates the maturation and extracellular matrix formation of chondrocytes. The expression of RUNX1 in mesenchymal stem cells, chondrocytes, and osteoblasts is of great significance for maintaining normal bone development and the mass and quality of bones. RUNX1 also inhibits the differentiation and bone resorptive activities of osteoclasts, which may be influenced by sexual dimorphism. In addition, RUNX1 deficiency contributes to the pathogenesis of osteoarthritis, delayed fracture healing, and osteoporosis, which was revealed by the RUNX1 conditional knockout modeling in mice. However, the roles of RUNX1 in regulating the hypertrophic differentiation of chondrocytes, the sexual dimorphism of activities of osteoclasts, as well as bone loss in diabetes mellitus, senescence, infection, chronic inflammation, etc, are still not fully understood. This review provides a systematic summary of the research progress concerning RUNX1 in the field of bone biology, offering new ideas for using RUNX1 as a potential target for bone related diseases, especially osteoarthritis, delayed fracture healing, and osteoporosis.


Subject(s)
Bone Development , Cell Differentiation , Chondrocytes , Core Binding Factor Alpha 2 Subunit , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Humans , Animals , Bone Development/physiology , Bone Development/genetics , Chondrocytes/metabolism , Osteoblasts/metabolism , Osteoblasts/cytology , Osteoclasts/metabolism , Osteoclasts/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Bone Diseases/genetics , Bone Diseases/metabolism , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/etiology
10.
Anat Sci Int ; 99(3): 268-277, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38598056

ABSTRACT

Because experimental studies to determine the developmental toxicity of exposure to various substances in children are impossible, many studies use immature male rats. This study aimed to provide normative data for longitudinal bone growth with age during the puberty in male rats. In order to evaluate long bone growth and mineralization we examined bone size and bone density by dual-energy X-ray absorptiometry, analyzed histomorphometry of the growth plate, and serum hormone levels relevant to bone growth from postnatal day (PD)20 to PD60. The length and weight of long bones increased strongly by PD40, and no further increase was observed after PD50. On the other hand, tibial growth plate height decreased sharply after PD50 along with a reduction in the number of cells and columns, which was probably responsible for the absence of further lengthening of long bones. Parameters related to bone formation such as bone area ratio, and the thickness and number of trabeculae, also increased significantly between PD40 and PD50. Furthermore, serum levels of IGF-1 peaked at PD30 and testosterone increased rapidly on and after PD40, when IGF-1 levels were going down. These changes may participate in the parallel increase in mineral acquisition, as well as lengthening of long bones. Our findings provide comprehensive data for changes in bone density, histomorphometry of long bones, and hormone levels relevant to bone growth during the growth spurt. This will be useful for planning animal toxicological studies, particularly for deciding on the appropriate age of animals to use in given experiments.


Subject(s)
Absorptiometry, Photon , Bone Density , Bone Development , Insulin-Like Growth Factor I , Animals , Male , Rats , Insulin-Like Growth Factor I/metabolism , Testosterone/blood , Tibia/growth & development , Growth Plate/growth & development , Rats, Wistar , Sexual Maturation/physiology
11.
Biol Res ; 57(1): 16, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38644509

ABSTRACT

Protein-encoding genes only constitute less than 2% of total human genomic sequences, and 98% of genetic information was previously referred to as "junk DNA". Meanwhile, non-coding RNAs (ncRNAs) consist of approximately 60% of the transcriptional output of human cells. Thousands of ncRNAs have been identified in recent decades, and their essential roles in the regulation of gene expression in diverse cellular pathways associated with fundamental cell processes, including proliferation, differentiation, apoptosis, and metabolism, have been extensively investigated. Furthermore, the gene regulation networks they form modulate gene expression in normal development and under pathological conditions. In this review, we integrate current information about the classification, biogenesis, and function of ncRNAs and how these ncRNAs support skeletal development through their regulation of critical genes and signaling pathways in vivo. We also summarize the updated knowledge of ncRNAs involved in common skeletal diseases and disorders, including but not limited to osteoporosis, osteoarthritis, rheumatoid arthritis, scoliosis, and intervertebral disc degeneration, by highlighting their roles established from in vivo, in vitro, and ex vivo studies.


