1
|
Takata K, Yugami M, Karata S, Karasugi T, Uehara Y, Masuda T, Nakamura T, Tokunaga T, Hisanaga S, Sugimoto K, Yonemitsu R, Ideo K, Fukuma Y, Uragami M, Arima T, Kawakami J, Maeda K, Yoshimura N, Matsunaga H, Kai Y, Tanimura S, Shimada M, Shibata Y, Tateyama M, Takata S, Goshogawa H, Yumoto M, Takashima Y, Inoue S, Ueno S, Kubo R, Tajiri R, Tian X, Honma F, Kawamura Y, Miyamoto T. Plates made from magnesium alloy with a long period stacking ordered structure promote bone formation in a rabbit fracture model. Sci Rep 2025; 15:12210. [PMID: 40204858 PMCID: PMC11982537 DOI: 10.1038/s41598-025-96853-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Operative treatment is an option for fractures when the fracture is unstable or the patient wishes to return early to daily life or social activities. Metal plates such as titanium and stainless steel are often used in fracture surgery, but the metal plate lacks bone-healing activity and is not bioabsorbable, requiring a second surgery to remove it after bone union. Here we show that a magnesium (Mg) plate made from an alloy of yttrium, zinc, and aluminum with magnesium as the main component in a long-period stacking ordered structure promotes bone formation in a rabbit tibia fracture model and is also bioabsorbable. We show that the Mg plate significantly promoted bone and callus formation compared to a titanium plate in the rabbit tibia fracture model. Moreover, the Mg plate was mostly bioabsorbed once bone union was achieved, but rabbits showed no evidence of biotoxic effects, such as weight loss or increased blood magnesium levels. We also demonstrate that treatment with exogenous magnesium significantly enhanced calcium deposition in an in vitro osteoblast culture system. Magnesium is an essential element, and its radiolucency facilitates observation of the fracture site during Mg plate fixation, while its lack of magnetic properties allows its use in patients who require MRI scans. Accordingly, we propose that a use of a Mg plate could be beneficial in treating bone fracture.
Collapse
Affiliation(s)
- Kosei Takata
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Masaki Yugami
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Soichiro Karata
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Tatsuki Karasugi
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yusuke Uehara
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Tetsuro Masuda
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takayuki Nakamura
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takuya Tokunaga
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Satoshi Hisanaga
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kazuki Sugimoto
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Ryuji Yonemitsu
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Katsumasa Ideo
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yuko Fukuma
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Masaru Uragami
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takahiro Arima
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Jyunki Kawakami
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kazuya Maeda
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Naoto Yoshimura
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Hideto Matsunaga
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yuki Kai
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Shuntaro Tanimura
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Masaki Shimada
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yuto Shibata
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Makoto Tateyama
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Shu Takata
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Hikaru Goshogawa
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Mizuho Yumoto
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yusuke Takashima
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Shinichi Inoue
- Magnesium Research Center & Department of Material Science, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-85565, Japan
| | - Syotaro Ueno
- Magnesium Research Center & Department of Material Science, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-85565, Japan
| | - Ryuta Kubo
- Department of Oral and Maxillofacial Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Rui Tajiri
- Department of Oral and Maxillofacial Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Xiao Tian
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Fuka Honma
- Department of Oral and Maxillofacial Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yoshihito Kawamura
- Magnesium Research Center & Department of Material Science, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-85565, Japan
| | - Takeshi Miyamoto
- Faculty of Life Sciences, Department of Orthopedic Surgery, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
| |
Collapse
|
2
|
Harissa Z, Kim Y, Dicks AR, Steward N, Guilak F. Skeletal dysplasia-causing mutations in TRPV4 alter the chondrocyte transcriptomic response to mechanical loading. Am J Physiol Cell Physiol 2025; 328:C1135-C1149. [PMID: 40019039 DOI: 10.1152/ajpcell.01066.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a mechanosensitive ion channel highly expressed in chondrocytes that supports cartilage development and homeostasis. Mutations in the channel can cause skeletal dysplasias, including the gain-of-function mutations V620I and T89I, which lead to brachyolmia and metatropic dysplasia, respectively. These mutations suppress hypertrophic differentiation, but the mechanisms by which they alter chondrocyte response to mechanical load remain to be elucidated. To determine the effect of these mutations on chondrocyte mechanotransduction, tissue-engineered cartilage was derived from differentiated clustered regularly interspaced short palindromic repeats (CRISPR)-edited human-induced pluripotent stem cells (hiPSCs) harboring the moderate V620I or severe T89I TRPV4 mutations. Wild-type and mutant tissue-engineered hiPSC-derived cartilage contructs were subjected to compressive mechanical loading at physiological levels, and transcriptomic signatures were assessed by RNA-sequencing. Our results demonstrate that the V620I and T89I mutations diminish the mechanoresponsiveness of chondrocytes, as evidenced by reduced gene expression downstream of TRPV4 activation, including those involved in endochondral ossification. Changes in the expression of genes involved in extracellular matrix production and organization were found to contribute toward the phenotype in V620I mutant chondrocytes, whereas dysregulated retinoic acid signaling was linked to T89I, and disrupted proliferation was common to both. Our findings suggest that dysfunctional mechanotransduction due to V620I and T89I mutations in TRPV4 contribute to the developmental phenotypes, supporting TRPV4 modulation as a potential pharmacologic target.NEW & NOTEWORTHY Gain-of-function mutations in TRPV4, a mechano- and osmosensitive ion channel, are linked to skeletal dysplasias, but their effects on chondrocyte mechanotransduction remain unknown. Using human iPSCs harboring skeletal dysplasia-causing mutations, we developed and mechanically loaded tissue-engineered cartilage. Our findings show that V620I and T89I mutations reduce chondrocyte mechanoresponsiveness, evidenced by decreased gene expression downstream of TRPV4 activation, providing insight into TRPV4-related skeletal disorders and potential pharmacological targets.
Collapse
Affiliation(s)
- Zainab Harissa
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Yuseon Kim
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Amanda R Dicks
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
3
|
Bergamasco MI, Ogier JM, Garnham AL, Whitehead L, Rogers K, Smyth GK, Burt RA, Voss AK, Thomas T. Loss of KAT6B causes premature ossification and promotes osteoblast differentiation during development. Dev Biol 2025; 520:141-154. [PMID: 39832706 DOI: 10.1016/j.ydbio.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
The MYST family histone acetyltransferase gene, KAT6B (MYST4, MORF, QKF) is mutated in two distinct human congenital disorders characterised by intellectual disability, facial dysmorphogenesis and skeletal abnormalities; the Say-Barber-Biesecker-Young-Simpson variant of Ohdo syndrome and Genitopatellar syndrome. Despite its requirement in normal skeletal development, the cellular and transcriptional effects of KAT6B in skeletogenesis have not been thoroughly studied. Here, we show that germline deletion of the Kat6b gene in mice causes premature ossification in vivo, resulting in shortened craniofacial elements and increased bone density, as well as shortened tibias with an expanded pre-hypertrophic layer, as compared to wild type controls. Mechanistically, we show that the loss of KAT6B in mesenchymal progenitor cells promotes transition towards an osteoblast-progenitor state with upregulation of gene targets of RUNX2, a master regulator of osteoblast development and concomitant downregulation of SOX9, a critical gene in chondrocyte development. Moreover, we find that compound heterozygosity at Kat6b and Runx2 loci partially rescues the reduction in ossification of Runx2 heterozygous, but not homozygous mice, suggesting that KAT6B may limit the action of RUNX2, possibly through a role in maintaining progenitors in an undifferentiated state. Moreover, our results show that KAT6B has essential roles in regulating the expression of a large number of genes involved in skeletogenesis and bone development.
Collapse
Affiliation(s)
- Maria I Bergamasco
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Jacqueline M Ogier
- The Department of Audiology and Speech Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Kelly Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Rachel A Burt
- Department of Genetics, The Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, 3052, Australia
| | - Anne K Voss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
4
|
Cai C, Ma Y, Zhang L, An Z, Zhou E, Liu X, Li H, Li W, Li Z, Li G, Liu X, Zhang Y, Han R. Genome-wide methylation and transcriptome differential analysis of skeletal muscle in broilers with valgus-varus deformity. Br Poult Sci 2025; 66:175-186. [PMID: 39504239 DOI: 10.1080/00071668.2024.2410368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/03/2024] [Indexed: 11/08/2024]
Abstract
1. Valgus-varus deformity (VVD) is a disease that severely affects leg function in broilers and for which there is no effective control method current available. Although DNA methylation has an important impact on most physiological and pathological processes, its involvement in skeletal muscle growth and development in VVD broilers is unknown. In this study, genome-wide DNA methylation was analysed in VVD-affected and normal broilers using whole genome resulphite sequencing.2. The results showed that in the cytosine-phosphoric acid-guanine (CG) sequence environment there was a methylation rate of about 55% and 4,265 differentially methylated regions (DMRs) were found in the CG. Of these, 550 were located in the promoter, 547 in the exon region, and 1,718 in the intron region.3. All differentially methylated genes (DMGs) were analysed for enrichment of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. The GO was enriched in pathways related to protein degradation such as proteasome complex, endopeptidase complex and extracellular region. The KEGG pathways were enriched in signalling pathways related to protein degradation and catabolism such as proteasome, nitrogen metabolism, adherens junction and alanine.4. Protein interactions analysis revealed that FOS, MYL9, and FRAS1 had a high degree of interactions, in which the DNA methylation level of the MYL9 promoter region was negatively correlated with mRNA expression level. Further studies showed that 5-azacytidine (5-AzaC) inhibited DNMT1 and DNMT3A gene expression and promoted MYL9 expression.5. This study systematically investigated overall DNA methylation patterns in the leg muscle of VVD and normal broilers. It screened common differential genes in conjunction with transcriptomic data to further identify genes associated with muscle growth and development. This study provides new insights to better understand the pathogenesis of VVD from an epigenetic perspective.
Collapse
Affiliation(s)
- C Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| | - Y Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| | - L Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| | - Z An
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| | - E Zhou
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| | - X Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| | - H Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - W Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| | - Z Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - G Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| | - X Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Y Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - R Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- The Shennong Laboratory, Zhengzhou, China
| |
Collapse
|
5
|
Thakore P, Delany AM. miRNA-based regulation in growth plate cartilage: mechanisms, targets, and therapeutic potential. Front Endocrinol (Lausanne) 2025; 16:1530374. [PMID: 40225327 PMCID: PMC11985438 DOI: 10.3389/fendo.2025.1530374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
MicroRNAs (miRNAs) are critical regulators of the skeleton. In the growth plate, these small non-coding RNAs modulate gene networks that drive key stages of chondrogenesis, including proliferation, differentiation, extracellular matrix synthesis and hypertrophy. These processes are orchestrated through the interaction of pivotal pathways including parathyroid hormone-related protein (PTHrP), Indian hedgehog (IHH), and bone morphogenetic protein (BMP) signaling. This review highlights the miRNA-mRNA target networks essential for chondrocyte differentiation. Many miRNAs are differentially expressed in resting, proliferating and hypertrophic cartilage zones. Moreover, differential enrichment of specific miRNAs in matrix vesicles is also observed, providing means for chondrocytes to influence the function and differentiation of their neighbors by via matrix vesicle protein and RNA cargo. Notably, miR-1 and miR-140 emerge as critical modulators of chondrocyte proliferation and hypertrophy by regulating multiple signaling pathways, many of them downstream from their mutual target Hdac4. Demonstration that a human gain-of-function mutation in miR-140 causes skeletal dysplasia underscores the clinical relevance of understanding miRNA-mediated regulation. Further, miRNAs such as miR-26b have emerged as markers for skeletal disorders such as idiopathic short stature, showcasing the translational relevance of miRNAs in skeletal health. This review also highlights some miRNA-based therapeutic strategies, including innovative delivery systems that could target chondrocytes via cartilage affinity peptides, and potential applications related to treatment of physeal bony bridge formation in growing children. By synthesizing current research, this review offers a nuanced understanding of miRNA functions in growth plate biology and their broader implications for skeletal health. It underscores the translational potential of miRNA-based therapies in addressing skeletal disorders and aims to inspire further investigations in this rapidly evolving field.
Collapse
|
6
|
Zhao B, Suh J, Zhang Y, Yin E, Kadota-Watanabe C, Chang IW, Yaung J, Lao-Ngo I, Young NM, Kim RH, Klein OD, Hong C. p75 neurotrophin receptor regulates craniofacial growth and morphology in postnatal development. Front Cell Dev Biol 2025; 13:1569533. [PMID: 40171227 PMCID: PMC11959563 DOI: 10.3389/fcell.2025.1569533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
Craniofacial abnormalities are among the most prevalent congenital defects, significantly affecting appearance, function, and quality of life. While the role of genetic mutations in craniofacial malformations is recognized, the underlying molecular mechanisms remain poorly understood. In this study, we investigate the role of p75 neurotrophin receptor (p75NTR) in craniofacial development by comparing wild-type (p75NTR+/+) mice against p75NTR-deficient (p75NTR-/-) knockout mice. We employed histology, micro-CT surface distance, volumetric analysis, and geometric morphometric analysis to assess craniofacial development and growth. On postnatal day 7 (P7), p75NTR-/- mice exhibited reduced skull length compared to wild-type controls. By P28, micro-CT analysis revealed significant reductions in calvarial bone volume and trabecular bone thickness in p75NTR-/- mice. Geometric morphometric analysis identified significant shape alterations in the nasal, parietal, and occipital regions, with p75NTR-/- mice showing a shortened cranium and tapered nasal bone morphology. These findings highlight the critical role of p75NTR in regulating postnatal craniofacial development. Disruption of p75NTR signaling impairs both the growth and morphological integrity of craniofacial structures, which may contribute to the pathogenesis of congenital craniofacial abnormalities. In the future, a better understanding of the molecular mechanisms through which p75NTR mediates craniofacial development may offer valuable insights for future targeted therapeutic strategies for craniofacial defects.
