1
|
Ren Q, Xing W, Jiang B, Feng H, Hu X, Suo J, Wang L, Zou W. Tenascin-C promotes bone regeneration via inflammatory macrophages. Cell Death Differ 2025:10.1038/s41418-024-01429-9. [PMID: 39794452 DOI: 10.1038/s41418-024-01429-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/13/2025] Open
Abstract
During the early stage of tissue injury, macrophages play important roles in the activation of stem cells for further regeneration. However, the regulation of macrophages during bone regeneration remains unclear. Here, the extracellular matrix (ECM) tenascin-C (TNC) is found to express in the periosteum and recruit inflammatory macrophages. TNC-deficiency in the periosteum delays bone repair. Transplantation of macrophages derived from injured periosteum is able to rescue the decreased skeletal stem cells and impaired bone regeneration caused by TNC deficiency. The cell communication analysis identifies ITGA7 as a TNC receptor contributing to the recruitment of inflammatory macrophages. TNC expression declines in aged mice and the exogenous delivery of TNC significantly promotes bone regeneration after aging through the recruitment of macrophages. Taken together, this study reveals the regulation of macrophage recruitment and its function in the activation of skeletal stem cells after bone injury, providing a strategy to accelerate bone regeneration by TNC delivery.
Collapse
Affiliation(s)
- Qian Ren
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wenhui Xing
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- Hainan Medical University, Haikou, Hainan, China
| | - Bo Jiang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Heng Feng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xuye Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Lijun Wang
- Hainan Medical University, Haikou, Hainan, China.
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
- Hainan Medical University, Haikou, Hainan, China.
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
2
|
Sheng MHC, Rundle CH, Baylink DJ, Lau KHW. Conditional Deletion of Gremlin-1 in Cathepsin K-expressing Mature Osteoclasts Altered the Skeletal Response to Calcium Depletion in Sex-Dependent Manner. Calcif Tissue Int 2025; 116:28. [PMID: 39789342 PMCID: PMC11717885 DOI: 10.1007/s00223-024-01337-7] [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: 08/02/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025]
Abstract
This study assessed the novel concept that osteoclast-derived Grem1 has regulatory functions in the skeletal response to calcium stress using an osteoclastic Grem1 conditional knockout (cKO) mouse model. The calcium stress was initiated by feeding cKO mutants and wildtype (WT) littermates a calcium-deficient diet for 2 weeks. Deletion of Grem1 in mature osteoclasts did not affect developmental bone growth nor basal bone turnover. In response to calcium depletion, male cKO mutants showed greater increases in osteoclastic resorption and trabecular bone loss than male WT littermates, indicating an enhanced skeletal sensitivity to calcium depletion in male mutants. The enhanced sensitivity to calcium depletion was sex-dependent, as female cKO mutants showed lower increases in osteoclastic resorption and bone loss than female WT littermates as well as male cKO mutants. The sex disparity in osteoclastic resorption response to calcium stress was intrinsic to osteoclasts since osteoclasts of male but not female cKO mutants showed greater in vitro bone resorption activity than osteoclasts of WT littermates of respective sex. Male cKO mutants displayed smaller bone formation response to calcium depletion than male WT littermates, while female mutants showed bigger bone formation response than female WT littermates, indicating that cKO mutants also displayed sex disparity in bone formation response. The sex disparity in bone formation response was not caused by intrinsic differences in osteoblasts but might be due to sex-dependent differential osteoclastic release of osteogenic factors. In summary, osteoclast-derived gremlin-1 has complicated and sex-dependent regulatory roles in skeletal response to calcium stress.
Collapse
Affiliation(s)
- Matilda H-C Sheng
- Musculoskeletal Disease Center (151), Jerry L. Pettis Memorial VA Medical Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA.
- Department of Medicine and Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Charles H Rundle
- Musculoskeletal Disease Center (151), Jerry L. Pettis Memorial VA Medical Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA
- Department of Medicine and Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - David J Baylink
- Department of Medicine and Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Kin-Hing William Lau
- Musculoskeletal Disease Center (151), Jerry L. Pettis Memorial VA Medical Center, VA Loma Linda Healthcare System, 11201 Benton Street, Loma Linda, CA, 92357, USA
- Department of Medicine and Biochemistry, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
3
|
Wang Z, Wang K, Yu Y, Fu J, Zhang S, Li M, Yang J, Zhang X, Liu X, Lv F, Ma L, Cai H, Tian W, Liao L. Identification of human cranio-maxillofacial skeletal stem cells for mandibular development. SCIENCE ADVANCES 2025; 11:eado7852. [PMID: 39742474 DOI: 10.1126/sciadv.ado7852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
Compared with long bone that arises from the mesoderm, the major portion of the maxillofacial bones and the front bone of the skull are derived from cranial neural crest cells and undergo intramembranous ossification. Human skeletal stem cells have been identified in embryonic and fetal long bones. Here, we describe a single-cell atlas of the human embryonic mandible and identify a population of cranio-maxillofacial skeletal stem cells (CMSSCs). These CMSSCs are marked by interferon-induced transmembrane protein 5 (IFITM5) and are specifically located around the periosteum of the jawbone and frontal bone. Additionally, these CMSSCs exhibit strong self-renewal and osteogenic differentiation capacities but lower chondrogenic differentiation potency, mediating intramembranous bone formation without cartilage formation. IFITM5+ cells are also observed in the adult jawbone and exhibit functions similar to those of embryonic CMSSCs. Thus, this study identifies CMSSCs that orchestrate the intramembranous ossification of cranio-maxillofacial bones, providing a deeper understanding of cranio-maxillofacial skeletal development and promising seed cells for bone repair.
Collapse
Affiliation(s)
- Zhuo Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Kun Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yejia Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jing Fu
- Department of Reproductive Endocrinology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
| | - Siyuan Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Maojiao Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jian Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xuanhao Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xiaodong Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Fengqiong Lv
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
- Department of Operating Room Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
- Department of Operating Room Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Haoyang Cai
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Li Liao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Engineering Research Center of Oral Translational Medicine, Ministry of Education and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
4
|
Hojo H, Tani S, Ohba S. Modeling of skeletal development and diseases using human pluripotent stem cells. J Bone Miner Res 2024; 40:5-19. [PMID: 39498496 DOI: 10.1093/jbmr/zjae178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/28/2024] [Accepted: 11/02/2024] [Indexed: 01/07/2025]
Abstract
Human skeletal elements are formed from distinct origins at distinct positions of the embryo. For example, the neural crest produces the facial bones, the paraxial mesoderm produces the axial skeleton, and the lateral plate mesoderm produces the appendicular skeleton. During skeletal development, different combinations of signaling pathways are coordinated from distinct origins during the sequential developmental stages. Models for human skeletal development have been established using human pluripotent stem cells (hPSCs) and by exploiting our understanding of skeletal development. Stepwise protocols for generating skeletal cells from different origins have been designed to mimic developmental trails. Recently, organoid methods have allowed the multicellular organization of skeletal cell types to recapitulate complicated skeletal development and metabolism. Similarly, several genetic diseases of the skeleton have been modeled using patient-derived induced pluripotent stem cells and genome-editing technologies. Model-based drug screening is a powerful tool for identifying drug candidates. This review briefly summarizes our current understanding of the embryonic development of skeletal tissues and introduces the current state-of-the-art hPSC methods for recapitulating skeletal development, metabolism, and diseases. We also discuss the current limitations and future perspectives for applications of the hPSC-based modeling system in precision medicine in this research field.
Collapse
Affiliation(s)
- Hironori Hojo
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shoichiro Tani
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Shinsuke Ohba
- Department of Tissue and Developmental Biology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
5
|
Xu N, Yang K, Wang M. CCN3: lactational bone booster. Cell Biosci 2024; 14:155. [PMID: 39734229 DOI: 10.1186/s13578-024-01344-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: 07/15/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024] Open
Abstract
Mammalian reproduction requires that nursing mothers transfer large amounts of calcium to their offspring through milk. Meeting this demand requires the activation of a brain-breast-bone circuit during lactation that coordinates changes in systemic hormones, dietary calcium intake, skeletal turnover, and calcium transport into milk. Classically, increased bone resorption via increased parathyroid hormone-related protein and low estrogen levels is the main source of calcium for milk production during lactation. Over the past few decades, investigators have described many aspects of this brain-breast-bone axis during lactation, yet many unanswered questions remain. Using a comprehensive set of parabiosis coupled with in vivo µCT, bone transplant studies, cell culturing and differentiation assays, mouse genetic models, pharmacologic interventions, hepatic viral transduction, and sequencing analysis, a recent study discovered that cellular communication network factor 3 (CCN3), derived from ARHERα/Kiss1 neurons, functions as an osteogenic hormone to sustain bone formation and progeny survival during lactation. Compelling evidence has been presented to show that (1) CCN3 expression in ARHERα/Kiss1 neurons fluctuates, almost exclusively appearing during lactation; (2) CCN3 stimulates mouse and human skeletal stem cell activity, increases bone remodeling and fracture repair in young and old mice of both sexes; (3) knockdown Ccn3 transcripts in the ARHKiss1 neurons in lactating dams causes devastating bone loss and failure to sustain progeny survival. These findings suggested that the stage-specific expression of CCN3 in female ARHERα/Kiss1 neurons during lactation is a newly identified brain-bone axis evolved to sustain the skeleton in mammalian mothers and offspring.