Subject(s)
RNA, Untranslated , Humans , RNA, Untranslated/genetics , Bone Development/genetics , Bone Development/physiology , Bone Diseases/genetics , Animals
12.
Pak J Biol Sci ; 27(3): 113-118, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38686732

ABSTRACT

<b>Background and Objective:</b> Malnutrition and stunting are major unresolved problems in Indonesia. Protein deficiency can cause stunted growth, as well as make physical and cognitive abilities cannot reach their maximum potential. During childhood the need for protein must be fulfilled so that the peak of bone formation during adolescence can be perfect. In malnourished children, a low protein diet will lead to thinning of the bone cortex. Due to the high rate of stunting and malnutrition in children due to protein deficiency, a study was conducted on the effects of feeding low protein diet on rat bones. <b>Materials and Methods:</b> Male Wistar rats (n = 10) at 6-8 weeks old (body weight around 250 g), control groups were fed a normal chow diet and low protein diet groups were given low protein chow diet (protein 5%) for 18 weeks, then the rats were sacrificed and the femoral bones were isolated. Body weight, femur weight, femur length were checked and bone density was examined using X-ray. <b>Results:</b> The body proportions of the low protein group rats were smaller and thinner than those of the control group. This difference is supported by the significant weight loss starting from the sixth week after low protein feeding. There are significant differences in body weight and femur weight between the control and low protein diet groups. Bone density decreases significantly in low protein diet group. Macroscopically, the femur length of the low protein group was shorter than the control group, however the femur length did not show significant differences statistically between the two groups. <b>Conclusion:</b> A low protein diet decreased the body weight of the rats, also causing impaired bone growth characterized by decreasing femur weight. The low protein diet also caused osteoporosis in the bones.


Subject(s)
Bone Density , Diet, Protein-Restricted , Femur , Rats, Wistar , Animals , Male , Femur/metabolism , Rats , Body Weight , Bone Development , Bone and Bones/metabolism , Dietary Proteins/administration & dosage , Dietary Proteins/metabolism
13.
BMC Pediatr ; 24(1): 247, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594697

ABSTRACT

BACKGROUND: Sports practice during adolescence is important to enhance bone development, although it may provide different effects depending on the mechanical impact present in the sport. Besides, resistance training (RT) may also induce bone changes directly (via muscle contractions) and indirectly (via myokines). However, there have been no studies analyzing the longitudinal influence of engaging in sport with and without added mechanical load. Thus, this study aims to analyze the combined effects of sports participation and resistance training on areal bone mineral density (aBMD) accrual in adolescent athletes participating in swimming and impact sports for 12-months. METHODS: This was a 12-month longitudinal study. The sample comprised 91 adolescents (21 females) aged 10 to 18 years, engaged in impact sports (basketball, tennis, track & field, baseball and gymnastics, n = 66) and non-impact sport (swimming, n = 25). The sample was divided according to resistance training participation: impact sports only (n = 45), impact sports + resistance training (n = 21), swimming-only (n = 17) and swimming + resistance training (n = 8). aBMD and soft tissues were measured using dual-energy X-ray absorptiometry. Generalized linear models analysis was used for the resistance training (RT) x type of sport interaction in predicting aBMD changes overtime, adjusting for maturation, sex and baseline aBMD. RESULTS: After 12-months, all groups showed a significant increase in aBMD, except for the swimming groups (regardless of resistant training), which showed a significant loss in spine aBMD (-0.045 [-0.085 to -0.004] g/cm2 in swimming-only and - 0.047 [-0.073 to -0.021] g/cm2 in swimming + RT). In comparisons between groups, only swimming + RT group, compared with swimming-only group presented higher upper limbs aBMD (0.096 g/cm2 [0.074 to 0.118] in swimming + RT vs. 0.046 [0.032 to 0.060] g/cm2 in swimming only; p < 0.05) and whole body less head (WBLH) aBMD (0.039 [0.024 to 0.054] g/cm2 in swimming + RT vs. 0.017 [0.007 to 0.027] g/cm2 swimming-only; p < 0.05). CONCLUSION: Despite the significant gain in aBMD in all groups and body sites after 12-months, except for the spine site of swimmers, the results indicate that participation in RT seems to improve aBMD accrual in swimmers at the upper limbs and WBLH.