Collapse
Affiliation(s)
- Byron Zhao
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Jinsook Suh
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Yan Zhang
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Eric Yin
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Chiho Kadota-Watanabe
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
- Division of Maxillofacial and Neck Reconstruction, Department of Maxillofacial Orthognathics, Institute of Science Tokyo, Tokyo, Japan
| | - In Won Chang
- Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jun Yaung
- Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Isabelle Lao-Ngo
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Nathan M. Young
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Reuben H. Kim
- Shapiro Family Laboratory of Viral Oncology and Aging Research, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ophir D. Klein
- Department of Orofacial Sciences, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, Los Angeles, CA, United States
| | - Christine Hong
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
7
|
Hu Z, Wang C, Wang C, He J, Yan Y, Xu Z, Yu Y, Yu Y, Cheng H, Liu L, Tang M, Zhang C, Yu H, Jing J, Cheng W. The comparative efficacy of L-glutamine, celecoxib, and glucosamine sulfate in osteoarthritis management. Sci Rep 2025; 15:8992. [PMID: 40089639 PMCID: PMC11910618 DOI: 10.1038/s41598-025-93357-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
To explore the therapeutic efficacy of L-glutamine (L-Gln) on pathological progression and clinical symptoms of osteoarthritis (OA), and compare with glucosamine sulfate (GS), and celecoxib (CXB). Rats were administered sodium chloride, L-Gln, GS, or CXB via gavage for eight weeks starting from the fifth week after sham operation or Anterior Cruciate Ligament Transection (ACLT) + Medial Meniscectomy (MMx). Then the severity of knee OA in rats was evaluated by serological analysis, histological examination and imaging examination. In addition, patients with mild primary OA were administered L-Gln, GS, or CXB orally for 12 weeks in accordance with the randomization principle. The efficacy end points were the change from baseline to week 24 in the pain and physical function subscale scores of the Western Ontario and McMaster Universities OA Index (WOMAC), and Lequesne score. Treatment with L-Gln alleviated the increased concentration of serum cartilage degradation markers caused by OA in rats. Histological tests showed improvement in knee joint cartilage destruction after treatment. Three-dimensional CT scans and reconstructions revealed a reduction in osteophyte formation and subchondral bone loss. L-glutamine performed as well as or better than glucosamine sulfate and celecoxib in all comparative measures among the three treatment groups. In clinical trials, the WOMAC pain and physical function subscale scores, as well as the Lequesne score, decreased from baseline in all three patient groups during follow-up, with no significant differences observed between the groups. Our research indicates that L-Gln is comparable to GS and CXB in improving the pathological progression and clinical efficacy of OA, which makes it a promising drug for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Zhongyao Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | | | - Chen Wang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Junyan He
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yiqun Yan
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zelin Xu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yangmang Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ya Yu
- Huoshan County Hospital, Lu'an, 237299, Anhui, China
| | - Huan Cheng
- Huoshan County Hospital of Traditional Chinese Medicine, Lu'an, 237200, Anhui, China
| | - Lei Liu
- Suzhou Municipal Hospital in Anhui Province, Suzhou, 234000, Anhui, China
| | - Miao Tang
- Suzhou Municipal Hospital in Anhui Province, Suzhou, 234000, Anhui, China
| | - Chun Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Haoran Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Wendan Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
8
|
Reno PL, Wallace S, Doelp SN, Biancaniello M, Kjosness KM. The role of the PTHrP/Ihh feedback loop in the unusual growth plate location in mammalian metatarsals and pisiforms. Dev Dyn 2025. [PMID: 40088130 DOI: 10.1002/dvdy.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Longitudinal skeletal growth takes place in the cartilaginous growth plates. While growth plates are found at either end of conventional long bones, they occur at a variety of locations in the mammalian skeleton. For example, the metacarpals and metatarsals (MT) in the hands and feet form only a single growth plate at one end, and the pisiform in the wrist is the only carpal bone to contain a growth plate. We take advantage of this natural anatomical variation to test which components of the PTHrP/Ihh feedback loop, a fundamental regulator of chondrocyte differentiation, are specific to growth plate function. RESULTS Parathyroid hormone-like hormone (Pthlh), the gene that transcribes parathyroid hormone-related peptide (PTHrP), is expressed in the reserve zone of the growth plate-forming end of the MT. At the opposite end, the absence of a PTHrP+ reserve zone results in premature chondrocyte differentiation and Indian hedgehog (Ihh) expression. Pthlh is expressed in the reserve zone of the developing pisiform, confirming the existence of a true growth plate. CONCLUSION A pool of PTHrP+ reserve zone chondrocytes is a defining characteristic of growth plates, and its patterning may be key to evolved differences in growth plate location in the mammalian skeleton.
Collapse
Affiliation(s)
- Philip L Reno
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | - Sherrie Wallace
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | - Sarah N Doelp
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | - Maria Biancaniello
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | - Kelsey M Kjosness
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Rizzo MG, Briglia M, Zammuto V, Morganti D, Faggio C, Impellitteri F, Multisanti CR, Graziano ACE. Innovation in Osteogenesis Activation: Role of Marine-Derived Materials in Bone Regeneration. Curr Issues Mol Biol 2025; 47:175. [PMID: 40136429 PMCID: PMC11941683 DOI: 10.3390/cimb47030175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Marine-derived biomaterials are emerging as promising candidates for tissue regeneration due to their sustainability, biocompatibility, bioactivity, and unique chemical structure. This review provides an overview of different marine-derived inorganic and organic materials, such as calcium carbonate, magnesium salts, silica, polysaccharides, bioactive peptides, and lipid-based compounds, and their effects in promoting osteogenesis. Specifically, the osteoinductive, osteoconductive, and osteointegrative activities of traditional and innovative materials that influence key molecular pathways such as BMP/Smad and Wnt/β-catenin signaling underlying bone formation will be evaluated. This review also prospects innovative approaches, i.e., phage display technology, to optimize marine-derived peptides for targeted bone regeneration. In the context of innovative and sustainable materials, this review suggests some interesting applications of unusual materials able to overcome the limitations of conventional ones and stimulate cellular regeneration of bone tissue by activating specific molecular pathways.
Collapse
Affiliation(s)
- Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy;
| | - Marilena Briglia
- Department of Medicine and Surgery, “Kore” University of Enna, 94100 Enna, Italy; (M.B.); (A.C.E.G.)
| | - Vincenzo Zammuto
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy;
| | - Dario Morganti
- Consiglio Nazionale delle Ricerche DSFTM, Department of Physical Sciences and Technologies of Matter, Piazzale Aldo Moro, 7, 00185 Roma, Italy
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy;
- Department of Eco-Sustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, 80122 Naples, Italy
| | - Federica Impellitteri
- Department of Veterinary Sciences, University of Messina, Viale Giovanni Palatucci, 98168 Messina, Italy; (F.I.); (C.R.M.)
| | - Cristiana Roberta Multisanti
- Department of Veterinary Sciences, University of Messina, Viale Giovanni Palatucci, 98168 Messina, Italy; (F.I.); (C.R.M.)
| | | |
Collapse
|
10
|
Zhang G, Moya A, Scherberich A, Martin I. Challenges of engineering a functional growth plate in vitro. Front Bioeng Biotechnol 2025; 13:1550713. [PMID: 40104770 PMCID: PMC11913844 DOI: 10.3389/fbioe.2025.1550713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Several cartilage and bone organoids have been developed in vitro and in vivo using adult mesenchymal stromal/stem cells (MSCs) or pluripotent stem cells (PSCs) to mimic different phases of endochondral ossification (ECO), as one of the main processes driving skeletal development and growth. While cellular and molecular features of growth plate-like structures have been observed through the generation and in vivo implantation of hypertrophic cartilage tissues, no functional analogue or model of the growth plate has yet been engineered. Herein, after a brief introduction about the growth plate architecture and function, we summarize the recent progress in dissecting the biology of the growth plate and indicate the knowledge gaps to better understand the mechanisms of its development and maintenance. We then discuss how this knowledge could be integrated with state-of-art bioengineering approaches to generate a functional in vitro growth plate model.
Collapse
Affiliation(s)
- Gangyu Zhang
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Adrien Moya
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| |
Collapse
|
11
|
Kodama J, Oichi T, Wilkinson KJ, Abzug JM, Kaito T, Enomoto-Iwamoto M, Iwamoto M, Otsuru S. Apolipoprotein E is a marker of all chondrocytes in the growth plate resting zone. Bone Res 2025; 13:31. [PMID: 40025030 PMCID: PMC11873292 DOI: 10.1038/s41413-025-00407-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 03/04/2025] Open
Abstract
The resting zone (RZ) in mammalian growth plates is critical for maintaining and regulating chondrocyte turnover during longitudinal bone growth as a control tower and stem cell reservoir. Although recent lineage tracing studies have identified several markers for stem cells in the RZ, these markers only partially label chondrocytes in the RZ, suggesting that the resting chondrocytes (RCs) are a heterogeneous population with different types of stem cells. Since a comprehensive marker for RCs is still lacking, the RZ is generally determined based on ambiguous histological criteria, such as small and round chondrocytes without columnar formation, which may lead to inconsistencies among researchers. Therefore, in this study, we used single-cell RNA sequencing (scRNAseq) of growth plate chondrocytes followed by validation by fluorescence in situ hybridization (FISH) to precisely annotate cell clusters in scRNAseq and search for a marker of RCs. The scRNAseq analysis revealed that apolipoprotein E (Apoe) was the top-hit gene, which was ubiquitously expressed in the RC cluster. FISH confirmed that Apoe was exclusively localized to the histologically defined RZ. In newly generated ApoemCherry knock-in mice, we further confirmed that mCherry expression mirrored the distribution of Apoe-expressing chondrocytes in the RZ particularly after the formation of the secondary ossification center. These mCherry+ RCs were slow cycling in vivo and exhibited stem cell properties in vitro. Moreover, APOE was detected in human growth plate RCs. These findings suggest that apolipoprotein E is a novel pan-RC marker in both mouse and human growth plates.
Collapse
Affiliation(s)
- Joe Kodama
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Takeshi Oichi
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kevin J Wilkinson
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joshua M Abzug
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Masahiro Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Satoru Otsuru
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Alzarka B, Charnaya O, Gunay-Aygun M. Diseases of the primary cilia: a clinical characteristics review. Pediatr Nephrol 2025; 40:611-627. [PMID: 39340573 DOI: 10.1007/s00467-024-06528-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024]
Abstract
Ciliopathies encompass a broad spectrum of diseases stemming from dysfunction of the primary (non-motile) cilia, present on almost all cells in the human body. These disorders include autosomal dominant and recessive polycystic kidney diseases, nephronophthisis, and multisystem ciliopathies such as Joubert, Meckel, Bardet-Biedl, Alström, oral-facial-digital syndromes, and skeletal ciliopathies. The majority of these ciliopathies are associated with fibrocystic kidney disease resulting in progressive kidney dysfunction. In addition, many ciliopathies are associated with extra-renal manifestations including congenital hepatic fibrosis, retinal dystrophy, obesity, and brain and skeletal anomalies. The diagnoses may be challenging due to their overlapping clinical features and molecular heterogeneity. To date, over 190 genes encoding proteins that localize to the primary cilia have been identified as disease-causing. This review will discuss the clinical features of the most frequently encountered disorders of primary cilia.
Collapse
Affiliation(s)
- Bakri Alzarka
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Olga Charnaya
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Meral Gunay-Aygun
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA.
| |
Collapse
|
13
|
Wang J, Liu W, Zhang T, Cui M, Gao K, Lu P, Yao S, Cao Z, Zheng Y, Tian W, Li Y, Yin R, Hu J, Han G, Liang J, Zhou F, Chai J, Zhang H. An epitranscriptomic program maintains skeletal stem cell quiescence via a METTL3-FEM1B-GLI1 axis. EMBO J 2025:10.1038/s44318-025-00399-z. [PMID: 40016417 DOI: 10.1038/s44318-025-00399-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
Skeletal stem cells (SSCs) maintain the skeletal system via pluripotency and differentiation capacity. However, it remains largely unknown how these cells precisely regulate their function to maintain skeletal organization. Here, we delineate the RNA m6A modification landscape across skeletal cell populations in the mouse epiphysis. Our findings show that m6A modifications are prevalent in skeletal stem cell and progenitor populations and play critical roles in cell fate determination. Genetic deletion of Mettl3, the core catalytic subunit of the m6A-methyltransferase complex, in murine skeletal stem and progenitors impaired bone development, leading to shortened limbs, disrupted growth plate zonation, and decreased bone mass. Moreover, Mettl3 deficiency induced quiescence exit in SSCs, together with compromised self-renewal capacity and differentiation potential. Mechanistically, Mettl3-mediated m6A modification reduced mRNA stability of the Cul2-RING E3 ligase complex subunit Fem1b, which subsequently stabilizes Gli1 protein, a key transcription factor of Hedgehog pathway for maintaining SSC identity and function. Thus, we present a comprehensive RNA m6A modification landscape of skeletal cell hierarchy and uncover the essential function of epitranscriptomically-regulated proteostasis in maintaining SSCs quiescence and potency.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weidong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Tiantian Zhang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Manman Cui
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Kexin Gao
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Pengbo Lu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Shuxin Yao
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Ziyan Cao
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yanbing Zheng
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Wen Tian
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Yan Li
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Rong Yin
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jin Hu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Guoqiang Han
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jianfei Liang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 710004, Xi'an, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jihua Chai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Haojian Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China.