Collapse
Affiliation(s)
- Nathan Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, USA
| | - Kyle Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, USA
| | - Mengjie Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, USA.
- , 1100 Bates Street #8066, Houston, TX, 77030, USA.
| |
Collapse
|
6
|
Cong T, Morse KW, Sosa BR, Lane JM, Rodeo SA, Greenblatt MB. Skeletal Stem Cells: A Basis for Orthopaedic Pathology and Tissue Repair. J Bone Joint Surg Am 2024:00004623-990000000-01286. [PMID: 39693451 DOI: 10.2106/jbjs.24.00905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
➢ Skeletal stem cells (SSCs) continually replenish mature cell populations to support skeletal homeostasis.➢ SSCs repopulate by self-renewal, have multilineage potential, and are long-lived in vivo.➢ SSCs express specific combinations of cell surface markers that reflect their lineage identity.➢ SSCs adapt to their anatomic environment to support regional differences in skeletal behavior and pathology.
Collapse
Affiliation(s)
- Ting Cong
- Department of Orthopaedic Surgery, UPMC Sports Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Orthopedic Surgery, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Kyle W Morse
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Branden R Sosa
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Joseph M Lane
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Scott A Rodeo
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
- Research Division, Hospital for Special Surgery, New York, NY
| |
Collapse
|
7
|
Reeves J, Tournier P, Becquart P, Carton R, Tang Y, Vigilante A, Fang D, Habib SJ. Rejuvenating aged osteoprogenitors for bone repair. eLife 2024; 13:RP104068. [PMID: 39692737 DOI: 10.7554/elife.104068] [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] [Indexed: 12/19/2024] Open
Abstract
Aging is marked by a decline in tissue regeneration, posing significant challenges to an increasingly older population. Here, we investigate age-related impairments in calvarial bone healing and introduce a novel two-part rejuvenation strategy to restore youthful repair. We demonstrate that aging negatively impacts the calvarial bone structure and its osteogenic tissues, diminishing osteoprogenitor number and function and severely impairing bone formation. Notably, increasing osteogenic cell numbers locally fails to rescue repair in aged mice, identifying the presence of intrinsic cellular deficits. Our strategy combines Wnt-mediated osteoprogenitor expansion with intermittent fasting, which leads to a striking restoration of youthful levels of bone healing. We find that intermittent fasting improves osteoprogenitor function, benefits that can be recapitulated by modulating NAD+-dependent pathways or the gut microbiota, underscoring the multifaceted nature of this intervention. Mechanistically, we identify mitochondrial dysfunction as a key component in age-related decline in osteoprogenitor function and show that both cyclical nutrient deprivation and Nicotinamide mononucleotide rejuvenate mitochondrial health, enhancing osteogenesis. These findings offer a promising therapeutic avenue for restoring youthful bone repair in aged individuals, with potential implications for rejuvenating other tissues.
Collapse
Affiliation(s)
- Joshua Reeves
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Pierre Tournier
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Pierre Becquart
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Robert Carton
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Yin Tang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Alessandra Vigilante
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Dong Fang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Shukry J Habib
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
8
|
Perrin S, Ethel M, Bretegnier V, Goachet C, Wotawa CA, Luka M, Coulpier F, Masson C, Ménager M, Colnot C. Single-nucleus transcriptomics reveal the differentiation trajectories of periosteal skeletal/stem progenitor cells in bone regeneration. eLife 2024; 13:RP92519. [PMID: 39642053 PMCID: PMC11623931 DOI: 10.7554/elife.92519] [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] [Indexed: 12/08/2024] Open
Abstract
Bone regeneration is mediated by skeletal stem/progenitor cells (SSPCs) that are mainly recruited from the periosteum after bone injury. The composition of the periosteum and the steps of SSPC activation and differentiation remain poorly understood. Here, we generated a single-nucleus atlas of the periosteum at steady state and of the fracture site during the early stages of bone repair (https://fracture-repair-atlas.cells.ucsc.edu). We identified periosteal SSPCs expressing stemness markers (Pi16 and Ly6a/SCA1) and responding to fracture by adopting an injury-induced fibrogenic cell (IIFC) fate, prior to undergoing osteogenesis or chondrogenesis. We identified distinct gene cores associated with IIFCs and their engagement into osteogenesis and chondrogenesis involving Notch, Wnt, and the circadian clock signaling, respectively. Finally, we show that IIFCs are the main source of paracrine signals in the fracture environment, suggesting a crucial paracrine role of this transient IIFC population during fracture healing. Overall, our study provides a complete temporal topography of the early stages of fracture healing and the dynamic response of periosteal SSPCs to injury, redefining our knowledge of bone regeneration.
Collapse
Affiliation(s)
- Simon Perrin
- Univ Paris Est Creteil, INSERM, IMRBCreteilFrance
| | - Maria Ethel
- Univ Paris Est Creteil, INSERM, IMRBCreteilFrance
| | | | | | | | - Marine Luka
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163ParisFrance
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163ParisFrance
| | | | - Cécile Masson
- Bioinformatics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163ParisFrance
- INSERM US24/CNRS UAR3633, Paris Cité UniversityParisFrance
| | - Mickael Ménager
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163ParisFrance
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163ParisFrance
| | | |
Collapse
|
9
|
Khan NM, Wilderman A, Kaiser JM, Kamalakar A, Goudy SL, Cotney J, Drissi H. Enhanced osteogenic potential of iPSC-derived mesenchymal progenitor cells following genome editing of GWAS variants in the RUNX1 gene. Bone Res 2024; 12:70. [PMID: 39643619 PMCID: PMC11624199 DOI: 10.1038/s41413-024-00369-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: 03/19/2024] [Revised: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 12/09/2024] Open
Abstract
Recent genome-wide association studies (GWAS) identified 518 significant loci associated with bone mineral density (BMD), including variants at the RUNX1 locus (rs13046645, rs2834676, and rs2834694). However, their regulatory impact on RUNX1 expression and bone formation remained unclear. This study utilized human induced pluripotent stem cells (iPSCs) differentiated into osteoblasts to investigate these variants' regulatory roles. CRISPR/Cas9 was employed to generate mutant (Δ) iPSC lines lacking these loci at the RUNX1 locus. Deletion lines (Δ1 and Δ2) were created in iPSCs to assess the effects of removing regions containing these loci. Deletion lines exhibited enhanced osteogenic potential, with increased expression of osteogenic marker genes and Alizarin Red staining. Circularized chromosome conformation capture (4C-Seq) was utilized to analyze interactions between BMD-associated loci and the RUNX1 promoter during osteogenesis. Analysis revealed altered chromatin interactions with multiple gene promoters including RUNX1 isoform, as well as SETD4, a histone methyltransferase, indicating their regulatory influence. Interestingly, both deletion lines notably stimulated the expression of the long isoform of RUNX1, with more modest effects on the shorter isoform. Consistent upregulation of SETD4 and other predicted targets within the Δ2 deletion suggested its removal removed a regulatory hub constraining expression of multiple genes at this locus. In vivo experiments using a bone defect model in mice demonstrated increased bone regeneration with homozygous deletion of the Δ2 region. These findings indicate that BMD-associated variants within the RUNX1 locus regulate multiple effector genes involved in osteoblast commitment, providing valuable insights into genetic regulation of bone density and potential therapeutic targets.
Collapse
Affiliation(s)
- Nazir M Khan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Andrea Wilderman
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Jarred M Kaiser
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Archana Kamalakar
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven L Goudy
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, USA
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, USA.
- Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
10
|
Mizoguchi T. In vivo dynamics of hard tissue-forming cell origins: Insights from Cre/loxP-based cell lineage tracing studies. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:109-119. [PMID: 38406212 PMCID: PMC10885318 DOI: 10.1016/j.jdsr.2024.01.003] [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: 11/20/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Bone tissue provides structural support for our bodies, with the inner bone marrow (BM) acting as a hematopoietic organ. Within the BM tissue, two types of stem cells play crucial roles: mesenchymal stem cells (MSCs) (or skeletal stem cells) and hematopoietic stem cells (HSCs). These stem cells are intricately connected, where BM-MSCs give rise to bone-forming osteoblasts and serve as essential components in the BM microenvironment for sustaining HSCs. Despite the mid-20th century proposal of BM-MSCs, their in vivo identification remained elusive owing to a lack of tools for analyzing stemness, specifically self-renewal and multipotency. To address this challenge, Cre/loxP-based cell lineage tracing analyses are being employed. This technology facilitated the in vivo labeling of specific cells, enabling the tracking of their lineage, determining their stemness, and providing a deeper understanding of the in vivo dynamics governing stem cell populations responsible for maintaining hard tissues. This review delves into cell lineage tracing studies conducted using commonly employed genetically modified mice expressing Cre under the influence of LepR, Gli1, and Axin2 genes. These studies focus on research fields spanning long bones and oral/maxillofacial hard tissues, offering insights into the in vivo dynamics of stem cell populations crucial for hard tissue homeostasis.
Collapse
|
11
|
Jia S, Liu W, Zhang M, Wang L, Ren C, Feng C, Zhang T, Lv H, Hou Z, Zou W, Zhang Y, Tong W, Wang J, Chen W. Insufficient Mechanical Loading Downregulates Piezo1 in Chondrocytes and Impairs Fracture Healing Through ApoE-Induced Senescence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400502. [PMID: 39418070 PMCID: PMC11633519 DOI: 10.1002/advs.202400502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 08/10/2024] [Indexed: 10/19/2024]
Abstract
Insufficient mechanical loading impairs fracture healing; however, the underlying mechanisms remain unclear. Increasing evidence indicates that Piezo1 plays an important role in fracture healing, although the effect of Piezo1 on the endochondral ossification of chondrocytes has been overlooked. This study reports that mechanical unloading down-regulates the expression of Piezo1 in chondrocytes and leads to fracture nonunion. Single-cell sequencing of calluses revealed that specific deletion of Piezo1 in chondrocytes upregulated the expression of apolipoprotein E (ApoE) in hypertrophic chondrocytes, resulting in delayed cartilage-to-bone transition due to enhanced chondrocyte senescence. Based on these results, an injectable and thermosensitive hydrogel is developed, which released an ApoE antagonist in situ at the fracture site. This hydrogel effectively attenuated chondrocyte senescence and, thus, promoted cartilage-to-bone transition as well as the fracture healing process. Overall, this data provide a new perspective on the activity of chondrocytes in fracture healing and a new direction for the treatment of fracture nonunion caused by insufficient mechanical loading.
Collapse
Affiliation(s)
- Siming Jia
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
- Hebei Medical University Clinical Medicine Postdoctoral Station (Hebei Medical University Third Hospital)ShijiazhuangHebei050051China
| | - Weijian Liu
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430022China
| | - Mo Zhang
- Hebei Medical University Clinical Medicine Postdoctoral Station (Hebei Medical University Third Hospital)ShijiazhuangHebei050051China
- School of PharmacyKey Laboratory of Innovative Drug Development and EvaluationHebei Medical UniversityShijiazhuang050017China
| | - Lijun Wang
- Hainan Institute of Regenerative Orthopedics and Sports Medicine, Hainan Academy of Medical Sciences and School of Basic MedicineHainan Medical UniversityHainan570000China
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Chuan Ren
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Chen Feng
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Tao Zhang
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Hongzhi Lv
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Weiguo Zou
- Hainan Institute of Regenerative Orthopedics and Sports Medicine, Hainan Academy of Medical Sciences and School of Basic MedicineHainan Medical UniversityHainan570000China
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Yingze Zhang
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Wei Tong
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430022China
| | - Juan Wang
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Wei Chen
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| |
Collapse
|
12
|
Xu R, Zhang X, Lin W, Wang Y, Zhang D, Jiang S, Liu L, Wang J, Luo X, Zhang X, Jing J, Yuan Q, Zhou C. Cathepsin K-Positive Cell Lineage Promotes In Situ Dentin Formation Controlled by Nociceptive Sonic Hedgehog. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310048. [PMID: 39474995 DOI: 10.1002/advs.202310048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 09/23/2024] [Indexed: 12/19/2024]
Abstract
Oral diseases affect nearly half of the global population throughout their lifetime causing pain, as estimated by the World Health Organization. Preservation of vital pulp is the therapeutic core as well as a challenge to protect natural teeth. Current bottleneck lies in that the regenerative capacity of injured pulp is undetermined. In this study, we identified a lifelong lineage that is labelled by cathepsin K (Ctsk) contributing to the physiological, reactionary and reparative odontogenesis of mouse molars. Ctsk+ cell-mediated dentin formation is regulated by nociceptive nerve-derived Sonic Hedgehog (Shh), especially rapidly responsive to acute injury. Notably, exogenous Shh protein to the injury pulp can preserve Ctsk+ cell capacity of odontogenesis for the nearby crown pulp and even remote root apex growth, alleviating conventionally developmental arrest in youth pulpitis. Exposed to chronical attrition, aged pulp Ctsk+ cells still hold the capacity to respond to acute stimuli and promote reparative odontogenesis, also enhanced by exogenous Shh capping. Therefore, Ctsk+ cells may be one of the lineages for accelerating precision medicine for efficient pulp treatment across ages. Shh application can be a candidate for vital pulp preservation and pulp injury repair by promoting regenerative odontogenesis to a certain extent from young adults to older individuals.
Collapse
Affiliation(s)
- Ruoshi Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaohan Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yushun Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Danting Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shuang Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Linfeng Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiaying Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xutao Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiao Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junjun Jing
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
13
|
Jacob G, Shimomura K, Nakamura N. Biologic therapies in stress fractures: Current concepts. J ISAKOS 2024; 9:100256. [PMID: 38631518 DOI: 10.1016/j.jisako.2024.04.008] [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: 11/13/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Stress fractures, a common overuse injury in physically active individuals, present a significant challenge for athletes and military personnel. Patients who sustain stress fractures have demanding training regimes where periods of rest and immobilisation have unacceptable negative consequences on sports goals and finances. Aside from being an overuse injury, there are various contributing risk factors that put certain individuals at risk of a stress fracture. The main two being nutritional deficiencies and hormonal variations, which have significant effects on bone metabolism and turnover. Historically, treatment of stress fractures focused on conservative strategies such as rest and immobilisation. Calcium and vitamin D deficiencies have been closely linked to stress fractures and so over time supplementation has also played a role in treatment. With the introduction of biologics into orthopaedics, newer treatment strategies have been applied to accelerate fracture healing and perhaps improve fracture callus quality. If such therapies can reduce time spent away from sport and activity, it would be ideal for treating stress fractures. This article aims to offer insights into the evolving landscape of stress fracture management. It investigates the pre-clinical evidence and available published clinical applications. Though fracture healing is well understood, the role of biologics for fracture healing is still indeterminate. Available literature for the use of biologic therapies in stress fractures are restricted and most reports have used biologics as a supplement to surgical fixation in subjects in studies that lack control groups. Randomised control trials have been proposed and registered by a few groups, with results awaited. Assessing individuals for risk factors, addressing hormonal imbalances and nutritional deficiencies seems like an effective approach to addressing the burden of stress fractures. We await better designed trials and studies to accurately determine the clinical benefit of adding biologics to the management of these injuries.