Subject(s)
Resistance Training , Swimming , Female , Adolescent , Humans , Swimming/physiology , Longitudinal Studies , Bone Density/physiology , Absorptiometry, Photon/methods , Bone Development/physiology
14.
Physiol Res ; 73(1): 157-172, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38466013

ABSTRACT

Effects of pre/postnatal 2.45 GHz continuous wave (CW), Wireless-Fidelity (Wi-Fi) Microwave (MW) irradiation on bone have yet to be well defined. The present study used biochemical and histological methods to investigate effects on bone formation and resorption in the serum and the tibia bone tissues of growing rats exposed to MW irradiation during the pre/postnatal period. Six groups were created: one control group and five experimental groups subjected to low-level different electromagnetic fields (EMF) of growing male rats born from pregnant rats. During the experiment, the bodies of all five groups were exposed to 2.45 GHz CW-MW for one hour/day. EMF exposure started after fertilization in the experimental group. When the growing male rats were 45 days old in the postnatal period, the control and five experimental groups' growing male and maternal rats were sacrificed, and their tibia tissues were removed. Maternal rats were not included in the study. No differences were observed between the control and five experimental groups in Receptor Activator Nuclear factor-kB (RANK) biochemical results. In contrast, there was a statistically significant increase in soluble Receptor Activator of Nuclear factor-kB Ligand (sRANKL) and Osteoprotegerin (OPG) for 10 V/m and 15 V/m EMF values. Histologically, changes in the same groups supported biochemical results. These results indicate that pre/postnatal exposure to 2.45 GHz EMF at 10 and 15 V/m potentially affects bone development.


Subject(s)
Electromagnetic Fields , Microwaves , Pregnancy , Female , Rats , Animals , Male , Rats, Sprague-Dawley , Microwaves/adverse effects , Electromagnetic Fields/adverse effects , Bone Development
15.
Nutrients ; 16(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474792

ABSTRACT

Colostrum basic protein (CBP) is a trace protein extracted from bovine colostrum. Previous studies have shown that CBP can promote bone cell differentiation and increase bone density. However, the mechanism by which CBP promotes bone activity remains unclear. This study investigated the mechanism of the effect of CBP on bone growth in mice following dietary supplementation of CBP at doses that included 0.015%, 0.15%, 1.5%, and 5%. Compared with mice fed a normal diet, feeding 5% CBP significantly enhanced bone rigidity and improved the microstructure of bone trabeculae. Five-percent CBP intake triggered significant positive regulation of calcium metabolism in the direction of bone calcium accumulation. The expression levels of paracellular calcium transport proteins CLDN2 and CLDN12 were upregulated nearly 1.5-fold by 5% CBP. We conclude that CBP promotes calcium absorption in mice by upregulating the expression of the calcium-transporting paracellular proteins CLND2 and CLND12, thereby increasing bone density and promoting bone growth. Overall, CBP contributes to bone growth by affecting calcium metabolism.


Subject(s)
Calcium , Colostrum , Pregnancy , Female , Animals , Mice , Cattle , Calcium/metabolism , Colostrum/metabolism , Calcium, Dietary/metabolism , Bone and Bones/metabolism , Bone Development , Bone Density , Dietary Proteins/pharmacology
16.
Gene ; 915: 148396, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38552750