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- RNA Institute, Wuhan University, Wuhan, China.
| |
Collapse
|
14
|
Midla SC, Omi-Sugihara M, Cha M, Chen C, Cavalcante RC, Pan H, Mishina Y, Ueharu H. Augmented Bone Morphogenetic Protein Signaling During TMJ Development Alters Morphology in a Timepoint-Dependent Manner. Int J Mol Sci 2025; 26:1655. [PMID: 40004119 PMCID: PMC11855487 DOI: 10.3390/ijms26041655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
The temporomandibular joint (TMJ) is unique in both developmental origin and functional maintenance. The role of bone morphogenic protein (BMP) signaling in endochondral ossification has been widely investigated but not in the context of the TMJ. We employed a histomorphometric analysis approach to understand how augmented BMP signaling in the cranial neural crest affects the postnatal development of the TMJ. Our analysis showed that cartilage length in the mandibular condyle was reduced in Wnt1 Cre;caBmpr1a mice before the weaning stage (P17). However, following weaning, the mandibular condylar cartilage showed recovered length (P28 and P42). Furthermore, the changes in cartilage length coincide with alterations in cell death in the superficial region of the mandibular condyle. These results suggest that BMP signaling influences chondrocyte cell death and TMJ development in a timepoint-specific manner.
Collapse
Affiliation(s)
- Susannah C. Midla
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; (S.C.M.); (M.O.-S.); (M.C.); (C.C.); (R.C.C.); (H.P.)
| | - Maiko Omi-Sugihara
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; (S.C.M.); (M.O.-S.); (M.C.); (C.C.); (R.C.C.); (H.P.)
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Madeline Cha
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; (S.C.M.); (M.O.-S.); (M.C.); (C.C.); (R.C.C.); (H.P.)
| | - Coral Chen
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; (S.C.M.); (M.O.-S.); (M.C.); (C.C.); (R.C.C.); (H.P.)
| | - Rafael Correia Cavalcante
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; (S.C.M.); (M.O.-S.); (M.C.); (C.C.); (R.C.C.); (H.P.)
| | - Haichun Pan
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; (S.C.M.); (M.O.-S.); (M.C.); (C.C.); (R.C.C.); (H.P.)
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; (S.C.M.); (M.O.-S.); (M.C.); (C.C.); (R.C.C.); (H.P.)
| | - Hiroki Ueharu
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; (S.C.M.); (M.O.-S.); (M.C.); (C.C.); (R.C.C.); (H.P.)
| |
Collapse
|
15
|
Liu Y, Zhang ZX, Fu CS, Ye ZB, Jin HM, Yang XH. Association of aberrant mineral metabolic markers with fracture risk in chronic kidney disease: a comprehensive meta-analysis. BMC Nephrol 2025; 26:68. [PMID: 39934684 DOI: 10.1186/s12882-025-03992-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND This meta-analysis aims to investigate the impact of abnormalities in mineral metabolic markers, including serum phosphate and calcium, intact parathyroid hormone (iPTH), and fibroblast growth factor 23 (FGF23) on the risk of fractures in patients with chronic kidney disease (CKD). METHODS A systematic search was conducted across MEDLINE, Web of Science, EMBASE, ClinicalTrials.gov, and the Cochrane Central Register for Controlled Trials. The outcomes were association of mineral metabolic markers with the risk of fractures in patients with chronic kidney disease. Pooled risk estimates and 95% confidence intervals (CIs) were calculated using fixed-effects or random-effects models. RESULTS Thirty-two studies were included in the meta-analysis. High and low levels of serum phosphate in hemodialysis (HD) patients were both associated with an increased risk of fractures (RR = 1.08, 95% CI 1.02-1.15, P = 0.013; RR = 1.13, 95% CI 1.02-1.25, P = 0.022, respectively). Similarly, abnormal levels of iPTH in CKD patients, both high and low, were associated with increased fracture risk (RR = 1.25, 95% CI 1.20-1.31, P < 0.001; RR = 1.41, 95% CI 1.10-1.82, P = 0.007, respectively). Elevated FGF23 levels were also linked to an increased risk of fractures (RR = 1.32, 95% CI 1.06-1.66, P = 0.015). While a higher level of calcium exhibited a trend towards reduced fracture incidence without statistical significance (RR = 0.90, 95% CI 0.77-1.05, P = 0.181), lower calcium levels tended to increase fracture risk without statistical significance (RR = 1.11, 95% CI 0.99-1.24, P = 0.087). Notably, subjects treated with calcium and phosphorus modulating drugs demonstrated a statistically significant reduction in fractures among CKD patients undergoing dialysis (phosphate binders, RR = 0.79, 95% CI 0.70-0.89; cinacalcet, RR = 0.74, 95% CI 0.59-0.93; vitamin D analogues, RR = 0.82, 95% CI 0.74-0.92, respectively). CONCLUSION This meta-analysis underscores the association between abnormal mineral metabolic markers, including high serum phosphate, iPTH, and FGF23, and an increased risk of fractures in CKD patients. Notably, both elevated and decreased levels of phosphate and iPTH contribute to fracture risk. The efficacy of active vitamin D, phosphorus binders, and cinacalcet in preventing fractures was observed in HD patients but not in the non-dialysis CKD population. TRIAL REGISTRATION PROSPERO CRD42023493951.
Collapse
Affiliation(s)
- Yao Liu
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Zhen Xing Zhang
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Chen Sheng Fu
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, 200040, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Zhi Bin Ye
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, 200040, China.
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, China.
| | - Hui Min Jin
- Department of Nephrology, the People's Hospital of Wenshan Prefecture, Yunnan Province, China.
- Department of Nephrology, Pudong Medical Center, Shanghai Pudong Hospital, Fudan University, 2800 Gong Wei Road, Shanghai, China.
| | - Xiu Hong Yang
- Department of Nephrology, Huadong Hospital, Fudan University, Shanghai, 200040, China.
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
16
|
Perepletchikova D, Malashicheva A. Communication between endothelial cells and osteoblasts in regulation of bone homeostasis: Notch players. Stem Cell Res Ther 2025; 16:56. [PMID: 39920854 PMCID: PMC11806792 DOI: 10.1186/s13287-025-04176-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
Endothelial cells coat blood vessels and release molecular signals to affect the fate of other cells. Endothelial cells can adjust their behavior in response to the changing microenvironmental conditions. During bone regeneration, bone tissue cells release factors that promote blood vessel growth. Notch is a key signaling that regulates cell fate decisions in many tissues and plays an important role in bone tissue development and homeostasis. Understanding the interplay between angiogenesis and osteogenesis is currently a focus of research efforts in order to facilitate and improve osteogenesis when needed. Our review explores the cellular and molecular mechanisms including Notch-dependent endothelial-MSC communication that drive osteogenesis-angiogenesis processes and their effects on bone remodeling and repair.
Collapse
Affiliation(s)
| | - Anna Malashicheva
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia, 194064.
| |
Collapse
|
17
|
Zhao X, Yao M, Wang Y, Feng C, Yang Y, Tian L, Bao C, Li X, Zhu X, Zhang X. Neuroregulation during Bone Formation and Regeneration: Mechanisms and Strategies. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7223-7250. [PMID: 39869030 DOI: 10.1021/acsami.4c16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The skeleton is highly innervated by numerous nerve fibers. These nerve fibers, in addition to transmitting information within the bone and mediating bone sensations, play a crucial role in regulating bone tissue formation and regeneration. Traditional bone tissue engineering (BTE) often fails to achieve satisfactory outcomes when dealing with large-scale bone defects, which is frequently related to the lack of effective reconstruction of the neurovascular network. In recent years, increasing research has revealed the critical role of nerves in bone metabolism. Nerve fibers regulate bone cells through neurotransmitters, neuropeptides, and peripheral glial cells. Furthermore, nerves also coordinate with the vascular and immune systems to jointly construct a microenvironment favorable for bone regeneration. As a signaling driver of bone formation, neuroregulation spans the entire process of bone physiological activities from the embryonic formation to postmaturity remodeling and repair. However, there is currently a lack of comprehensive summaries of these regulatory mechanisms. Therefore, this review sketches out the function of nerves during bone formation and regeneration. Then, we elaborate on the mechanisms of neurovascular coupling and neuromodulation of bone immunity. Finally, we discuss several novel strategies for neuro-bone tissue engineering (NBTE) based on neuroregulation of bone, focusing on the coordinated regeneration of nerve and bone tissue.
Collapse
Affiliation(s)
- Xiangrong Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meilin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuyi Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
18
|
Stoop J, Yokoyama Y, Adachi T. Timing of resting zone parathyroid hormone-related protein expression affects maintenance of the growth plate during secondary ossification: a computational study. Biomech Model Mechanobiol 2025; 24:125-137. [PMID: 39549120 PMCID: PMC11846766 DOI: 10.1007/s10237-024-01899-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/17/2024] [Indexed: 11/18/2024]
Abstract
Secondary ossification and maintenance of the growth plate are crucial aspects of long bone formation. Parathyroid hormone-related protein (PTHrP) has been implicated as a key factor in maintaining the growth plate, and studies suggest that PTHrP expression in the resting zone is closely related with formation of the secondary ossification center (SOC). However, details of the relationship between resting zone PTHrP expression and preservation of the growth plate remain unclear. In this study, we aim to investigate the role of resting zone PTHrP expression on maintenance of the growth plate using a computational method. We extend an existing continuum-based particle model of tissue morphogenesis to include PTHrP and Indian hedgehog (Ihh) signaling, allowing the model to capture biochemical and mechanical regulation of individual cell activities. Our model indicates that the timing of resting zone PTHrP expression-specifically the rate of increase in production at the onset of SOC formation-is potentially a crucial mechanism for maintenance of the growth plate.
Collapse
Affiliation(s)
- Jorik Stoop
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Yuka Yokoyama
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Taiji Adachi
- Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
- Department of Medicine and Medical Science, Graduate School of Medicine, Kyoto University, 53 Shogoin-Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
19
|
Bao M, Wang X, Li X, Sun R, Wang Z, Jiang T, Wang H, Feng J. Single-Cell Landscape of the Cochlea Revealed Cell-Type-Specific Diversification in Hipposideros armiger Based on PacBio Long-Read Sequencing. Biomolecules 2025; 15:211. [PMID: 40001514 PMCID: PMC11853400 DOI: 10.3390/biom15020211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Echolocation represents one of the most rapid adaptive sensorimotor modulation behaviors observed in mammals, establishing bats as one of the most evolutionarily successful mammals. Bats rely on high-frequency hearing for survival, but our understanding of its cellular molecular basis is scattered and segmented. Herein, we constructed the first single-cell transcriptomic landscape of the cochlea in Hipposideros armiger, a CF-FM bat, using a PacBio-optimized genome and compared it with the results obtained from unoptimized original genomes. Sixteen distinct cell types were distributed across five spatial regions of the cochlea. Notably, through hematoxylin and eosin staining and fluorescence in situ hybridization, we identified new types of spiral ganglion neuron (SGN) cells in the cochlea of H. armiger. These SGN cells are likely critical for auditory perception and may have driven the adaptive evolution of high-frequency hearing in this species. Furthermore, we uncovered the differentiation relationships of among specific cell types, such as the transition from supporting cells to hair cells. Using the cochlear cell atlas as a reference, cell types susceptible to deafness-associated genes (in the human) were also identified. In summary, this study provides novel insights into the cellular and molecular mechanisms underlying the adaptive high-frequency hearing in bats and highlights potential candidate cell types and genes for therapeutic interventions in hearing loss.
Collapse
Affiliation(s)
- Mingyue Bao
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Xue Wang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Xintong Li
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Ruyi Sun
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Zhiqiang Wang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| | - Tinglei Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| | - Hui Wang
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
| | - Jiang Feng
- College of Life Science, Jilin Agricultural University, Changchun 130118, China; (M.B.)
- Jilin Provincial International Cooperation Key Laboratory for Biological Control of Agricultural Pests, Changchun 130118, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130117, China
| |
Collapse
|
20
|
Wang L, Liu Z, Zhao S, Xu K, Aceves V, Qiu C, Feng HC, Bian F, He J, Song CJ, Troutwine B, Liu L, Ma S, Niu Y, Wang S, Yuan S, Li X, Zhao L, Liu X, Qiu G, Wu Z, Zhang TJ, Gray RS, Wu N. Variants in the SOX9 transactivation middle domain induce axial skeleton dysplasia and scoliosis. Proc Natl Acad Sci U S A 2025; 122:e2313978121. [PMID: 39854231 PMCID: PMC11789016 DOI: 10.1073/pnas.2313978121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/30/2024] [Indexed: 01/30/2025] Open
Abstract
SOX9 is a crucial transcriptional regulator of cartilage development and homeostasis. Dysregulation of SOX9 is associated with a wide spectrum of skeletal disorders, including campomelic dysplasia, acampomelic campomelic dysplasia, and scoliosis. Yet how SOX9 variants contribute to the spectrum of axial skeletal disorders is not well understood. Here, we report four pathogenic variants of SOX9 identified in a cohort of patients with congenital vertebral malformations. We report a pathogenic missense variant in the transactivation middle (TAM) domain of SOX9 associated with mild skeletal dysplasia and scoliosis. We isolated a Sox9 mutant mouse with an in-frame microdeletion in the TAM domain (Sox9Asp272del), which exhibits skeletal dysplasia including kinked tails, rib cage anomalies, and scoliosis in homozygous mutants. We find that both the human missense and the mouse microdeletion mutations resulted in reduced SOX9 protein stability in cell culture, while Sox9Asp272del mutant mice show decreased SOX9 expression in the growth plate and annulus fibrosus tissues of the spine. This reduction in SOX9 expression was correlated with the reduction of extracellular matrix components, such as tenascin-X and the Adhesion G-protein coupled receptor ADGRG6. In summary, our work identified and modeled a pathologic variant of SOX9 within the TAM domain and demonstrated its importance for SOX9 protein stability. Our work demonstrates that SOX9 stability is important for the regulation of ADGRG6 expression, which is a known regulator of postnatal spine homeostasis, underscoring the essential role of SOX9 dosage in a spectrum of axial skeleton dysplasia in humans.