Collapse
Affiliation(s)
- George Jacob
- Department of Orthopaedic Surgery, Lakeshore Hospital, Cochin, India
| | - Kazunori Shimomura
- Department of Rehabilitation, Kansai University of Welfare Sciences, Osaka, Japan; Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norimasa Nakamura
- Institute for Medical Science in Sports, Osaka Health Science University, Osaka, Japan; Global Centre for Medical Engineering and Informatics, Osaka University, Osaka, Japan.
| |
Collapse
|
14
|
Shu Y, Tan Z, Pan Z, Chen Y, Wang J, He J, Wang J, Wang Y. Inhibition of inflammatory osteoclasts accelerates callus remodeling in osteoporotic fractures by enhancing CGRP +TrkA + signaling. Cell Death Differ 2024; 31:1695-1706. [PMID: 39223264 PMCID: PMC11618598 DOI: 10.1038/s41418-024-01368-5] [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: 06/08/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Impaired callus remodeling significantly contributes to the delayed healing of osteoporotic fractures; however, the underlying mechanisms remain unclear. Sensory neuronal signaling plays a crucial role in bone repair. In this study, we aimed to investigate the pathological mechanisms hindering bone remodeling in osteoporotic fractures, particularly focusing on the role of sensory neuronal signaling. We demonstrate that in ovariectomized (OVX) mice, the loss of CGRP+TrkA+ sensory neuronal signaling during callus remodeling correlates with increased Cx3cr1+iOCs expression within the bone callus. Conditional knockout of Cx3cr1+iOCs restored CGRP+TrkA+ sensory neuronal, enabling normal callus remodeling progression. Mechanistically, we further demonstrate that Cx3cr1+iOCs secrete Sema3A in the osteoporotic fracture repair microenvironment, inhibiting CGRP+TrkA+ sensory neurons' axonal regeneration and suppressing nerve-bone signaling exchange, thus hindering bone remodeling. Lastly, in human samples, we observed an association between the loss of CGRP+TrkA+ sensory neuronal signaling and increased expression of Cx3cr1+iOCs. In conclusion, enhancing CGRP+TrkA+ sensory nerve signaling by inhibiting Cx3cr1+iOCs activity presents a potential strategy for treating delayed healing in osteoporotic fractures. Inhibition of inflammatory osteoclasts enhances CGRP+TrkA+ signaling and accelerates callus remodeling in osteoporotic fractures.
Collapse
Affiliation(s)
- Yuexia Shu
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, China
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Zhenyu Tan
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
| | - Zhen Pan
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, China
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yujie Chen
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, China
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Jielin Wang
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, China
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Jieming He
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Jia Wang
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Wang
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, China.
- Department of Orthopedics, TongRen Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
15
|
Weng Y, Feng Y, Li Z, Xu S, Wu D, Huang J, Wang H, Wang Z. Zfp260 choreographs the early stage osteo-lineage commitment of skeletal stem cells. Nat Commun 2024; 15:10186. [PMID: 39582024 PMCID: PMC11586402 DOI: 10.1038/s41467-024-54640-0] [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: 06/21/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
The initial fine-tuning processes are crucial for successful bone regeneration, as they guide skeletal stem cells through progenitor differentiation toward osteo- or chondrogenic fate. While fate determination processes are well-documented, the mechanisms preceding progenitor commitment remain poorly understood. Here, we identified a transcription factor, Zfp260, as pivotal for stem cell maturation into progenitors and directing osteogenic differentiation. Zfp260 is markedly up-regulated as cells transition from stem to progenitor stages; its dysfunction causes lineage arrest at the progenitor stage, impairing bone repair. Zfp260 is required for maintaining chromatin accessibility and regulates Runx2 expression by forming super-enhancer complexes. Furthermore, the PKCα kinase phosphorylates Zfp260 at residues Y173, S182, and S197, which are essential for its functional activity. Mutations at these residues significantly impair its functionality. These findings position Zfp260 as a vital factor bridging stem cell activation with progenitor cell fate determination, unveiling a element fundamental to successful bone regeneration.
Collapse
Affiliation(s)
- Yuteng Weng
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Yanhuizhi Feng
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Zeyuan Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Shuyu Xu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Di Wu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Jie Huang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Haicheng Wang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Zuolin Wang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology, Shanghai, 200072, China.
- Department of Oral and Maxillofacial Surgery, Department of Oral Implantology, Shanghai Tongji Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
16
|
Jeong Y, Deveza L, Ortinau L, Lei K, Dawson JR, Park D. Identification of LRP1+CD13+ human periosteal stem cells that require LRP1 for bone repair. JCI Insight 2024; 9:e173831. [PMID: 39405183 PMCID: PMC11601900 DOI: 10.1172/jci.insight.173831] [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: 07/10/2023] [Accepted: 10/04/2024] [Indexed: 11/29/2024] Open
Abstract
Human periosteal skeletal stem cells (P-SSCs) are critical for cortical bone maintenance and repair. However, their in vivo identity, molecular characteristics, and specific markers remain unknown. Here, single-cell sequencing revealed human periosteum contains SSC clusters expressing known SSC markers, podoplanin (PDPN) and PDGFRA. Notably, human P-SSCs, but not bone marrow SSCs, selectively expressed identified markers low density lipoprotein receptor-related protein 1 (LRP1) and CD13. These LRP1+CD13+ human P-SSCs were perivascular cells with high osteochondrogenic but minimal adipogenic potential. Upon transplantation into bone injuries in mice, they preserved self-renewal capability in vivo. Single-cell analysis of mouse periosteum further supported the preferential expression of LRP1 and CD13 in Prx1+ P-SSCs. When Lrp1 was conditionally deleted in Prx1 lineage cells, it led to severe bone deformity, short stature, and periosteal defects. By contrast, local treatment with an LRP1 agonist at the injury sites induced early P-SSC proliferation and bone healing. Thus, human and mouse periosteum contains unique osteochondrogenic stem cell subsets, and these P-SSCs express specific markers, LRP1 and CD13, with a regulatory mechanism through LRP1 that enhances P-SSC function and bone repair.
Collapse
Affiliation(s)
| | - Lorenzo Deveza
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, USA
| | | | - Kevin Lei
- Department of Molecular and Human Genetics and
| | - John R. Dawson
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Dongsu Park
- Department of Molecular and Human Genetics and
| |
Collapse
|
17
|
Zhou JH, Wang JL, Yang D, Wu YX, Zhang W, Qin HZ, Wang C, Li J. Characteristics and Clinical Significance of Skull Defect Restoration in Young Patients: A Single-center Report and Literature Review. J Craniofac Surg 2024:00001665-990000000-02153. [PMID: 39527727 DOI: 10.1097/scs.0000000000010851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE The objective of this study is to observe cranial regeneration following cranial defects in young patients and analyze the factors influencing the regeneration process. METHODS In this retrospective study, a comparative and analytical assessment was conducted on the clinical data of 2 distinct cohorts: the observation group comprised patients aged ≤22 years with a regenerated cranial area ≥4 cm2 following cranial defects, while the control group consisted of individuals with cranial defects persisting for over 1 year and a regenerated cranial area measuring <4 cm2. These patients had been admitted to our department from August 2008 to October 2022. In addition, an analysis was undertaken to discern the pertinent factors influencing the process of cranial regeneration. RESULTS Among the 32 patients in the observation group, 9 patients developed complications after decompressive craniectomy (DC) (hereafter referred to as post-DC complications); all the patients in this group, except one, underwent cranioplasty. The control group consisted of 12 patients, all of whom had post-DC complications and underwent cranioplasty; among them, 4 patients with a coexisting complication of hydrocephalus also underwent ventriculoperitoneal shunting. Cranial regeneration was observed in all 23 patients who did not have post-DC complications and in 9 of the 21 patients with post-DC complications. The rate of cranial regeneration was lower in the group with post-DC complications than in the group without post-DC complications, and the observation time (the time from cranial defect to the completion of cranioplasty) was longer in patients in the group with post-DC complications than those in the group without post-DC complications. CONCLUSION Cranial bone regeneration can be achieved in young patients with cranial defects if factors favorable to cranial regeneration are effectively managed. These include preserving the dura mater, periosteum, and diploe during the surgery and maintaining good physical conditions postoperatively. The distribution pattern of the regenerated bones aligns with the shape of the flap. Post-DC complications represent adverse factors that can negatively affect cranial regeneration.
Collapse
Affiliation(s)
- Jia-Hua Zhou
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University
| | - Ju-Lei Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University
| | - Di Yang
- Department of Radiology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Ying-Xi Wu
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University
| | - Wei Zhang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University
| | - Huai-Zhou Qin
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University
| | - Chao Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University
| | - Jiang Li
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University
| |
Collapse
|
18
|
Cai W, Mao S, Wang Y, Gao B, Zhao J, Li Y, Chen Y, Zhang D, Yang J, Yang G. An Engineered Hierarchical Hydrogel with Immune Responsiveness and Targeted Mitochondrial Transfer to Augmented Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406287. [PMID: 39258577 PMCID: PMC11558138 DOI: 10.1002/advs.202406287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Indexed: 09/12/2024]
Abstract
Coordinating the immune response and bioenergy metabolism in bone defect environments is essential for promoting bone regeneration. Mitochondria are important organelles that control internal balance and metabolism. Repairing dysfunctional mitochondria has been proposed as a therapeutic approach for disease intervention. Here, an engineered hierarchical hydrogel with immune responsiveness can adapt to the bone regeneration environment and mediate the targeted mitochondria transfer between cells. The continuous supply of mitochondria by macrophages can restore the mitochondrial bioenergy of bone marrow mesenchymal stem cells (BMSC). Fundamentally solving the problem of insufficient energy support of BMSCs caused by local inflammation during bone repair and regeneration. This discovery provides a new therapeutic strategy for promoting bone regeneration and repair, which has research value and practical application prospects in the treatment of various diseases caused by mitochondrial dysfunction.