ABSTRACT

Family with sequence similarity 20 member C (FAM20C) is a Golgi casein kinase that phosphorylates extracellularly-secreted regulatory proteins involved in bone development and mineralization, but its specific role in bone development is still largely unknown. In this study, to examine the specific mechanisms that FAM20C influences bone development, we cross-bred Osx-Cre with FAM20Cflox/flox mice to establish a Osx-Cre; FAM20Cflox/flox knockout (oKO) mouse model; FAM20C was KO in pre-osteoblasts. oKO development was examined at 1-10 weeks, in which compared to control FAM20Cflox/flox, they had lower body weights and bone tissue mineralization. Furthermore, oKO had lower bone volume fractions, thickness, and trabecular numbers, along with higher degrees of trabecular separation. These mice also had decreased femoral metaphyseal cartilage proliferation layer, along with thickened hypertrophic layer and increased apoptotic cell counts. Transcriptomic analysis found that differentially-expressed genes in oKO were concentrated in the osteoclast differentiation pathway, in line with increased osteoclast presence. Additionally, up-regulation of osteoclast-related, and down-regulation of osteogenesis-related genes, were identified, in which the most up-regulated genes were signal regulatory protein ß-1 family (Sirpb1a-c) and mitogen-activated protein kinase 13. Overall, FAM20C KO in pre-osteoblasts leads to abnormal long bone development, likely due to subsequent up-regulation of osteoclast differentiation-associated genes.


Subject(s)
Bone Development , Calcium-Binding Proteins , Casein Kinase I , Cell Differentiation , Mice, Knockout , Osteoblasts , Osteoclasts , Osteogenesis , Up-Regulation , Animals , Mice , Bone Development/genetics , Casein Kinase I/metabolism , Casein Kinase I/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Male , Female
17.
J Clin Invest ; 134(10)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530358

ABSTRACT

Gender-affirming hormone therapy (GAHT) is often prescribed to transgender (TG) adolescents to alleviate gender dysphoria, but the effect of GAHT on the growing skeleton is unclear. We found GAHT to improve trabecular bone structure via increased bone formation in young male mice and not to affect trabecular structure in female mice. GAHT modified gut microbiome composition in both male and female mice. However, fecal microbiota transfers (FMTs) revealed that GAHT-shaped gut microbiome was a communicable regulator of bone structure and turnover in male, but not in female mice. Mediation analysis identified 2 species of Bacteroides as significant contributors to the skeletal effects of GAHT in male mice, with Bacteroides supplementation phenocopying the effects of GAHT on bone. Bacteroides have the capacity to expand Treg populations in the gut. Accordingly, GAHT expanded intestinal Tregs and stimulated their migration to the bone marrow (BM) in male but not in female mice. Attesting to the functional relevance of Tregs, pharmacological blockade of Treg expansion prevented GAHT-induced bone anabolism. In summary, in male mice GAHT stimulated bone formation and improved trabecular structure by promoting Treg expansion via a microbiome-mediated effect, while in female mice, GAHT neither improved nor impaired trabecular structure.


Subject(s)
Gastrointestinal Microbiome , T-Lymphocytes, Regulatory , Animals , Gastrointestinal Microbiome/drug effects , Mice , Female , Male , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Bone Development/drug effects , Osteogenesis/drug effects , Bacteroides , Fecal Microbiota Transplantation , Humans
18.
JCI Insight ; 9(6)2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38516888

ABSTRACT

Recently, skeletal stem cells were shown to be present in the epiphyseal growth plate (epiphyseal skeletal stem cells, epSSCs), but their function in connection with linear bone growth remains unknown. Here, we explore the possibility that modulating the number of epSSCs can correct differences in leg length. First, we examined regulation of the number and activity of epSSCs by Hedgehog (Hh) signaling. Both systemic activation of Hh pathway with Smoothened agonist (SAG) and genetic activation of Hh pathway by Patched1 (Ptch1) ablation in Pthrp-creER Ptch1fl/fl tdTomato mice promoted proliferation of epSSCs and clonal enlargement. Transient intra-articular administration of SAG also elevated the number of epSSCs. When SAG-containing beads were implanted into the femoral secondary ossification center of 1 leg of rats, this leg was significantly longer 1 month later than the contralateral leg implanted with vehicle-containing beads, an effect that was even more pronounced 2 and 6 months after implantation. We conclude that Hh signaling activates growth plate epSSCs, which effectively leads to increased longitudinal growth of bones. This opens therapeutic possibilities for the treatment of differences in leg length.