Collapse
Affiliation(s)
- Lianlei Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
| | - Zhaoyang Liu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Sen Zhao
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
| | - Kexin Xu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Valeria Aceves
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Cheng Qiu
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
| | - Hong Colleen Feng
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
| | - Fangzhou Bian
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
| | - Jingyu He
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
| | - Christina J. Song
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Benjamin Troutwine
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Lian Liu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
| | - Samuel Ma
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Yuchen Niu
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Shengru Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Suomao Yuan
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
| | - Xiaoxin Li
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Lina Zhao
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Xinyu Liu
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
| | - Guixing Qiu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Zhihong Wu
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Deciphering disorders Involving Scoliosis and COmorbidities (DISCO) study group
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Ryan S. Gray
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Nan Wu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| |
Collapse
|
21
|
Fang F, Casserly M, Robbins J, Thomopoulos S. Hedgehog signaling directs cell differentiation and plays a critical role in tendon enthesis healing. NPJ Regen Med 2025; 10:3. [PMID: 39833191 PMCID: PMC11747568 DOI: 10.1038/s41536-025-00392-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
A high prevalence of rotator cuff tears presents a major clinical challenge. A better understanding of the molecular mechanisms underlying enthesis development and healing is needed for developing treatments. We recently identified hedgehog (Hh)-lineage cells critical for enthesis development and repair. This study revealed cell-cell communication within the Hh-lineage cell population. To further characterize the role of Hh signaling, we used mouse models to activate and inactivate the Hh pathway in enthesis progenitors. Activation of Hh target genes during enthesis development increased its mineralization and mechanical properties. Activation of Hh signaling at the injured mature enthesis promoted fibrocartilage formation, enhanced mineralization, and increased expression of chondrogenic and osteogenic markers, which implies that Hh signaling drives cell differentiation to regenerate the damaged enthesis. Conversely, deletion of Hh target genes impaired enthesis healing. In summary, this study revealed a new strategy for enthesis repair via activation of Hh signaling in endogenous cells.
Collapse
Affiliation(s)
- Fei Fang
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthew Casserly
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Robbins
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| |
Collapse
|
22
|
Dong DL, Jin GZ. Targeting Chondrocyte Hypertrophy as Strategies for the Treatment of Osteoarthritis. Bioengineering (Basel) 2025; 12:77. [PMID: 39851351 PMCID: PMC11760869 DOI: 10.3390/bioengineering12010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by pain and functional impairment, which severely impacts the quality of life of middle-aged and elderly individuals. During normal bone development, chondrocyte hypertrophy is a natural physiological process. However, in the progression of OA, chondrocyte hypertrophy becomes one of its key pathological features. Although there is no definitive evidence to date confirming that chondrocyte hypertrophy is the direct cause of OA, substantial experimental data indicate that it plays an important role in the disease's pathogenesis. In this review, we first explore the mechanisms underlying chondrocyte hypertrophy in OA and offer new insights. We then propose strategies for inhibiting chondrocyte hypertrophy from the perspectives of targeting signaling pathways and tissue engineering, ultimately envisioning the future prospects of OA treatment.
Collapse
Affiliation(s)
- Da-Long Dong
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
| |
Collapse
|
23
|
Tailor K, van Ree J, Stowe T, Ventura B, Sisk C, Courtis J, Camp A, Elzamzami F, van Deursen J, O’Brien R, Baron J, Lui JC. Efficacy of cartilage-targeted IGF-1 in a mouse model of growth hormone insensitivity. Front Endocrinol (Lausanne) 2025; 15:1523931. [PMID: 39850478 PMCID: PMC11756323 DOI: 10.3389/fendo.2024.1523931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/13/2024] [Indexed: 01/25/2025] Open
Abstract
Recombinant human IGF-1 is used to treat severe primary IGF-1 deficiency, but this treatment requires twice-daily injection, often does not fully correct the growth deficit, and has important off-target effects. We therefore sought to target IGF-1 to growth plate cartilage by generating fusion proteins combining IGF-1 with single-chain human antibody fragments that target matrilin-3, a cartilage matrix protein. We previously showed that this cartilage-targeting IGF-1 fusion protein (CV1574-1) promoted growth plate function in a GH-deficient (lit) mouse model. Here, we studied CV1574-1 in a second mouse model, C57BL/6 wild-type mice treated with pegvisomant to induce GH resistance. In this model, once-daily injections of CV1574-1 for 5 days partially restored the pegvisomant-induced decrease in growth plate height without increasing kidney cell proliferation. Furthermore, we found that subcutaneous CV1574-1 showed significantly reduced hypoglycemic effect compared to injection of IGF-1 itself. Lastly, to gain mechanistic insights into the role of matrilin-3 targeting, we assessed the ability of CV1574-1 to activate AKT signaling in vitro and found that CV1574-1 caused a prolonged increase in AKT signaling compared to IGF-1 and that this effect was dependent on matrilin-3. Taken together, our findings provide further evidence that cartilage-targeted therapy could provide new pharmacological approaches for the treatment of childhood growth disorders, such as GH-insensitivity syndrome.
Collapse
Affiliation(s)
- Krishma Tailor
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
- Cavalry Biosciences, Inc., San Francisco, CA, United States
| | - Janine van Ree
- Cavalry Biosciences, Inc., San Francisco, CA, United States
| | - Timothy Stowe
- Cavalry Biosciences, Inc., San Francisco, CA, United States
| | - Brit Ventura
- Cavalry Biosciences, Inc., San Francisco, CA, United States
| | - Connor Sisk
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
| | - Joanna Courtis
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
| | - Anna Camp
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
| | - Fatima Elzamzami
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
| | | | - Robert O’Brien
- Cavalry Biosciences, Inc., San Francisco, CA, United States
| | - Jeffrey Baron
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
| | - Julian C. Lui
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
| |
Collapse
|
24
|
Boonrungsiman S, Allen C, Nudelman F, Shefelbine S, Farquharson C, Porter AE, Fleck RA. Endochondral ossification: Insights into the cartilage mineralization processes achieved by an anhydrous freeze substitution protocol. Acta Biomater 2025; 191:149-157. [PMID: 39542200 DOI: 10.1016/j.actbio.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Growth plate cartilage (GP) serves as a dynamic site of active mineralization and offers a unique opportunity to investigate the cell-regulated matrix mineralization process. Transmission electron microscopy (TEM) provides a means for the direct observation of these mechanisms, offering the necessary resolution and chemical analysis capabilities. However, as mineral crystallinity is prone to artifacts using aqueous fixation protocols, sample preparation techniques are critical to preserve the mineralized tissue in its native form. We optimized cryofixation by high-pressure freezing followed by freeze substitution in anhydrous acetone containing 0.5 % uranyl acetate to prepare murine GP for TEM analysis. This sample preparation workflow maintains cellular and extracellular protein structural integrity with sufficient contrast for observation and without compromising mineral crystallinity. By employing appropriate sample preparation techniques, we were able to observe two parallel mineralization processes driven by chondrocytes: 1) intracellular- and 2) extracellular-originating mineralized vesicles. Both mechanisms are based on sequestering calcium phosphate (CaP) within a membrane-limited structure, albeit originating from different compartments of the chondrocytes. In the intracellular originating pathway, CaP accumulates within mitochondria as globular CaP granules, which are incorporated into intracellular vesicles (500-1000 nm) and transported as granules to the extracellular matrix (ECM). In contrast, membrane budding vesicles with a size of approximately 100-200 nm, filled with needle-shaped minerals were observed only in the ECM. Both processes transport CaP to the collagenous matrix via vesicles, they can be differentiated based on the vesicle size and mineral morphologies. Their individual importance to the cartilage mineralization process is yet to be determined. STATEMENT OF SIGNIFICANCE: We do not fully understand the process by which epiphyseal cartilage mineralizes - a vital step in endochondral bone formation. Previous work has proposed that mitochondria and intracellular vesicles are storage sites for the delivery of mineral to collagen fibrils. However, these concepts are founded on results from in vitro models of mineralization; no prior work has observed mineral-containing intracellular vesicles or mitochondria in developing epiphyseal cartilage. Here we developed a new cryofixation preparation route for transmission electron microscopy (TEM) imaging that has disclosed a cell-regulated process of mineralization in epiphyseal cartilage. High resolution TEM images revealed an involvement of mitochondria and intracellular and extracellular vesicles in delivering transient mineral phases to the collagen fibrils to promote cartilage mineralization.
Collapse
Affiliation(s)
- Suwimon Boonrungsiman
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Christopher Allen
- Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, Oxfordshire OX11 0DE, UK
| | - Fabio Nudelman
- School of Chemistry, University of Edinburgh, King's Buildings, Edinburgh EH9 3FJ, UK
| | - Sandra Shefelbine
- Department of Mechanical and Industrial Engineering and Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Colin Farquharson
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | | | - Roland A Fleck
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK.
| |
Collapse
|
25
|
Hata K, Wakamori K, Hirakawa‐Yamamura A, Ichiyama‐Kobayashi S, Yamaguchi M, Okuzaki D, Takahata Y, Murakami T, Uzawa N, Yamashiro T, Nishimura R. Serinc5 Regulates Sequential Chondrocyte Differentiation by Inhibiting Sox9 Function in Pre-Hypertrophic Chondrocytes. J Cell Physiol 2025; 240:e31490. [PMID: 39568258 PMCID: PMC11747958 DOI: 10.1002/jcp.31490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024]
Abstract
The growth plate is the primary site of longitudinal bone growth with chondrocytes playing a pivotal role in endochondral bone development. Chondrocytes undergo a series of differentiation steps, resulting in the formation of a unique hierarchical columnar structure comprising round, proliferating, pre-hypertrophic, and hypertrophic chondrocytes. Pre-hypertrophic chondrocytes, which exist in the transitional stage between proliferating and hypertrophic stages, are a critical cell population in the growth plate. However, the molecular basis of pre-hypertrophic chondrocytes remains largely undefined. Here, we employed scRNA-seq analysis on fluorescently labeled growth plate chondrocytes for their molecular characterization. Serine incorporator 5 (Serinc5) was identified as a marker gene for pre-hypertrophic chondrocytes. Histological analysis revealed that Serinc5 is specifically expressed in pre-hypertrophic chondrocytes, overlapping with Indian hedgehog (Ihh). Serinc5 represses cell proliferation and Col2a1 and Acan expression by inhibiting the transcriptional activity of Sox9 in primary chondrocytes. Chromatin profiling using ChIP-seq and ATAC-seq revealed an active enhancer of Serinc5 located in intron 1, with its chromatin status progressively activated during chondrocyte differentiation. Collectively, our findings suggest that Serinc5 regulates sequential chondrocyte differentiation from proliferation to hypertrophy by inhibiting Sox9 function in pre-hypertrophic chondrocytes, providing novel insights into the mechanisms underlying chondrocyte differentiation in growth plates.
Collapse
Affiliation(s)
- Kenji Hata
- Department of Molecular and Cellular BiochemistryOsaka University Graduate School of DentistryOsakaJapan
| | - Kanta Wakamori
- Department of Molecular and Cellular BiochemistryOsaka University Graduate School of DentistryOsakaJapan
- Department of Oral & Maxillofacial Oncology and SurgeryOsaka University Graduate School of DentistryOsakaJapan
| | - Akane Hirakawa‐Yamamura
- Department of Molecular and Cellular BiochemistryOsaka University Graduate School of DentistryOsakaJapan
- Department of Orthodontics and Dentofacial OrthopedicsOsaka University Graduate School of DentistryOsakaJapan
| | - Sachi Ichiyama‐Kobayashi
- Department of Molecular and Cellular BiochemistryOsaka University Graduate School of DentistryOsakaJapan
- Department of Oral & Maxillofacial Oncology and SurgeryOsaka University Graduate School of DentistryOsakaJapan
| | - Masaya Yamaguchi
- Bioinformatics Research UnitOsaka University Graduate School of DentistryOsakaJapan
- Department of MicrobiologyOsaka University Graduate School of DentistryOsakaJapan
- Center for Infectious Diseases Education and ResearchOsaka UniversityOsakaJapan
| | - Daisuke Okuzaki
- Laboratory for Human Immunology (Single Cell Genomics)WPI Immunology Frontier Research Center, Osaka UniversityOsakaJapan
| | - Yoshifumi Takahata
- Department of Molecular and Cellular BiochemistryOsaka University Graduate School of DentistryOsakaJapan
- Genome Editing Research and Development UnitOsaka University Graduate School of DentistryOsakaJapan
| | - Tomohiko Murakami
- Department of Molecular and Cellular BiochemistryOsaka University Graduate School of DentistryOsakaJapan
| | - Narikazu Uzawa
- Department of Oral & Maxillofacial Oncology and SurgeryOsaka University Graduate School of DentistryOsakaJapan
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial OrthopedicsOsaka University Graduate School of DentistryOsakaJapan
| | - Riko Nishimura
- Department of Molecular and Cellular BiochemistryOsaka University Graduate School of DentistryOsakaJapan
| |
Collapse
|
26
|
Fu YF, Shi SW, Wu JJ, Yuan ZD, Wang LS, Nie H, Zhang ZY, Wu X, Chen YC, Ti HB, Zhang KY, Mao D, Ye JX, Li X, Yuan FL. Osteoclast Secretes Stage-Specific Key Molecules for Modulating Osteoclast-Osteoblast Communication. J Cell Physiol 2025; 240:e31484. [PMID: 39606839 DOI: 10.1002/jcp.31484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
In most cases of bone metabolic disorders, such as osteoporosis and osteomalacia, these conditions are often attributed to dysfunctional osteoclasts, leading to their common characterization as "destructors." In addition to the widely documented regulatory process where osteoblasts direct osteoclastic bone resorption, there is increasing evidence suggesting that osteoclasts also in turn influence osteoblastic bone formation through direct and indirect mechanisms. It is well-known that differentiation of osteoclasts involves several stages, each characterized by specific cellular features and functions. Stage-specific key molecules secreted during these stages play a critical role in mediating osteoclast-osteoblast communication. In this review, we described the different stages of osteoclast differentiation and reviewed stage-specific key molecules involved in osteoclasts-osteoblasts communication. We highlighted that a detailed understanding of these processes and molecular mechanism could facilitate the development of novel treatments for bone metabolic disorders.