Collapse
Affiliation(s)
- Wenjin Cai
- Stomatology HospitalSchool of StomatologyZhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityEngineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000P. R. China
| | - Shihua Mao
- Stomatology HospitalSchool of StomatologyZhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityEngineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000P. R. China
- Zhejiang Key Laboratory of Plastic Modification and Processing TechnologyCollege of Materials Science & EngineeringZhejiang University of TechnologyHangzhou310014P. R. China
| | - Ying Wang
- Stomatology HospitalSchool of StomatologyZhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityEngineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000P. R. China
| | - Bicong Gao
- Stomatology HospitalSchool of StomatologyZhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityEngineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000P. R. China
| | - Jiaying Zhao
- Stomatology HospitalSchool of StomatologyZhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityEngineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000P. R. China
| | - Yongzheng Li
- Stomatology HospitalSchool of StomatologyZhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityEngineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000P. R. China
| | - Yani Chen
- Stomatology HospitalSchool of StomatologyZhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityEngineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000P. R. China
| | - Dong Zhang
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30318USA
| | - Jintao Yang
- Zhejiang Key Laboratory of Plastic Modification and Processing TechnologyCollege of Materials Science & EngineeringZhejiang University of TechnologyHangzhou310014P. R. China
| | - Guoli Yang
- Stomatology HospitalSchool of StomatologyZhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityEngineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000P. R. China
| |
Collapse
|
19
|
Zhang S, Zhu J, Jin S, Sun W, Ji W, Chen Z. Jawbone periosteum-derived cells with high osteogenic potential controlled by R-spondin 3. FASEB J 2024; 38:e70079. [PMID: 39340242 DOI: 10.1096/fj.202400988rr] [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: 05/02/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024]
Abstract
The jawbone periosteum, the easily accessible tissue responding to bone repair, has been overlooked in the recent development of cell therapy for jawbone defect reconstruction. Therefore, this study aimed to elucidate the in vitro and in vivo biological characteristics of jawbone periosteum-derived cells (jb-PDCs). For this purpose, we harvested the jb-PDCs from 8-week-old C57BL/6 mice. The in vitro cultured jb-PDCs (passages 1 and 3) contained skeletal stem/progenitor cells and exhibited clonogenicity and tri-lineage differentiation capacity. When implanted in vivo, the jb-PDCs (passage 3) showed evident ectopic bone formation after 4-week subcutaneous implantation, and active contribution to repair the critical-size jawbone defects in mice. Molecular profiling suggested that R-spondin 3 was strongly associated with the superior in vitro and in vivo osteogenic potentials of jb-PDCs. Overall, our study highlights the significance of comprehending the biological characteristics of the jawbone periosteum, which could pave the way for innovative cell-based therapies for the reconstruction of jawbone defects.
Collapse
Affiliation(s)
- Shu 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
| | - Jingxian Zhu
- 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
| | - Siyu Jin
- 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
| | - Wei Sun
- Department of Implantology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Ji
- 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 Implantology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- 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
| |
Collapse
|
20
|
Zhu M, Hu L, Liu Y, Chen P, Wang X, Tang B, Liu C, Zhang R, Fang J, Ren F. A Surface-Mediated Biomimetic Porous Polyether-Ether-Ketone Scaffold for Regulating Immunity and Promoting Osteogenesis. ACS Biomater Sci Eng 2024; 10:6120-6134. [PMID: 39295122 DOI: 10.1021/acsbiomaterials.4c00725] [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] [Indexed: 09/21/2024]
Abstract
The repair of critical-sized bone defects remains a major challenge for clinical orthopedic surgery. Here, we develop a surface biofunctionalized three-dimensional (3D) porous polyether-ether-ketone (PEEK) scaffold that can simultaneously promote osteogenesis and regulate macrophage polarization. The scaffold is created using polydopamine (PDA)-assisted immobilization of silk fibroin (SF) and the electrostatic self-assembly of nanocrystalline hydroxyapatite (nano-HA) on a 3D-printed porous PEEK scaffold. The SF/nano-HA functionalized surface provides a bone-like microenvironment for osteoblastic cells' adhesion, proliferation, mineralization and osteogenic differentiation. Moreover, the biofunctionalized surface can effectively drive macrophages polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Integrin β1-specific cell-matrix binding and the activation of Ca2+ receptor-mediated signaling pathway play critical roles in the regulation of macrophage polarization. Compared with the as-printed scaffold, the SF/nano-HA functionalized porous PEEK scaffold induces minimal inflammatory response, enhanced angiogenesis, and substantial new bone formation, resulting in improved osseointegration in vivo. This study not only develops a promising candidate for bone repair but also demonstrates a facile surface biofunctionalization strategy for orthopedic implants to improve osseointegration by stimulating osteogenesis and regulating immunity.
Collapse
Affiliation(s)
- Mingyu Zhu
- Department of Materials Science and Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Room 3069, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Liqiu Hu
- Department of Biomedical Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yang Liu
- Department of Biomedical Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Pinghang Chen
- Department of Materials Science and Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xiaofei Wang
- Department of Materials Science and Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Bin Tang
- Department of Biomedical Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Chao Liu
- Department of Biomedical Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Rui Zhang
- Department of Prosthodontics, Stomatology Center, Peking University Shenzhen Hospital Shenzhen, Guangdong 518036, China
| | - Ju Fang
- Department of Materials Science and Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Fuzeng Ren
- Department of Materials Science and Engineering Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
21
|
Yin B, Shen F, Ma Q, Liu Y, Han X, Cai X, Shi Y, Ye L. Identification of Postn+ periosteal progenitor cells with bone regenerative potential. JCI Insight 2024; 9:e182524. [PMID: 39377227 PMCID: PMC11466188 DOI: 10.1172/jci.insight.182524] [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: 04/29/2024] [Accepted: 08/13/2024] [Indexed: 10/09/2024] Open
Abstract
Bone contains multiple pools of skeletal stem/progenitor cells (SSPCs), and SSPCs in periosteal compartments are known to exhibit higher regenerative potential than those in BM and endosteal compartments. However, the in vivo identity and hierarchical relationships of periosteal SSPCs (P-SSPCs) remain unclear due to a lack of reliable markers to distinguish BM SSPCs and P-SSPCs. Here, we found that periosteal mesenchymal progenitor cells (P-MPs) in periosteum can be identified based on Postn-CreERT2 expression. Postn-expressing periosteal subpopulation produces osteolineage descendants that fuel bones to maintain homeostasis and support regeneration. Notably, Postn+ P-MPs are likely derived from Gli1+ skeletal stem cells (SSCs). Ablation of Postn+ cells results in impairments in homeostatic cortical bone architecture and defects in fracture repair. Genetic deletion of Igf1r in Postn+ cells dampens bone fracture healing. In summary, our study provides a mechanistic understanding of bone regeneration through the regulation of region-specific Postn+ P-MPs.
Collapse
Affiliation(s)
- Bei Yin
- State Key Laboratory of Oral Diseases
- National Center for Stomatology
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology
- Department of Endodontics, West China School of Stomatology
| | - Fangyuan Shen
- State Key Laboratory of Oral Diseases
- National Center for Stomatology
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology
| | - Qingge Ma
- State Key Laboratory of Oral Diseases
- National Center for Stomatology
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology
- Department of Endodontics, West China School of Stomatology
| | | | - Xianglong Han
- State Key Laboratory of Oral Diseases
- National Center for Stomatology
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology
- Department of Orthodontics, West China School of Stomatology, and
| | - Xuyu Cai
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Shi
- State Key Laboratory of Oral Diseases
- National Center for Stomatology
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology
| | - Ling Ye
- State Key Laboratory of Oral Diseases
- National Center for Stomatology
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology
- Department of Endodontics, West China School of Stomatology
| |
Collapse
|
22
|
Melis S, Trompet D, Chagin AS, Maes C. Skeletal stem and progenitor cells in bone physiology, ageing and disease. Nat Rev Endocrinol 2024:10.1038/s41574-024-01039-y. [PMID: 39379711 DOI: 10.1038/s41574-024-01039-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 10/10/2024]
Abstract
Skeletal stem cells (SSCs) and related progenitors with osteogenic potential, collectively termed skeletal stem and/or progenitor cells (SSPCs), are crucial for providing osteoblasts for bone formation during homeostatic tissue turnover and fracture repair. Besides mediating normal bone physiology, they also have important roles in various metabolic bone diseases, including osteoporosis. SSPCs are of tremendous interest because they represent prime future targets for osteoanabolic therapies and bone regenerative medicine. Remarkable progress has been made in characterizing various SSC and SSPC populations in postnatal bone. SSPCs exist in the periosteum and within the bone marrow stroma, including subsets localizing around arteriolar and sinusoidal blood vessels; they can display osteogenic, chondrogenic, adipogenic and/or fibroblastic potential, and exert critical haematopoiesis-supportive functions. However, much remains to be clarified. By the current markers, bona fide SSCs are commonly contained within broader SSPC populations characterized by considerable heterogeneity and overlap, whose common versus specific functions in health and disease have not been fully unravelled. Here, we review the present knowledge of the identity, fates and relationships of SSPC populations in the postnatal bone environment, their contributions to bone maintenance, the changes observed upon ageing, and the effect of metabolic diseases such as osteoporosis and diabetes mellitus.