Subject(s)
Growth Plate , Hedgehog Proteins , Red Fluorescent Protein , Mice , Rats , Animals , Hedgehog Proteins/metabolism , Bone Development , Stem Cells/metabolism
19.
J Bone Miner Res ; 39(5): 595-610, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38477809

ABSTRACT

Ablation of Cyp27b1 eliminates calcitriol but does not disturb fetal mineral homeostasis or skeletal development. However, independent of fetal genotypes, maternal loss of Cyp27b1 altered fetal mineral and hormonal levels compared to offspring of WT dams. We hypothesized that these maternal influences would alter postnatal skeletal development. Cyp27b1 null and WT females were mated to bear only Cyp27b1+/- offspring. Forty-eight hours after birth, pups were cross-fostered to dams of the same or opposite genotype that bore them. Maternal and offspring samples were collected on days 21 (weaning) and 42. Offspring measurements included minerals and hormones, BMC by DXA, ash weight and mineral content, gene expression, 3-point bending tests, and microCT. Maternal lactational behavior was evaluated. Milk was analyzed for nutritional content. At day 21, offspring fostered by nulls, independent of birth dam, had ~20% lower weight, BMC, ash weight, and ash calcium than pups fostered by WT dams. Adjustment for body weight accounted for the lower BMC but not the lower ash weight and ash calcium. Hormones and serum/urine minerals did not differ across offspring groups. Offspring fostered by nulls had shorter femurs and lower cortical thickness, mean polar moment of inertia, cortical area, trabecular bone volume, and trabecular number. Dam lactational behaviors and milk nutritional content did not differ between groups. At day 42, body weight, ash weight, lengths, BMC, and tibial bone strength were no longer different between pups fostered by null vs WT dams. In summary, pups fostered by Cyp27b1 nulls, regardless of birth dam, have proportionately smaller skeletons at 21 d, impaired microstructure, but normal mineral homeostasis. The skeletal effects are largely recovered by day 42 (3 wk after weaning). In conclusion, maternal loss of calcitriol impairs early postnatal cortical bone growth and trabecular bone mass, but affected offspring catch up after weaning.


Subject(s)
Bone Development , Calcitriol , Animals , Female , Calcitriol/blood , Calcitriol/metabolism , Bone Development/drug effects , Mice , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Bone Density/drug effects , Lactation , Male , Pregnancy , Mice, Knockout , Body Weight/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism
20.
Bone ; 182: 117055, 2024 May.
Article in English | MEDLINE | ID: mdl-38412894

ABSTRACT

The length of long bones is determined by column formation of proliferative chondrocytes and subsequent chondrocyte hypertrophy in the growth plate during bone development. Despite the importance of mechanical loading in long bone development, the mechanical conditions of the cells within the growth plate, such as the stress field, remain unclear owing to the difficulty in investigating spatiotemporal changes within dynamically growing tissues. In this study, the mechanisms of longitudinal bone growth were investigated from a mechanical perspective through column formation of proliferative chondrocytes within the growth plate before secondary ossification center formation using continuum-based particle models (CbPMs). A one-factor model, which simply describes essential aspects of a biological signaling cascade regulating cell activities within the growth plate, was developed and incorporated into CbPM. Subsequently, the developmental process and maintenance of the growth plate structure and resulting bone morphogenesis were simulated. Thus, stress anisotropy in the proliferative zone that affects bone elongation through chondrocyte column formation was identified and found to be promoted by chondrocyte hypertrophy. These results provide further insights into the mechanical regulation of multicellular dynamics during bone development.


Subject(s)
Chondrocytes , Growth Plate , Humans , Anisotropy , Bone Development/physiology , Cell Differentiation , Hypertrophy
SELECTION OF CITATIONS
SEARCH DETAIL
...