Collapse
Affiliation(s)
- Yi-Fei Fu
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Shu-Wen Shi
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
| | - Lei-Sheng Wang
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Hao Nie
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
| | - Zheng-Yu Zhang
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Xian Wu
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Yue-Chun Chen
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Hui-Bo Ti
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Ke-Yue Zhang
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Dong Mao
- Orthopaedic Institute, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Jun-Xing Ye
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Xia Li
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- Wuxi Medical College, Jiangnan University, Wuxi, China
| |
Collapse
|
27
|
Pierre-Jerome C. The peripheral nervous system: peripheral neuropathies in the diabetic foot. MYOPATHIES AND TENDINOPATHIES OF THE DIABETIC FOOT 2025:451-482. [DOI: 10.1016/b978-0-443-13328-2.00022-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
Uptegrove A, Chen C, Sahagun-Bisson M, Kulkarni AK, Louie KW, Ueharu H, Mishina Y, Omi-Sugihara M. Influence of bone morphogenetic protein (BMP) signaling and masticatory load on morphological alterations of the mouse mandible during postnatal development. Arch Oral Biol 2025; 169:106096. [PMID: 39341045 PMCID: PMC11609011 DOI: 10.1016/j.archoralbio.2024.106096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/15/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
OBJECTIVE Bone homeostasis relies on several contributing factors, encompassing growth factors and mechanical stimuli. While bone morphogenetic protein (BMP) signaling is acknowledged for its essential role in skeletal development, its specific impact on mandibular morphogenesis remains unexplored. Here, we investigated the involvement of BMP signaling and mechanical loading through mastication in postnatal mandibular morphogenesis. DESIGN We employed conditional deletion of Bmpr1a in osteoblasts and chondrocytes via Osterix-Cre. Cre activity was induced at birth for the 3-week group and at three weeks for the 9-week and 12-week groups, respectively. The conditional knockout (cKO) and control mice were given either a regular diet (hard diet, HD) or a powdered diet (soft diet, SD) from 3 weeks until sample collection, followed by micro-CT and histological analysis. RESULTS The cKO mice exhibited shorter anterior lengths and a posteriorly inclined ramus across all age groups compared to the control mice. The cKO mice displayed an enlarged hypertrophic cartilage area along with fewer osteoclast numbers in the subchondral bone of the condyle compared to the control group at three weeks, followed by a reduction in the cartilage area in the posterior region at twelve weeks. Superimposed imaging and histomorphometrical analysis of the condyle revealed that BMP signaling primarily affects the posterior part of the condyle, while mastication affects the anterior part. CONCLUSIONS Using 3D landmark-based geometric morphometrics and histological assessments of the mandible, we demonstrated that BMP signaling and mechanical loading reciprocally contribute to the morphological alterations of the mandible and condyle during postnatal development.
Collapse
Affiliation(s)
- Amber Uptegrove
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Coral Chen
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Madison Sahagun-Bisson
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Anshul K Kulkarni
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Ke'ale W Louie
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Hiroki Ueharu
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA.
| | - Maiko Omi-Sugihara
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, USA; Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Osaka, Japan.
| |
Collapse
|
29
|
Boueya IL, Sandhow L, Albuquerque JRP, Znaidi R, Passaro D. Endothelial heterogeneity in bone marrow: insights across development, adult life and leukemia. Leukemia 2025; 39:8-24. [PMID: 39528790 PMCID: PMC11717709 DOI: 10.1038/s41375-024-02453-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
The central role of the endothelial microenvironment in orchestrating bone marrow (BM) homeostasis and hematopoietic support has been confirmed at various developmental stages and in adult life. The BM vasculature is crucial in mediating communication between BM parenchyma and circulating blood, displaying remarkable heterogeneity in structure and function. While vascular cell diversity in other tissues has long been recognized, the molecular basis of this phenomenon in BM is just now emerging. Over the past decade, single-cell approaches and microscopic observations have expanded our understanding of BM vasculature. While solely characterized for their paracrine properties in the past, recent advances have revolutionized our perception of endothelial function, revealing distinct anatomical locations associated with diverse endothelial cell states. The identification of phenotypic differences between normal and pathological conditions has therefore deepened our understanding of vascular dynamics and their impact on hematopoiesis in health and disease. In this review, we highlight key milestones and recent advances in understanding vascular heterogeneity within BM microenvironment during development, adulthood and aging. We also explore how leukemia affects this heterogeneity and how we can take this knowledge forward to improve clinical practices. By synthesizing existing literature, we aim to address unresolved questions and outline future research directions.
Collapse
Affiliation(s)
- I L Boueya
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - L Sandhow
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - J R P Albuquerque
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - R Znaidi
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - D Passaro
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France.
| |
Collapse
|
30
|
Yamaguchi H, Kitami M, Li M, Swaminathan S, Darabi R, Takemaru KI, Komatsu Y. Disruption of distal appendage protein CEP164 causes skeletal malformation in mice. Biochem Biophys Res Commun 2024; 741:151063. [PMID: 39612644 DOI: 10.1016/j.bbrc.2024.151063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
The primary cilium is a cellular antenna to orchestrate cell growth and differentiation. Deficient or dysfunctional cilia are frequently linked to skeletal abnormalities. Previous research demonstrated that ciliary proteins regulating axoneme elongation are essential for skeletogenesis. However, the role of the ciliary proteins responsible for initiating cilium assembly in skeletal development remains unknown. Here, we investigate the function of centrosomal protein of 164 kDa (CEP164), a key ciliogenesis regulator that localizes at the distal appendages of the mother centriole, during skeletal development in mice. Interestingly, the mesodermal cell-specific Cep164 deletion resulted in severe bone defects and osteoblast-specific deletion of Cep164 affected bone development. In contrast, chondrocyte-specific Cep164 deletion did not cause overt skeletal abnormalities, indicating that CEP164 functions in a cell type-specific manner within skeletal tissues. Importantly, Cep164-mutant osteoblasts not only displayed a lack of cilia but also showed an increased number of γH2AX-positive cells, indicating the involvement of defective DNA damage response in the etiology of skeletal lesions of Cep164-mutant mice. These results demonstrate that CEP164 has both ciliary and non-ciliary functions to control osteoblast growth and survival. Our study therefore reveals a novel understanding of the pathogenesis of skeletal ciliopathies associated with CEP164 dysfunction.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Megumi Kitami
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Margaret Li
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Kinesiology, Rice University Wiess School of Natural Science, Houston, TX, USA
| | - Sowmya Swaminathan
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Radbod Darabi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA; Institute of Muscle Biology and Cachexia, University of Houston, Houston, TX, USA
| | - Ken-Ichi Takemaru
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
31
|
Feng L, Lang Y, Sun L, Shi W, Chen X, Xia Y, Xu H, Liu Y. Ghrelin alleviated TiO 2 NPs-induced inhibition of endochondral osteogenesis and promoted longitudinal growth of long bones in juvenile rats via Wnt/β-catenin signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125185. [PMID: 39454809 DOI: 10.1016/j.envpol.2024.125185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) are widely used in children's daily necessities and foods, and their health hazards to children have attracted particular attention. Childhood is a critical time for accelerated bone growth and development. Current studies revealed that TiO2 NPs exposure causes bone damage in juvenile rats; however, the underlying mechanism is unknown. Ghrelin is a polypeptide hormone that is considered to be a candidate factor for regulating bone growth and development. In this research, 3-week-old juvenile male rats were administered 0, 100 or 200 mg/kg TiO2 NPs and 50 μg/kg ghrelin for 4 weeks to explore the underlying mechanism of TiO2 NPs-induced bone damage, and the protective effect of ghrelin. Our results revealed that TiO2 NPs resulted in decreased synthesis of bone growth-related hormones, disturbed bone metabolism, and destruction of bone structure. Further mechanism studies showed that TiO2 NPs inhibited Wnt/β-catenin pathway, reduced collagen synthesis, inhibited chondrocyte proliferation and differentiation, promoted chondrocyte apoptosis, and inhibited endochondral osteogenesis, ultimately leading to long bone longitudinal growth retardation and osteoporosis. Ghrelin alleviated the negative effects of TiO2 NPs-induced bone growth in juvenile rats by upregulating the Wnt/β-catenin signaling pathway. This study provided a reference for the clinical treatment of growth retardation and idiopathic short stature in juvenile children caused by environmental pollutants.
Collapse
Affiliation(s)
- Lihua Feng
- Department of Pediatrics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yuanyuan Lang
- Medical Imaging Center, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Leke Sun
- Department of Pediatrics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Weihong Shi
- Department of Pediatrics, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Xiangxiang Chen
- Department of Pediatrics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yanan Xia
- Department of Pediatrics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Yang Liu
- Department of Pediatrics, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
32
|
Zhou Y, Lu Y, Xu H, Ji X, Deng Q, Wang X, Zhang Y, Li Q, Lu Y, Rustempasic A, Liu Y, Wang Y. The effect of miR-205a with RUNX2 towards proliferation and differentiation of chicken chondrocytes in thiram-induced tibial dyschondroplasia. Poult Sci 2024; 103:104535. [PMID: 39541878 PMCID: PMC11609359 DOI: 10.1016/j.psj.2024.104535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Tibial dyschondroplasia (TD) is a kind of metabolic bone disease in fast-growing broilers, which seriously restricts the development of poultry industry. Our previous studies have revealed a significant upregulation of miR-205a in TD cartilage tissue, suggesting its potential role as a regulatory factor in the pathogenesis of TD. However, the precise function implications and underlying regulatory mechanism remain elusive. Therefore, this study aims to elucidate the biological functions and regulatory mechanisms of miR-205a in the progression of TD by employing mehtodologies such as qRT-PCR, CCK-8 assay, EdU assays, and flow cytometry. The findings demonstrated that the transfection of miR-205a overexpression plasmid reduced chondrocytes growth and development in TD while enhancing apoptosis; conversely, blocking miR-205a had opposite effects. RUNX2 was identified as a target gene of miR-205a through biosynthesis and dual luciferase assays, and its overexpression helps chondrocytes in TD grow and develop. However, when both miR-205a and RUNX2 were overexpressed, the regulatory effect of RUNX2 was significantly suppressed. In conclusion, miR-205a plays a role in slowing the growth and development of chondrocytes in TD by targeting and reducing RUNX2 expression, which helps to initiate and progress TD.
Collapse
Affiliation(s)
- Yuxin Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yuxiang Lu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Hengyong Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Xuyang Ji
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Qingqing Deng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Xi Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yao Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Qiuhang Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yusheng Lu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Alma Rustempasic
- Faculty of Agriculture and Food Science, University in Sarajevo, Zmaja od Bosne 8, 71000 Sarajevo, Bosnia and Herzegovina
| | - Yiping Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China
| | - Yan Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu Campus, Chengdu 611130, PR China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, PR China.
| |
Collapse
|
33
|
Hao Y, Meng Q, Chang L, Qiu M, Han J, Wang Z, Li C, Ma J, Zhang X. IL-4 promotes chondrogenesis of bone marrow mesenchymal stem cells and blockade of IL-4Rα retards the endochondral ossification during rat embryonic bone development. Basic Clin Pharmacol Toxicol 2024; 135:693-704. [PMID: 39396908 DOI: 10.1111/bcpt.14088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/19/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024]
Abstract
Interleukin-4 (IL-4)/IL-4 receptor alpha (IL-4Rα) signalling pathways play important roles in the complex process of bone formation and bone remodelling. However, whether IL-4/IL-4Rα participates in skeletogenesis during embryonic development is not completely understood. We used the anti-IL-4Rα monoclonal antibody (anti-IL-4Rα mAb) as a powerful investigational tool to evaluate the potential roles of IL-4/IL-4Rα in the chondrogenic differentiation of rat bone marrow mesenchymal stem cells (BMSCs) in vitro. Simultaneously, we explored the effect of IL-4/IL-4Rα on bone ossification during rat embryo-fetal development. In this study, we found that, compared to the control group, IL-4 can significantly promote the chondrogenic differentiation of BMSCs. Furthermore, following exposure to anti-IL-4Rα mAb in pregnant rats, unexpected phenomena were observed in fetal bone development, including non-ossification of the fetal sternum, an incomplete ossification centre in long bones and a reduced number of ossification points in digit (toe) bones. To further investigate the underlying mechanism of the phenotype, we studied the rat sternum as the target organ, starting from different time points of sternum development in the embryonic stage. The results indicated that the retardation mainly occurred in the middle and late stages of embryonic development. This retardation was characterized by the inhibition of the differentiation process of mesenchymal stem cells into chondrocytes, resulting in reduced angiogenesis near the ossification centre, failure of osteoblasts to invade the centre of the cartilage body with the blood vessels and delayed formation of the primary ossification centre (POC). Overall, our study demonstrated the significant function of IL-4/IL-4Rα in chondrogenic differentiation of BMSCs and bone ossification during embryo-fetal development.