Collapse
Affiliation(s)
- Seppe Melis
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Andrei S Chagin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| |
Collapse
|
23
|
Suhardi VJ, Oktarina A, Hammad M, Niu Y, Li Q, Thomson A, Lopez J, McCormick J, Ayturk UM, Greenblatt MB, Ivashkiv LB, Bostrom MPG, Yang X. Prevention and treatment of peri-implant fibrosis by functionally inhibiting skeletal cells expressing the leptin receptor. Nat Biomed Eng 2024; 8:1285-1307. [PMID: 39085645 DOI: 10.1038/s41551-024-01238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024]
Abstract
The cellular and molecular mediators of peri-implant fibrosis-a most common reason for implant failure and for surgical revision after the replacement of a prosthetic joint-remain unclear. Here we show that peri-implant fibrotic tissue in mice and humans is largely composed of a specific population of skeletal cells expressing the leptin receptor (LEPR) and that these cells are necessary and sufficient to generate and maintain peri-implant fibrotic tissue. In a mouse model of tibial implantation and osseointegration that mimics partial knee arthroplasty, genetic ablation of LEPR+ cells prevented peri-implant fibrosis and the implantation of LEPR+ cells from peri-implant fibrotic tissue was sufficient to induce fibrosis in secondary hosts. Conditional deletion of the adhesion G-protein-coupled receptor F5 (ADGRF5) in LEPR+ cells attenuated peri-implant fibrosis while augmenting peri-implant bone formation, and ADGRF5 inhibition by the intra-articular or systemic administration of neutralizing anti-ADGRF5 in the mice prevented and reversed peri-implant fibrosis. Pharmaceutical agents that inhibit the ADGRF5 pathway in LEPR+ cells may be used to prevent and treat peri-implant fibrosis.
Collapse
Affiliation(s)
- Vincentius Jeremy Suhardi
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | | | - Mohammed Hammad
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Yingzhen Niu
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Joint Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Qingdian Li
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedics, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Andrew Thomson
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Juan Lopez
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Jason McCormick
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Ugur M Ayturk
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Matthew B Greenblatt
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Mathias P G Bostrom
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, NY, USA.
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
24
|
Li Z, Lin J, Wu J, Suo J, Wang Z. The Hippo signalling pathway in bone homeostasis: Under the regulation of mechanics and aging. Cell Prolif 2024; 57:e13652. [PMID: 38700015 PMCID: PMC11471399 DOI: 10.1111/cpr.13652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
The Hippo signalling pathway is a conserved kinase cascade that orchestrates diverse cellular processes, such as proliferation, apoptosis, lineage commitment and stemness. With the onset of society ages, research on skeletal aging-mechanics-bone homeostasis has exploded. In recent years, aging and mechanical force in the skeletal system have gained groundbreaking research progress. Under the regulation of mechanics and aging, the Hippo signalling pathway has a crucial role in the development and homeostasis of bone. We synthesize the current knowledge on the role of the Hippo signalling pathway, particularly its downstream effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), in bone homeostasis. We discuss the regulation of the lineage specification and function of different skeletal cell types by the Hippo signalling pathway. The interactions of the Hippo signalling pathway with other pathways, such as Wnt, transforming growth factor beta and nuclear factor kappa-B, are also mentioned because of their importance for modulating bone homeostasis. Furthermore, YAP/TAZ have been extensively studied as mechanotransducers. Due to space limitations, we focus on reviewing how mechanical forces and aging influence cell fate, communications and homeostasis through a dysregulated Hippo signalling pathway.
Collapse
Affiliation(s)
- Zhengda Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Junqing Lin
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Jing Wu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Zuoyun Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| |
Collapse
|
25
|
Nakamura K, Tsukasaki M, Tsunematsu T, Yan M, Ando Y, Huynh NCN, Hashimoto K, Gou Q, Muro R, Itabashi A, Iguchi T, Okamoto K, Nakamura T, Nakano K, Okamura T, Ueno T, Ito K, Ishimaru N, Hoshi K, Takayanagi H. The periosteum provides a stromal defence against cancer invasion into the bone. Nature 2024; 634:474-481. [PMID: 39169177 DOI: 10.1038/s41586-024-07822-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 07/12/2024] [Indexed: 08/23/2024]
Abstract
The periosteum is the layer of cells that covers nearly the entire surface of every bone. Upon infection, injury or malignancy the bone surface undergoes new growth-the periosteal reaction-but the mechanism and physiological role of this process remain unknown1,2. Here we show that the periosteal reaction protects against cancer invasion into the bone. Histological analyses of human lesions of head and neck squamous cell carcinomas (HNSCCs) show that periosteal thickening occurs in proximity to the tumour. We developed a genetically dissectible mouse model of HNSCC and demonstrate that inducible depletion of periosteal cells accelerates cancerous invasion of the bone. Single-cell RNA sequencing reveals that expression of the gene encoding the protease inhibitor TIMP1 is markedly increased in the periosteum at the pre-invasive stage. This increase is due to upregulation of HIF1α expression in the tumour microenvironment, and increased TIMP1 inactivates matrix-degrading proteases, promoting periosteal thickening to inhibit cancer invasion. Genetic deletion of Timp1 impairs periosteal expansion, exacerbating bone invasion and decreasing survival in tumour-bearing mice. Together, these data show that the periosteal reaction may act as a functional stromal barrier against tumour progression, representing a unique example of tissue immunity mediated by stromal cells.
Collapse
Affiliation(s)
- Kazutaka Nakamura
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Oral and Maxillofacial Surgery, Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masayuki Tsukasaki
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Takaaki Tsunematsu
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Minglu Yan
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yutaro Ando
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nam Cong-Nhat Huynh
- Laboratory of Oral-Maxillofacial Biology, Faculty of Odonto-Stomatology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Kyoko Hashimoto
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Qiao Gou
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryunosuke Muro
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Ayumi Itabashi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiro Iguchi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Immune Environment Dynamics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tomoya Ueno
- Department of Molecular Tumor Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kosei Ito
- Department of Molecular Tumor Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Naozumi Ishimaru
- Department of Oral Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuto Hoshi
- Oral and Maxillofacial Surgery, Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
26
|
Bok S, Sun J, Greenblatt MB. Are osteoblasts multiple cell types? A new diversity in skeletal stem cells and their derivatives. J Bone Miner Res 2024; 39:1386-1392. [PMID: 39052334 PMCID: PMC11425698 DOI: 10.1093/jbmr/zjae109] [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: 04/15/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 07/27/2024]
Abstract
Only in the past decade have skeletal stem cells (SSCs), a cell type displaying formal evidence of stemness and serving as the ultimate origin of mature skeletal cell types such as osteoblasts, been defined. Here, we discuss a pair of recent reports that identify that SSCs do not represent a single cell type, but rather a family of related cells that each have characteristic anatomic locations and distinct functions tailored to the physiology of those sites. The distinct functional properties of these SSCs in turn provide a basis for the diseases of their respective locations. This concept emerges from one report identifying a distinct vertebral skeletal stem cell driving the high rate of breast cancer metastasis to the spine over other skeletal sites and a report identifying 2 SSCs in the calvaria that interact to mediate both physiologic calvarial mineralization and pathologic calvarial suture fusion in craniosynostosis. Despite displaying functional differences, these SSCs are each united by shared features including a shared series of surface markers and parallel differentiation hierarchies. We propose that this diversity at the level of SSCs in turn translates into a similar diversity at the level of mature skeletal cell types, including osteoblasts, with osteoblasts derived from different SSCs each displaying different functional and transcriptional characteristics reflecting their cell of origin. In this model, osteoblasts would represent not a single cell type, but rather a family of related cells each with distinct functions, paralleling the functional diversity in SSCs.