Collapse
Affiliation(s)
- Yimeng Hao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Qinghe Meng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, China
| | - Leilei Chang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minglong Qiu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianxin Han
- Liaoning Qianyi Testing Technology Development Co. Ltd., Benxi, Liaoning, China
| | - Zhiqin Wang
- Liaoning Qianyi Testing Technology Development Co. Ltd., Benxi, Liaoning, China
| | - Changwei Li
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jing Ma
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Weaver SR, Peralta-Herrera E, Torres HM, Jessen E, Bradley EW, Westendorf JJ. Phlpp1 alters the murine chondrocyte phospho-proteome during endochondral bone formation. Bone 2024; 189:117265. [PMID: 39349089 PMCID: PMC11549792 DOI: 10.1016/j.bone.2024.117265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
Appendicular skeletal growth and bone mass acquisition are controlled by a variety of growth factors, hormones, and mechanical forces in a dynamic process called endochondral ossification. In long bones, chondrocytes in the growth plate proliferate and undergo hypertrophy to drive bone lengthening and mineralization. Pleckstrin homology (PH) domain and leucine rich repeat phosphatase 1 and 2 (Phlpp1 and Phlpp2) are serine/threonine protein phosphatases that regulate cell proliferation, survival, and maturation via Akt, PKC, Raf1, S6k, and other intracellular signaling cascades. Germline deletion of Phlpp1 suppresses bone lengthening in growth plate chondrocytes. Here, we demonstrate that Phlpp2 does not regulate endochondral ossification, and we define the molecular differences between Phlpp1 and Phlpp2 in chondrocytes. Phlpp2-/- mice were phenotypically indistinguishable from their wildtype (WT) littermates, with similar bone length, bone mass, and growth plate dynamics. Deletion of Phlpp2 had moderate effects on the chondrocyte transcriptome and proteome compared to WT cells. By contrast, Phlpp1/2-/- (double knockout) mice resembled Phlpp1-/- mice phenotypically and molecularly, as the chondrocyte phospho-proteomes of Phlpp1-/- and Phlpp1/2-/- chondrocytes had similarities and were significantly different from WT and Phlpp2-/- chondrocyte phospho-proteomes. Data integration via multiparametric analysis showed that the transcriptome explained less variation in the data as a result of Phlpp1 or Phlpp2 deletion than proteome or phospho-proteome. Alterations in cell proliferation, collagen fibril organization, and Pdpk1 and Pak1/2 signaling pathways were identified in chondrocytes lacking Phlpp1, while cell cycle processes and Akt1 and Aurka signaling pathways were altered in chondrocytes lacking Phlpp2. These data demonstrate that Phlpp1, and to a lesser extent Phlpp2, regulate multiple and complex signaling cascades across the chondrocyte transcriptome, proteome, and phospho-proteome and that multi-omic data integration can reveal novel putative kinase targets that regulate endochondral ossification.
Collapse
Affiliation(s)
- Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America
| | | | - Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America
| | - Erik Jessen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States of America
| | - Elizabeth W Bradley
- Department of Orthopedics, School of Medicine, University of Minnesota, Minneapolis, MN, United States of America
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
35
|
Hernández-García F, Fernández-Iglesias Á, Rodríguez Suárez J, Gil Peña H, López JM, Pérez RF. The Crosstalk Between Cartilage and Bone in Skeletal Growth. Biomedicines 2024; 12:2662. [PMID: 39767569 PMCID: PMC11727353 DOI: 10.3390/biomedicines12122662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
While the flat bones of the face, most of the cranial bones, and the clavicles are formed directly from sheets of undifferentiated mesenchymal cells, most bones in the human body are first formed as cartilage templates. Cartilage is subsequently replaced by bone via a very tightly regulated process termed endochondral ossification, which is led by chondrocytes of the growth plate (GP). This process requires continuous communication between chondrocytes and invading cell populations, including osteoblasts, osteoclasts, and vascular cells. A deeper understanding of these signaling pathways is crucial not only for normal skeletal growth and maturation but also for their potential relevance to pathophysiological processes in bones and joints. Due to limited information on the communication between chondrocytes and other cell types in developing bones, this review examines the current knowledge of how interactions between chondrocytes and bone-forming cells modulate bone growth.
Collapse
Affiliation(s)
- Frank Hernández-García
- Departamento de Medicina, Oviedo University, 33003 Oviedo, Spain; (F.H.-G.); (J.R.S.)
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
| | - Ángela Fernández-Iglesias
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
| | - Julián Rodríguez Suárez
- Departamento de Medicina, Oviedo University, 33003 Oviedo, Spain; (F.H.-G.); (J.R.S.)
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- AGC de Infancia y Adolescencia, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- RICORS-SAMID (RD21/0012), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Helena Gil Peña
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- AGC de Infancia y Adolescencia, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- RICORS2040 (RD21/0005/0011), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José M. López
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- Departamento de Morfología y Biología Celular, Oviedo University, 33003 Oviedo, Spain
| | - Rocío Fuente Pérez
- Universidad Europea de Madrid, Department of Nursing, Faculty of Medicine, Health and Sports, 28670 Madrid, Spain
| |
Collapse
|
36
|
Suhardi V, Oktarina A, Niu Y, Sosa B, Retzky J, Greenblatt M, Ivashkiv L, Bostrom M, Yang X. A Murine Model of Non-Wear-Particle-Induced Aseptic Loosening. Biomimetics (Basel) 2024; 9:673. [PMID: 39590245 PMCID: PMC11592190 DOI: 10.3390/biomimetics9110673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The current murine models of peri-implant osseointegration failure are associated with wear particles. However, the current clinical osseointegration failure is not associated with wear particles. Here, we develop a murine model of osseointegration failure not associated with wear particles and validate it by comparing the cellular composition of interfacial tissues with human samples collected during total joint arthroplasty revision for aseptic loosening. MATERIALS AND METHODS Thirty-two 16-week-old female C57BL/6 mice underwent implantation with a press-fitted roughened titanium implant (Control, n = 11) to induce normal osseointegration and a press-fitted smooth polymethylmethacrylate implant (PMMA, n = 11), a loosely fitted smooth titanium implant (Smooth-Ti, n = 5) or a loosely fitted roughened titanium implant (Overdrill, n = 5) to induce osseointegration failure. Pullout testing was used to determine the strength of the bone-implant interface (n = 6 of each for Control and PMMA groups) at 2 weeks after implantation. Histology (n = 2/group) and immunofluorescence (n = 3/group) were used to determine the cellular composition of bone-implant interfacial tissue, and this was compared with two human samples. RESULTS Osseointegration failure was confirmed with grossly loosening implants and the presence of fibrous tissue identified via histology. The maximum pullout load in the PMMA group was 87% lower than in the Control group (2.8 ± 0.6 N vs. 21 ± 1.5 N, p < 0.001). With immunofluorescence, abundant fibroblasts (PDGFRα+ TCF4+ and PDGFRα+ Pu1+) were observed in osseointegration failure groups and the human samples, but not in controls. Interestingly, CD146+PDGFRα+ and LepR+PDGFRα+ mesenchymal progenitors, osteoblasts (OPN+), vascular endothelium (EMCN+) cells were observed in all groups, indicating dynamic osteogenic activity. Macrophages, only M2, were observed in conditions producing fibrous tissue. CONCLUSIONS In this newly developed non-wear-particle-related murine osseointegration failure model, the cellular composition of human and murine interfacial tissue implicates specific populations of fibroblasts in fibrous tissue formation and implies that these cells may derive from mesenchymal stem cells.
Collapse
Affiliation(s)
- Vincentius Suhardi
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY 10021, USA; (V.S.); (M.B.)
- Research Institute, Hospital for Special Surgery, New York, NY 10021, USA (A.O.); (L.I.)
| | - Anastasia Oktarina
- Research Institute, Hospital for Special Surgery, New York, NY 10021, USA (A.O.); (L.I.)
| | - Yingzhen Niu
- Department of Joint Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050052, China
| | - Branden Sosa
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY 10021, USA; (V.S.); (M.B.)
| | - Julia Retzky
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY 10021, USA; (V.S.); (M.B.)
| | - Matthew Greenblatt
- Research Institute, Hospital for Special Surgery, New York, NY 10021, USA (A.O.); (L.I.)
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lionel Ivashkiv
- Research Institute, Hospital for Special Surgery, New York, NY 10021, USA (A.O.); (L.I.)
| | - Mathias Bostrom
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY 10021, USA; (V.S.); (M.B.)
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY 10021, USA
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, NY 10021, USA (A.O.); (L.I.)
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
37
|
Dinesh NEH, Baratang N, Rosseau J, Mohapatra R, Li L, Mahalingam R, Tiedemann K, Campeau PM, Reinhardt DP. Fibronectin isoforms promote postnatal skeletal development. Matrix Biol 2024; 133:86-102. [PMID: 39159790 DOI: 10.1016/j.matbio.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
Fibronectin (FN) is a ubiquitous extracellular matrix glycoprotein essential for the development of various tissues. Mutations in FN cause a unique form of spondylometaphyseal dysplasia, emphasizing its importance in cartilage and bone development. However, the relevance and functional role of FN during skeletal development has remained elusive. To address these aspects, we have generated conditional knockout mouse models targeting the cellular FN isoform in cartilage (cFNKO), the plasma FN isoform in hepatocytes (pFNKO), and both isoforms together in a double knockout (FNdKO). We used these mice to determine the relevance of the two principal FN isoforms in skeletal development from postnatal day one to the adult stage at two months. We identified a distinct topological FN deposition pattern in the mouse limb during different gestational and postnatal skeletal development phases, with prominent levels at the resting and hypertrophic chondrocyte zones and in the trabecular bone. Cartilage-specific cFN emerged as the predominant isoform in the growth plate, whereas circulating pFN remained excluded from the growth plate and confined to the primary and secondary ossification centers. Deleting either isoform independently (cFNKO or pFNKO) yielded only relatively subtle changes in the analyzed skeletal parameters. However, the double knockout of cFN in the growth plate and pFN in the circulation of the FNdKO mice significantly reduced postnatal body weight, body length, and bone length. Micro-CT analysis of the adult bone microarchitecture in FNdKO mice exposed substantial reductions in trabecular bone parameters and bone mineral density. The mice also showed elevated bone marrow adiposity. Analysis of chondrogenesis in FNdKO mice demonstrated changes in the resting, proliferating and hypertrophic growth plate zones, consistent alterations in chondrogenic markers such as collagen type II and X, decreased apoptosis of hypertrophic chondrocytes, and downregulation of bone formation markers. Transforming growth factor-β1 and downstream phospho-AKT levels were significantly lower in the FNdKO than in the control mice, revealing a crucial FN-mediated regulatory pathway in chondrogenesis and bone formation. In conclusion, the data demonstrate that FN is essential for chondrogenesis and bone development. Even though cFN and pFN act in different regions of the bone, both FN isoforms are required for the regulation of chondrogenesis, cartilage maturation, trabecular bone formation, and overall skeletal growth.
Collapse
Affiliation(s)
- Neha E H Dinesh
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | | | | | - Ronit Mohapatra
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Ling Li
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Ramshaa Mahalingam
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | | | | | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
38
|
To K, Fei L, Pett JP, Roberts K, Blain R, Polański K, Li T, Yayon N, He P, Xu C, Cranley J, Moy M, Li R, Kanemaru K, Huang N, Megas S, Richardson L, Kapuge R, Perera S, Tuck E, Wilbrey-Clark A, Mulas I, Memi F, Cakir B, Predeus AV, Horsfall D, Murray S, Prete M, Mazin P, He X, Meyer KB, Haniffa M, Barker RA, Bayraktar O, Chédotal A, Buckley CD, Teichmann SA. A multi-omic atlas of human embryonic skeletal development. Nature 2024; 635:657-667. [PMID: 39567793 PMCID: PMC11578895 DOI: 10.1038/s41586-024-08189-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/09/2024] [Indexed: 11/22/2024]
Abstract
Human embryonic bone and joint formation is determined by coordinated differentiation of progenitors in the nascent skeleton. The cell states, epigenetic processes and key regulatory factors that underlie lineage commitment of these cells remain elusive. Here we applied paired transcriptional and epigenetic profiling of approximately 336,000 nucleus droplets and spatial transcriptomics to establish a multi-omic atlas of human embryonic joint and cranium development between 5 and 11 weeks after conception. Using combined modelling of transcriptional and epigenetic data, we characterized regionally distinct limb and cranial osteoprogenitor trajectories across the embryonic skeleton and further described regulatory networks that govern intramembranous and endochondral ossification. Spatial localization of cell clusters in our in situ sequencing data using a new tool, ISS-Patcher, revealed mechanisms of progenitor zonation during bone and joint formation. Through trajectory analysis, we predicted potential non-canonical cellular origins for human chondrocytes from Schwann cells. We also introduce SNP2Cell, a tool to link cell-type-specific regulatory networks to polygenic traits such as osteoarthritis. Using osteolineage trajectories characterized here, we simulated in silico perturbations of genes that cause monogenic craniosynostosis and implicate potential cell states and disease mechanisms. This work forms a detailed and dynamic regulatory atlas of bone and cartilage maturation and advances our fundamental understanding of cell-fate determination in human skeletal development.
Collapse
Affiliation(s)
- Ken To
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Lijiang Fei
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - J Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Kenny Roberts
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Raphael Blain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Tong Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nadav Yayon
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
| | - Peng He
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - James Cranley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Madelyn Moy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Ruoyan Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Stathis Megas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Centre for AI in Medicine, Department of Applied Mathematics and Theoretical Physics, Cambridge, UK
| | | | - Rakesh Kapuge
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Elizabeth Tuck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Ilaria Mulas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Fani Memi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Batuhan Cakir
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - David Horsfall
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Simon Murray
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Pavel Mazin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Xiaoling He
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Newcastle University, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Omer Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Institut de Pathologie, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | | | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Cambridge Centre for AI in Medicine, Department of Applied Mathematics and Theoretical Physics, Cambridge, UK.
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- CIFAR Macmillan Multi-scale Human Programme, CIFAR, Toronto, Canada.
| |
Collapse
|
39
|
Alghonemy WY, Hegazy AA, Elmigdadi F, Atia GAN, Helal MB. Potential teratogenic effect of prenatal dexamethasone administration on palate development: Experimental study in rats. TRANSLATIONAL RESEARCH IN ANATOMY 2024; 37:100338. [DOI: 10.1016/j.tria.2024.100338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
|
40
|
Bian F, Hansen V, Feng HC, He J, Chen Y, Feng K, Ebrahimi B, Gray RS, Chai Y, Wu CL, Liu Z. The G protein-coupled receptor ADGRG6 maintains mouse growth plate homeostasis through IHH signaling. J Bone Miner Res 2024; 39:1644-1658. [PMID: 39236220 PMCID: PMC11523133 DOI: 10.1093/jbmr/zjae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/31/2024] [Accepted: 09/04/2024] [Indexed: 09/07/2024]
Abstract
The cartilage growth plate is essential for maintaining skeletal growth; however, the mechanisms governing postnatal growth plate homeostasis are still poorly understood. Using approaches of molecular mouse genetics and spatial transcriptomics applied to formalin-fixed, paraffin-embedded tissues, we show that ADGRG6/GPR126, a cartilage-enriched adhesion G protein-coupled receptor (GPCR), is essential for maintaining slow-cycling resting zone cells, appropriate chondrocyte proliferation and differentiation, and growth plate homeostasis in mice. Constitutive ablation of Adgrg6 in osteochondral progenitor cells with Col2a1Cre leads to a shortened resting zone, formation of cell clusters within the proliferative zone, and an elongated hypertrophic growth plate, marked by limited expression of parathyroid hormone-related protein (PTHrP) but increased Indian Hedgehog (IHH) signaling throughout the growth plate. Attenuation of smoothened-dependent hedgehog signaling restored the Adgrg6 deficiency-induced expansion of hypertrophic chondrocytes, confirming that IHH signaling can promote chondrocyte hypertrophy in a PTHrP-independent manner. In contrast, postnatal ablation of Adgrg6 in mature chondrocytes with AcanCreERT2, induced after the formation of the resting zone, does not affect PTHrP expression but causes an overall reduction of growth plate thickness marked by increased cell death specifically in the resting zone cells and a general reduction of chondrocyte proliferation and differentiation. Spatial transcriptomics reveals that ADGRG6 is essential for maintaining chondrocyte homeostasis by regulating osteogenic and catabolic genes in all the zones of the postnatal growth plates, potentially through positive regulation of SOX9 expression. Our findings elucidate the essential role of a cartilage-enriched adhesion GPCR in regulating cell proliferation and hypertrophic differentiation by regulation of PTHrP/IHH signaling, maintenance of slow-cycle resting zone chondrocytes, and safeguarding chondrocyte homeostasis in postnatal mouse growth plates.
Collapse
Affiliation(s)
- Fangzhou Bian
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Victoria Hansen
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - Hong Colleen Feng
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Jingyu He
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Yanshi Chen
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, United States
- Department of Biology, University of Rochester, Rochester, NY 14642, United States
| | - Kaining Feng
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Brenda Ebrahimi
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Ryan S Gray
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, United States
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Chia-Lung Wu
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - Zhaoyang Liu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| |
Collapse
|
41
|
Wang Y, Zhou R, Dong Z, Wang W, Guo L, Sun J, Rong X, Li P. Loss of Hdac4 in osteoprogenitors impairs postnatal trabecular and cortical bone formation, resulting in a dwarfism and osteopenia phenotype in mice. J Biol Chem 2024; 300:107941. [PMID: 39481602 DOI: 10.1016/j.jbc.2024.107941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 11/02/2024] Open
Abstract
HDAC4 is a class II histone deacetylation protein with a well-characterized role in chondrocyte differentiation and skeletal development, and dysregulated expression or haploinsufficiency of Hdac4 leads to skeletal formation and malformation disorders. The early lethality of Hdac4 ablation mice hindered further investigation of its role in postnatal bone growth and development. Therefore, this study aims to investigate the significant role of Hdac4 in postnatal endochondral bone development using two mouse models with conditional deletion of Hdac4 in Sp7-expressing osteoprogenitors or chondrocytes and monitored postnatal bone development. The phenotype of Acan-CreERT2; Hdac4fl/fl mice largely resembled that of conventional Hdac4-/- mice. But phenotypic characterizations of mice with Hdac4 inactivation in Sp7-expressing osteoprogenitors (Sp7-Cre; Hdac4fl/fl) showed dwarfism with body and limb shortening and remarkable skeletal defects. Microcomputed tomography analysis of tibias further demonstrated that loss of Hdac4 expression impaired bone formation and microarchitecture, mainly characterized by dysplasia of trabecular and cortical bone in young mice. Our in vivo and in vitro data support a crucial role for Hdac4 in regulating osteoblast proliferation and differentiation, bone matrix protein production, angiogenesis, and ultimately trabecular and cortical bone formation. Moreover, RNA-seq analysis implicated Hdac4 in the regulation of key genes and pathways necessary to affect the accumulation of extracellular matrix, biological processes related to signal transduction, and skeletal growth. Collectively, our data show that postnatal expression of Hdac4 in Sp7-expressing osteoprogenitors provides essential regulatory oversight of endochondral bone formation, bone morphology, and homeostasis.
Collapse
Affiliation(s)
- YunFei Wang
- Department of Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Raorao Zhou
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Zhengquan Dong
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Wenting Wang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, Shanxi Medical University, Taiyuan, China
| | - Li Guo
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Jian Sun
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Xueqin Rong
- Department of Pain Spinal Minimally Invasive Centre, Sanya Central Hospital, Sanya, Hainan, China.
| | - Pengcui Li
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China.
| |
Collapse
|
42
|
Zhou Z, Mao X, Jiang C, Li W, Zhou T, Liu M, Sun S, Wang M, Dong N, Wu Q, Zhou H. Deficiencies in corin and atrial natriuretic peptide-mediated signaling impair endochondral ossification in bone development. Commun Biol 2024; 7:1380. [PMID: 39443661 PMCID: PMC11500007 DOI: 10.1038/s42003-024-07077-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Corin is a protease that activates atrial natriuretic peptide (ANP), a hormone in cardiovascular homeostasis. Structurally, ANP is similar to C-type natriuretic peptide (CNP) crucial in bone development. Here, we examine the role of corin and ANP in chondrocyte differentiation and bone formation. We show that in Corin and Nppa (encoding ANP) knockout (KO) mice, chondrocyte differentiation is impaired, resulting in shortened limb long bones. In adult mice, Corin and Nppa deficiency impairs bone density and microarchitecture. Molecular studies in cartilages from newborn Corin and Nppa KO mice and in cultured chondrocytes indicate that corin and ANP act in chondrocytes via cGMP-dependent protein kinase G signaling to inhibit mitogen-activated protein kinase phosphorylation and stimulate glycogen synthase kinase-3β phosphorylation and β-catenin upregulation. These results indicate that corin and ANP signaling regulates chondrocyte differentiation in bone development and homeostasis, suggesting that enhancing ANP signaling may improve bone quality in patients with osteoporosis.
Collapse
Affiliation(s)
- Zibin Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Xiaoyu Mao
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Chun Jiang
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Wenguo Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Shijin Sun
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Mengting Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
| | - Haibin Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
43
|
Zwama J, Rosenberg NM, Verheij VA, Raijmakers PGHM, Yaqub M, Botman E, de Ruiter RD, Garrelfs MR, Bökenkamp A, Micha D, Schwarte LA, Teunissen BP, Lammertsma AA, Boellaard R, Eekhoff EMW. [ 18F]NaF PET/CT as a Marker for Fibrodysplasia Ossificans Progressiva: From Molecular Mechanisms to Clinical Applications in Bone Disorders. Biomolecules 2024; 14:1276. [PMID: 39456213 PMCID: PMC11505869 DOI: 10.3390/biom14101276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic bone disorder characterized by episodic flare-ups in connective tissue, which are frequently followed by the formation of heterotopic ossification. The absence of available plasma-soluble biomarkers for flare-ups or heterotopic bone formation poses severe challenges to the monitoring of disease activity to measure or predict disease progression. Recently, 18-fluor-sodium fluoride positron emission tomography/computed tomography ([18F]NaF PET/CT) was introduced as a potential marker for ossifying FOP activity. This review discusses the pharmacokinetics of [18F]NaF in relation to the pathophysiology of FOP, and its use as a marker of local bone metabolism in a variety of bone-related disorders. In addition, the review specifically addresses the applicability of [18F]NaF PET/CT imaging in FOP as a monitoring modality.
Collapse
Affiliation(s)
- Jolien Zwama
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Tissue Function and Regeneration, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Rare Bone Disease Centre, Amsterdam, The Netherlands
| | - Neeltje M. Rosenberg
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Tissue Function and Regeneration, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Rare Bone Disease Centre, Amsterdam, The Netherlands
| | - Vincent A. Verheij
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Rare Bone Disease Centre, Amsterdam, The Netherlands
| | - Pieter G. H. M. Raijmakers
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Esmée Botman
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, De Boelelaan 1117, Amsterdam, The Netherlands
- Rare Bone Disease Centre, Amsterdam, The Netherlands
| | - Ruben D. de Ruiter
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, De Boelelaan 1117, Amsterdam, The Netherlands
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Dijklander Hospital, Maelsonstraat 3, 1624 NP Hoorn, The Netherlands
| | - Mark R. Garrelfs
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Pediatric Endocrinology, Emma Children’s Hospital, Meibergdreef 9, Amsterdam, The Netherlands
| | - Arend Bökenkamp
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Pediatric Nephrology, Emma Children’s Hospital, Meibergdreef 9, Amsterdam, The Netherlands
| | - Dimitra Micha
- Amsterdam Movement Sciences, Tissue Function and Regeneration, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Human Genetics, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Lothar A. Schwarte
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Anesthesiology, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Bernd P. Teunissen
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Adriaan A. Lammertsma
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ronald Boellaard
- Rare Bone Disease Centre, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Elisabeth M. W. Eekhoff
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Movement Sciences, Tissue Function and Regeneration, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam, The Netherlands
- Rare Bone Disease Centre, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Hovis GE, Chandla A, Kolker SE, Yang I, Nagasawa DT. Ossified spinal epidermoid cyst: A systematic review and case report. Heliyon 2024; 10:e37093. [PMID: 39315203 PMCID: PMC11417560 DOI: 10.1016/j.heliyon.2024.e37093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Background Epidermoid cysts (ECs) are rare, benign lesions which comprise less than 1 % of all spinal tumors. Calcification of spinal ECs is rare, and EC ossification within the lumbar spine has never been documented. We report the only known congenital lumbar epidermoid tumor with ossification and a literature review of intradural lumbar ECs. Methods Studies meeting the following criteria were included: 1) EC as the primary tumor type, 2) intradural location, 3) involvement of the lumbar spinal level, and 4) primary presentation. Studies lacking individual patient data or published in a non-English language were excluded. Results A total of 172 studies were reviewed and 43 were included in analysis. Of the 83 total patients, 37 (45.1 %) were male and 45 (54.9 %) female, at an average age of 26 years. The L3 and L4 spinal levels were most frequently involved. Acquired etiology was reported in 49 (59.0 %) patients, and 24 (28.9 %) cases were congenital. Multivariate analyses demonstrated trends between decreased age and improved outcome, decreased delay in diagnosis and improved outcome, and increased extent of resection with reduced recurrence. Nine calcified spinal ECs were identified, with no previous report of EC ossification in the lumbar spine. Conclusion We present a case report of the only known ossified epidermoid tumor of the lumbar spine and a comprehensive literature review of 83 patients with intradural lumbar ECs. This review demonstrated trends between reduced age and improved outcome, reduced delay in diagnosis and improved outcome, and increased extent of resection with reduced recurrence.