Collapse
Affiliation(s)
- Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York NY 10065, United States
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York NY 10065, United States
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York NY 10065, United States
- Skeletal Health and Orthopedic Research Program, Hospital for Special Surgery, New York NY 10065, United States
| |
Collapse
|
27
|
Shi Y, Peng J, Liu M, Qi X, Li S, Li Q, Jiang Q, Zheng L, Xu J, Zhao Y, Zhang Y. Nicotinamide mononucleotide enhances fracture healing by promoting skeletal stem cell proliferation. Theranostics 2024; 14:5999-6015. [PMID: 39346542 PMCID: PMC11426247 DOI: 10.7150/thno.98149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/04/2024] [Indexed: 10/01/2024] Open
Abstract
The process of skeletal regeneration initiated by stem cells following injury, especially in fractures, is significantly impaired by aging and adverse factors. Nicotinamide mononucleotide (NMN), a critical endogenous precursor of nicotinamide adenine dinucleotide (NAD), has garnered extensive attention for its multifaceted regulatory functions in living organisms and its wide-ranging therapeutic potential. However, whether NMN contributes to trauma-induced skeletal regeneration remains unclear. Methods: The transverse femoral shaft fracture model was employed to evaluate the potential advantages of NMN administration for overall repair during the initial fracture stages in male mice through micro-CT analysis, histochemistry, and biomechanical testing. The pro-proliferative function of NMN on skeletal stem cells (SSCs) was investigated through flow cytometry, qRT-PCR, NAD content measurement, and cell proliferation assay. Results: In this study, we observed that the administration of NMN during the initial phase of fracture in mice led to a larger callus and corresponding improvement in micro-CT parameters. NMN enhances the cartilaginous component of the callus by elevating the NAD content, consequently accelerating subsequent endochondral ossification and the fracture healing process. Subsequent analyses elucidated that NMN was beneficial in promoting the expansion of diverse stem cells in vivo and in vitro potentially via modulation of the Notch signaling pathway. Moreover, the depletion of macrophages profoundly obstructs the proliferation of SSCs. Conclusion: Our discoveries provide a potential strategy for enhancing fracture healing through stimulation of callus SSC proliferation at an early stage, shedding light on the translational value of NMN as an enhancer for skeletal regeneration and highlighting the pivotal role of macrophage-stem cell interactions in governing the regenerative influence of NMN on stem cells.
Collapse
Affiliation(s)
- Yitian Shi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Jiayin Peng
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, PR China
| | - Mengfan Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Xiling Qi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Siyu Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
| | - Qiangqiang Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu 210008, PR China
| | - Liming Zheng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Yun Zhao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, PR China
| | - Yifeng Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, PR China
- Shanghai Clinical Research and Trial Center, Shanghai 200000, PR China
| |
Collapse
|
28
|
Xing W, Feng H, Jiang B, Gao B, Liu J, Xie Z, Zhang Y, Hu X, Sun J, Greenblatt MB, Zhou BO, Zou W. Itm2a expression marks periosteal skeletal stem cells that contribute to bone fracture healing. J Clin Invest 2024; 134:e176528. [PMID: 39225088 PMCID: PMC11364384 DOI: 10.1172/jci176528] [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: 10/10/2023] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
The periosteum contains skeletal stem/progenitor cells that contribute to bone fracture healing. However, the in vivo identity of periosteal skeletal stem cells (P-SSCs) remains unclear, and membrane protein markers of P-SSCs that facilitate tissue engineering are needed. Here, we identified integral membrane protein 2A (Itm2a) enriched in SSCs using single-cell transcriptomics. Itm2a+ P-SSCs displayed clonal multipotency and self-renewal and sat at the apex of their differentiation hierarchy. Lineage-tracing experiments showed that Itm2a selectively labeled the periosteum and that Itm2a+ cells were preferentially located in the outer fibrous layer of the periosteum. The Itm2a+ cells rarely expressed CD34 or Osx, but expressed periosteal markers such as Ctsk, CD51, PDGFRA, Sca1, and Gli1. Itm2a+ P-SSCs contributed to osteoblasts, chondrocytes, and marrow stromal cells upon injury. Genetic lineage tracing using dual recombinases showed that Itm2a and Prrx1 lineage cells generated spatially separated subsets of chondrocytes and osteoblasts during fracture healing. Bone morphogenetic protein 2 (Bmp2) deficiency or ablation of Itm2a+ P-SSCs resulted in defects in fracture healing. ITM2A+ P-SSCs were also present in the human periosteum. Thus, our study identified a membrane protein marker that labels P-SSCs, providing an attractive target for drug and cellular therapy for skeletal disorders.
Collapse
Affiliation(s)
- Wenhui Xing
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, China
| | - Heng Feng
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bo Jiang
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bo Gao
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Institute of Orthopaedic Surgery, Xijing Hospital, Air Force Military Medical University, Xi’an, Shaanxi, China
| | - Jiping Liu
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zaiqi Xie
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yazhuo Zhang
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xuye Hu
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Matthew B. Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
- Research Division, Hospital for Special Surgery, New York, New York, USA
| | - Bo O. Zhou
- Key Laboratory of Multi-Cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Weiguo Zou
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Kuroda Y, Yoda M, Kawaai K, Tatenuma M, Mizoguchi T, Ito S, Kasahara M, Wu Y, Takano H, Momose A, Matsuo K. Developing long bones respond to surrounding tissues by trans-pairing of periosteal osteoclasts and endocortical osteoblasts. Development 2024; 151:dev202194. [PMID: 39119717 PMCID: PMC11423808 DOI: 10.1242/dev.202194] [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: 07/18/2023] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Developing long bones alter their shape while maintaining uniform cortical thickness via coordinated activity of bone-forming osteoblasts and bone-resorbing osteoclasts at periosteal and endosteal surfaces, a process we designate trans-pairing. Two types of trans-pairing shift cortical bone in opposite orientations: peri-forming trans-pairing (peri-t-p) increases bone marrow space and endo-forming trans-pairing (endo-t-p) decreases it, via paired activity of bone resorption and formation across the cortex. Here, we focused on endo-t-p in growing bones. Analysis of endo-t-p activity in the cortex of mouse fibulae revealed osteoclasts under the periosteum compressed by muscles, and expression of RANKL in periosteal cells of the cambium layer. Furthermore, mature osteoblasts were localized on the endosteum, while preosteoblasts were at the periosteum and within cortical canals. X-ray tomographic microscopy revealed the presence of cortical canals more closely associated with endo- than with peri-t-p. Sciatic nerve transection followed by muscle atrophy and unloading induced circumferential endo-t-p with concomitant spread of cortical canals. Such canals likely supply the endosteum with preosteoblasts from the periosteum under endo-t-p, allowing bone shape to change in response to mechanical stress or nerve injury.
Collapse
Affiliation(s)
- Yukiko Kuroda
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masaki Yoda
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Katsuhiro Kawaai
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Motoharu Tatenuma
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | | | - Shinichirou Ito
- Department of Pharmacology, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Masataka Kasahara
- Department of Pharmacology, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Yanlin Wu
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Katahira 2-1-1, Aoba, Sendai Miyagi 980-8577, Japan
| | - Hidekazu Takano
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Katahira 2-1-1, Aoba, Sendai Miyagi 980-8577, Japan
| | - Atsushi Momose
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Katahira 2-1-1, Aoba, Sendai Miyagi 980-8577, Japan
- JASRI/SPring-8, 1-1-1 Kouto, Sayo-cho, Hyogo 679-5198, Japan
| | - Koichi Matsuo
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| |
Collapse
|
30
|
Vu EK, Karkache IY, Pham A, Koroth J, Bradley EW. Hdac3 deficiency limits periosteal reaction associated with Western diet feeding in female mice. J Cell Mol Med 2024; 28:e70081. [PMID: 39261913 PMCID: PMC11390340 DOI: 10.1111/jcmm.70081] [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: 07/03/2024] [Revised: 08/25/2024] [Accepted: 09/01/2024] [Indexed: 09/13/2024] Open
Abstract
Diet-induced obesity is associated with enhanced systemic inflammation that limits bone regeneration. HDAC inhibitors are currently being explored as anti-inflammatory agents. Prior reports show that myeloid progenitor-directed Hdac3 ablation enhances intramembranous bone healing in female mice. In this study, we determined if Hdac3 ablation increased intramembranous bone regeneration in mice fed a high-fat/high-sugar (HFD) diet. Micro-CT analyses demonstrated that HFD-feeding enhanced the formation of periosteal reaction tissue of control littermates, reflective of suboptimal bone healing. We confirmed enhanced bone volume within the defect of Hdac3-ablated females and showed that Hdac3 ablation reduced the amount of periosteal reaction tissue following HFD feeding. Osteoblasts cultured in a conditioned medium derived from Hdac3-ablated cells exhibited a four-fold increase in mineralization and enhanced osteogenic gene expression. We found that Hdac3 ablation elevated the secretion of several chemokines, including CCL2. We then confirmed that Hdac3 deficiency increased the expression of Ccl2. Lastly, we show that the proportion of CCL2-positve cells within bone defects was significantly higher in Hdac3-deficient mice and was further enhanced by HFD. Overall, our studies demonstrate that Hdac3 deletion enhances intramembranous bone healing in a setting of diet-induced obesity, possibly through increased production of CCL2 by macrophages within the defect.