Collapse
Affiliation(s)
- Gabrielle E.A. Hovis
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anubhav Chandla
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Steven E. Kolker
- Department of Pathology, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Isaac Yang
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Providence Saint John's Health Center, Santa Monica, CA, USA
- Department of Radiation Oncology, University of California, Los Angeles, California, USA
- Department of Head and Neck Surgery, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Los Angeles Biomedical Research Institute, University of California, Los Angeles, California, USA
- Harbor-UCLA Medical Center, University of California, Los Angeles, California, USA
| | | |
Collapse
|
45
|
Nakano N, Tashiro E, Shimada T, Ebisawa M, Kojima S, Ayabe K, Yamamoto Y, Maeda S, Itoh F, Itoh S. Involvement of mitogen- and stress-activated protein kinase 1 in BMP-6-induced chondrocyte differentiation. J Biol Chem 2024; 300:107806. [PMID: 39307301 PMCID: PMC11541777 DOI: 10.1016/j.jbc.2024.107806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 10/27/2024] Open
Abstract
Bone morphogenetic proteins (BMPs) are involved in several cellular responsive actions, such as development, cell differentiation, and apoptosis, via their specific transmembrane receptors. In particular, BMPs promote the differentiation and maturation of bone and cartilage from mesenchymal stem cells. Based on comprehensive analyses performed with a large number of antibodies, mitogen- and stress-activated protein kinase (MSK)1 was found to be immediately phosphorylated in the mouse chondrocyte precursor cell line, ATDC5, upon BMP-6 stimulation. The overexpression and knockdown of MSK1 in ATDC5 cells also enhanced and suppressed BMP-6-induced chondrocyte differentiation, respectively. Similar to ATDC5 cells, an ex vivo organ culture system using mouse embryonic metatarsal bones also demonstrated that BMP-6-mediated MSK1 activation might play a role in chondrocyte differentiation. Using several inhibitors, the p38 kinase pathway was confirmed to be implicated in BMP-6-induced phosphorylation of MSK1. Furthermore, MSK1 mutants lacking kinase activities and those lacking serine/threonine residues targeted by p38 kinase severely impaired their ability to potentiate BMP-6-induced chondrogenic differentiation of ATDC5 cells. Interestingly, a loss-of-function study for Smad4 perturbed BMP-6-induced phosphorylation of p38 kinase to inhibit BMP-6-mediated chondrocyte differentiation via MSK1 activation. Overall, both Smad-dependent and independent pathways require BMP-6-induced chondrocyte differentiation via MSK1 activation in ATDC5 cells.
Collapse
Affiliation(s)
- Naoko Nakano
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Etsu Tashiro
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Takayuki Shimada
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Masayasu Ebisawa
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Sayaka Kojima
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Kaho Ayabe
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Yohei Yamamoto
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Shingo Maeda
- Department of Bone and Joint Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Kagoshima, Japan
| | - Fumiko Itoh
- Laboratory of Stem Cell Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Susumu Itoh
- Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo, Japan.
| |
Collapse
|
46
|
Pei Y, Liu F, Zhao Y, Lin H, Huang X. Role of hedgehog signaling in the pathogenesis and therapy of heterotopic ossification. Front Cell Dev Biol 2024; 12:1454058. [PMID: 39364140 PMCID: PMC11447292 DOI: 10.3389/fcell.2024.1454058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
Heterotopic ossification (HO) is a pathological process that generates ectopic bone in soft tissues. Hedgehog signaling (Hh signaling) is a signaling pathway that plays an important role in embryonic development and involves three ligands: sonic hedgehog (Shh), Indian hedgehog (Ihh) and desert hedgehog (Dhh). Hh signaling also has an important role in skeletal development. This paper discusses the effects of Hh signaling on the process of HO formation and describes several signaling molecules that are involved in Hh-mediated processes: parathyroid Hormone-Related Protein (PTHrP) and Fkbp10 mediate the expression of Hh during chondrogenesic differentiation. Extracellular signal-regulated kinase (ERK), GNAs and Yes-Associated Protein (YAP) interact with Hh signaling to play a role in osteogenic differentiation. Runt-Related Transcription Factor 2 (Runx2), Mohawk gene (Mkx) and bone morphogenetic protein (BMP) mediate Hh signaling during both chondrogenic and osteogenic differentiation. This paper also discusses possible therapeutic options for HO, lists several Hh inhibitors and explores whether they could serve as emerging targets for the treatment of HO.
Collapse
Affiliation(s)
- Yiran Pei
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Fangzhou Liu
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yike Zhao
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoyan Huang
- The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
47
|
Wang L, Yang H, Wang C, Wang M, Huang J, Nyunt T, Osorio C, Sun SY, Pacifici M, Lefebvre V, Moore DC, Wang S, Yang W. SHP2 ablation mitigates osteoarthritic cartilage degeneration by promoting chondrocyte anabolism through SOX9. FASEB J 2024; 38:e70013. [PMID: 39225365 PMCID: PMC11404350 DOI: 10.1096/fj.202400642r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/21/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Articular cartilage phenotypic homeostasis is crucial for life-long joint function, but the underlying cellular and molecular mechanisms governing chondrocyte stability remain poorly understood. Here, we show that the protein tyrosine phosphatase SHP2 is differentially expressed in articular cartilage (AC) and growth plate cartilage (GPC) and that it negatively regulates cell proliferation and cartilage phenotypic program. Postnatal SHP2 deletion in Prg4+ AC chondrocytes increased articular cellularity and thickness, whereas SHP2 deletion in Acan+ pan-chondrocytes caused excessive GPC chondrocyte proliferation and led to joint malformation post-puberty. These observations were verified in mice and in cultured chondrocytes following treatment with the SHP2 PROTAC inhibitor SHP2D26. Further mechanistic studies indicated that SHP2 negatively regulates SOX9 stability and transcriptional activity by influencing SOX9 phosphorylation and promoting its proteasome degradation. In contrast to published work, SHP2 ablation in chondrocytes did not impact IL-1-evoked inflammation responses, and SHP2's negative regulation of SOX9 could be curtailed by genetic or chemical SHP2 inhibition, suggesting that manipulating SHP2 signaling has translational potential for diseases of cartilage dyshomeostasis.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Huiliang Yang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Changwei Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Mingliang Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jiahui Huang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Thedoe Nyunt
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Camilo Osorio
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Véronique Lefebvre
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Douglas C Moore
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Wentian Yang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Rhode Island Hospital, Providence, Rhode Island, USA
| |
Collapse
|
48
|
Rubin S, Agrawal A, Seewald A, Lian MJ, Gottdenker O, Villoutreix P, Baule A, Stern T, Zelzer E. Limited column formation in the embryonic growth plate implies divergent growth mechanisms during pre- and postnatal bone development. eLife 2024; 13:e95289. [PMID: 39269144 PMCID: PMC11509684 DOI: 10.7554/elife.95289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 09/09/2024] [Indexed: 09/15/2024] Open
Abstract
Chondrocyte columns, which are a hallmark of growth plate architecture, play a central role in bone elongation. Columns are formed by clonal expansion following rotation of the division plane, resulting in a stack of cells oriented parallel to the growth direction. In this work, we analyzed hundreds of Confetti multicolor clones in growth plates of mouse embryos using a pipeline comprising 3D imaging and algorithms for morphometric analysis. Surprisingly, analysis of the elevation angles between neighboring pairs of cells revealed that most cells did not display the typical stacking pattern associated with column formation, implying incomplete rotation of the division plane. Morphological analysis revealed that although embryonic clones were elongated, they formed clusters oriented perpendicular to the growth direction. Analysis of growth plates of postnatal mice revealed both complex columns, composed of ordered and disordered cell stacks, and small, disorganized clusters located in the outer edges. Finally, correlation between the temporal dynamics of the ratios between clusters and columns and between bone elongation and expansion suggests that clusters may promote expansion, whereas columns support elongation. Overall, our findings support the idea that modulations of division plane rotation of proliferating chondrocytes determines the formation of either clusters or columns, a multifunctional design that regulates morphogenesis throughout pre- and postnatal bone growth. Broadly, this work provides a new understanding of the cellular mechanisms underlying growth plate activity and bone elongation during development.
Collapse
Affiliation(s)
- Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Ankit Agrawal
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
- Würzburg Institute of Systems Immunology, Julius‐Maximilians‐Universität WürzburgWürzburgGermany
| | - Anne Seewald
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Meng-Jia Lian
- Department of Biologic and Materials & Prosthodontics, University of Michigan School of DentistryAnn ArborUnited States
| | - Olivia Gottdenker
- Department of Biologic and Materials & Prosthodontics, University of Michigan School of DentistryAnn ArborUnited States
| | - Paul Villoutreix
- Aix Marseille Univ, INSERM, MMG, UMR1251, Turing Center for Living SystemsMarseilleFrance
| | - Adrian Baule
- School of Mathematical Sciences, Queen Mary University of LondonLondonUnited Kingdom
| | - Tomer Stern
- Department of Biologic and Materials & Prosthodontics, University of Michigan School of DentistryAnn ArborUnited States
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
49
|
Li Q, Jiang S, Lei K, Han H, Chen Y, Lin W, Xiong Q, Qi X, Gan X, Sheng R, Wang Y, Zhang Y, Ma J, Li T, Lin S, Zhou C, Chen D, Yuan Q. Metabolic rewiring during bone development underlies tRNA m7G-associated primordial dwarfism. J Clin Invest 2024; 134:e177220. [PMID: 39255038 PMCID: PMC11473147 DOI: 10.1172/jci177220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
Translation of mRNA to protein is tightly regulated by transfer RNAs (tRNAs), which are subject to various chemical modifications that maintain structure, stability, and function. Deficiency of tRNA N7-methylguanosine (m7G) modification in patients causes a type of primordial dwarfism, but the underlying mechanism remains unknown. Here we report that the loss of m7G rewires cellular metabolism, leading to the pathogenesis of primordial dwarfism. Conditional deletion of the catalytic enzyme Mettl1 or missense mutation of the scaffold protein Wdr4 severely impaired endochondral bone formation and bone mass accrual. Mechanistically, Mettl1 knockout decreased abundance of m7G-modified tRNAs and inhibited translation of mRNAs relating to cytoskeleton and Rho GTPase signaling. Meanwhile, Mettl1 knockout enhanced cellular energy metabolism despite incompetent proliferation and osteogenic commitment. Further exploration revealed that impairment of Rho GTPase signaling upregulated the level of branched-chain amino acid transaminase 1 (BCAT1) that rewired cell metabolism and restricted intracellular α-ketoglutarate (αKG). Supplementation of αKG ameliorated the skeletal defect of Mettl1-deficient mice. In addition to the selective translation of metabolism-related mRNAs, we further revealed that Mettl1 knockout globally regulated translation via integrated stress response (ISR) and mammalian target of rapamycin complex 1 (mTORC1) signaling. Restoring translation by targeting either ISR or mTORC1 aggravated bone defects of Mettl1-deficient mice. Overall, our study unveils a critical role of m7G tRNA modification in bone development by regulation of cellular metabolism and indicates suspension of translation initiation as a quality control mechanism in response to tRNA dysregulation.
Collapse
Affiliation(s)
- Qiwen Li
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuang Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kexin Lei
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Han
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yaqian Chen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiuchan Xiong
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingying Qi
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyan Gan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Sheng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuan Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yarong Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jieyi Ma
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Li
- West China–Washington Mitochondria and Metabolism Center and Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuibin Lin
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demeng Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Campbell K, Naire S, Kuiper JH. A mathematical model of signalling molecule-mediated processes during regeneration of osteochondral defects after chondrocyte implantation. J Theor Biol 2024; 592:111874. [PMID: 38908475 DOI: 10.1016/j.jtbi.2024.111874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/12/2024] [Accepted: 06/06/2024] [Indexed: 06/24/2024]
Abstract
Treating bone-cartilage defects is a fundamental clinical problem. The ability of damaged cartilage to self-repair is limited due to its avascularity. Left untreated, these defects can lead to osteoarthritis. Details of osteochondral defect repair are elusive, but animal models indicate healing occurs via an endochondral ossification-like process, similar to that in the growth plate. In the growth plate, the signalling molecules parathyroid hormone-related protein (PTHrP) and Indian Hedgehog (Ihh) form a feedback loop regulating chondrocyte hypertrophy, with Ihh inducing and PTHrP suppressing hypertrophy. To better understand this repair process and to explore the regulatory role of signalling molecules on the regeneration process, we formulate a reaction-diffusion mathematical model of osteochondral defect regeneration after chondrocyte implantation. The drivers of healing are assumed to be chondrocytes and osteoblasts, and their interaction via signalling molecules. We model cell proliferation, migration and chondrocyte hypertrophy, and matrix production and conversion, spatially and temporally. We further model nutrient and signalling molecule diffusion and their interaction with the cells. We consider the PTHrP-Ihh feedback loop as the backbone mechanisms but the model is flexible to incorporate extra signalling mechanisms if needed. Our mathematical model is able to represent repair of osteochondral defects, starting with cartilage formation throughout the defect. This is followed by chondrocyte hypertrophy, matrix calcification and bone formation deep inside the defect, while cartilage at the surface is maintained and eventually separated from the deeper bone by a thin layer of calcified cartilage. The complete process requires around 48 months. A key highlight of the model demonstrates that the PTHrP-Ihh loop alone is insufficient and an extra mechanism is required to initiate chondrocyte hypertrophy, represented by a critical cartilage density. A parameter sensitivity study reveals that the timing of the repair process crucially depends on parameters, such as the critical cartilage density, and those describing the actions of PTHrP to suppress hypertrophy, such as its diffusion coefficient, threshold concentration and degradation rate.
Collapse
Affiliation(s)
- Kelly Campbell
- School of Computing and Mathematics, Keele University, Keele, ST5 5BG, UK
| | - Shailesh Naire
- School of Computing and Mathematics, Keele University, Keele, ST5 5BG, UK
| | - Jan Herman Kuiper
- School of Pharmacy and Bioengineering, Keele University, Keele, ST5 5BG, UK; Robert Jones and Agnes Hunt Orthopaedic & District Hospital NHS Trust, Oswestry, SY10 7AG, UK.
| |
Collapse
|