Collapse
Affiliation(s)
- Elizabeth K. Vu
- Department of Orthopedic SurgeryMedical School, University of MinnesotaMinneapolisMNUSA
| | - Ismael Y. Karkache
- Comparative Molecular BiosciencesSchool of Veterinary MedicineSt. PaulMNUSA
| | - Anthony Pham
- Department of Orthopedic SurgeryMedical School, University of MinnesotaMinneapolisMNUSA
| | - Jinsha Koroth
- Department of Orthopedic SurgeryMedical School, University of MinnesotaMinneapolisMNUSA
| | - Elizabeth W. Bradley
- Department of Orthopedic SurgeryMedical School, University of MinnesotaMinneapolisMNUSA
- Comparative Molecular BiosciencesSchool of Veterinary MedicineSt. PaulMNUSA
- Stem Cell Institute, University of MinnesotaMinneapolisMNUSA
| |
Collapse
|
31
|
Alikhani M, Alikhani M, Sangsuwon C, Oliveira SP, Abdullah F, Teixeira CC. Periosteum response to static forces stimulates cortical drifting: A new orthopedic target. J World Fed Orthod 2024:S2212-4438(24)00051-1. [PMID: 39209694 DOI: 10.1016/j.ejwf.2024.07.003] [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: 03/27/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The mechanism of cortical bone adaptation to static forces is not well understood. This is an important process because static forces are applied to the cortical bone in response to the growth of soft tissues and during Orthodontic and Orthopedic corrections. The aim of this study was to investigate the cortical bone response to expanding forces applied to the maxilla. METHODS Overall, 375 adult Sprague-Dawley rats were divided into three groups: 1) static force group, 2) static force plus stimulation group, and 3) sham group. In addition to static force across the maxilla, some animals were exposed to anti-inflammatory medication. Samples were collected at different time points and evaluated by micro-computed tomography, fluorescence microscopy, immunohistochemistry, and gene and protein analyses. RESULTS The application of expansion forces to the maxilla increased inflammation in the periosteum and activated osteoclasts on the surface of the cortical plate. This activation was independent of the magnitude of tooth movement but followed the pattern of skeletal displacement. Bone formation on the surface of the cortical plate occurred at a later stage and resulted in the relocation of the cortical boundary of the maxilla and cortical drifting. CONCLUSIONS This study demonstrates that cortical bone adaptation to static forces originates from the periosteum, and it is an inflammatory-based phenomenon that can be manipulated by the clinician. Our findings support a new theory for cortical adaptation to static forces and an innovative clinical approach to promote cortical drifting through periosteal stimulation. Being able to control cortical drift can have a significant impact on clinical orthodontic and dentofacial orthopedics by allowing corrections of severe deformities without the need for maxillofacial surgery.
Collapse
Affiliation(s)
- Mani Alikhani
- Advanced Education Program in Orthodontics & Dentofacial Orthopedics, CTOR Academy, Hoboken, New Jersey; Advanced Graduate Education Program in Orthodontics, Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Mona Alikhani
- Advanced Education Program in Orthodontics & Dentofacial Orthopedics, CTOR Academy, Hoboken, New Jersey
| | - Chinapa Sangsuwon
- Advanced Education Program in Orthodontics & Dentofacial Orthopedics, CTOR Academy, Hoboken, New Jersey
| | - Serafim P Oliveira
- Advanced Education Program in Orthodontics & Dentofacial Orthopedics, CTOR Academy, Hoboken, New Jersey; CISeD Research Center in Digital Services, Polytechnic University of Viseu, Viseu, Portugal
| | - Fanar Abdullah
- Advanced Education Program in Orthodontics & Dentofacial Orthopedics, CTOR Academy, Hoboken, New Jersey
| | - Cristina C Teixeira
- Department of Orthodontics, New York University College of Dentistry, New York, New York.
| |
Collapse
|
32
|
Li N, Shi B, Li Z, Han J, Sun J, Huang H, Yallowitz AR, Bok S, Xiao S, Wu Z, Chen Y, Xu Y, Qin T, Huang R, Zheng H, Shen R, Meng L, Greenblatt MB, Xu R. Schnurri-3 inhibition rescues skeletal fragility and vascular skeletal stem cell niche pathology in the OIM model of osteogenesis imperfecta. Bone Res 2024; 12:46. [PMID: 39183236 PMCID: PMC11345453 DOI: 10.1038/s41413-024-00349-1] [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: 07/09/2023] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 08/27/2024] Open
Abstract
Osteogenesis imperfecta (OI) is a disorder of low bone mass and increased fracture risk due to a range of genetic variants that prominently include mutations in genes encoding type I collagen. While it is well known that OI reflects defects in the activity of bone-forming osteoblasts, it is currently unclear whether OI also reflects defects in the many other cell types comprising bone, including defects in skeletal vascular endothelium or the skeletal stem cell populations that give rise to osteoblasts and whether correcting these broader defects could have therapeutic utility. Here, we find that numbers of skeletal stem cells (SSCs) and skeletal arterial endothelial cells (AECs) are augmented in Col1a2oim/oim mice, a well-studied animal model of moderate to severe OI, suggesting that disruption of a vascular SSC niche is a feature of OI pathogenesis. Moreover, crossing Col1a2oim/oim mice to mice lacking a negative regulator of skeletal angiogenesis and bone formation, Schnurri 3 (SHN3), not only corrected the SSC and AEC phenotypes but moreover robustly corrected the bone mass and spontaneous fracture phenotypes. As this finding suggested a strong therapeutic utility of SHN3 inhibition for the treatment of OI, a bone-targeting AAV was used to mediate Shn3 knockdown, rescuing the Col1a2oim/oim phenotype and providing therapeutic proof-of-concept for targeting SHN3 for the treatment of OI. Overall, this work both provides proof-of-concept for inhibition of the SHN3 pathway and more broadly addressing defects in the stem/osteoprogenitor niche as is a strategy to treat OI.
Collapse
Affiliation(s)
- Na Li
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Baohong Shi
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Zan Li
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Jie Han
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jun Sun
- Research Division, Hospital for Special Surgery, New York, NY, 10065, USA
| | - Haitao Huang
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Shuang Xiao
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Zuoxing Wu
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yu Chen
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yan Xu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Tian Qin
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Rui Huang
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Haiping Zheng
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Rong Shen
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Lin Meng
- Department of Electronic and Computer Engineering, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Matthew B Greenblatt
- Research Division, Hospital for Special Surgery, New York, NY, 10065, USA.
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| | - Ren Xu
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China.
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
33
|
Xing X, Li Z, Xu J, Chen AZ, Archer M, Wang Y, Xu M, Wang Z, Zhu M, Qin Q, Thottappillil N, Zhou M, James AW. Requirement of Pdgfrα+ cells for calvarial bone repair. Stem Cells Transl Med 2024; 13:791-802. [PMID: 38986535 PMCID: PMC11328938 DOI: 10.1093/stcltm/szae041] [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: 02/29/2024] [Accepted: 05/19/2024] [Indexed: 07/12/2024] Open
Abstract
Platelet-derived growth factor receptor α (PDGFRα) is often considered as a general marker of mesenchymal cells and fibroblasts, but also shows expression in a portion of osteoprogenitor cells. Within the skeleton, Pdgfrα+ mesenchymal cells have been identified in bone marrow and periosteum of long bones, where they play a crucial role in participating in fracture repair. A similar examination of Pdgfrα+ cells in calvarial bone healing has not been examined. Here, we utilize Pdgfrα-CreERTM;mT/mG reporter animals to examine the contribution of Pdgfrα+ mesenchymal cells to calvarial bone repair through histology and single-cell RNA sequencing (scRNA-Seq). Results showed that Pdgfrα+ mesenchymal cells are present in several cell clusters by scRNA-Seq, and by histology a dramatic increase in Pdgfrα+ cells populated the defect site at early timepoints to give rise to healed bone tissue overtime. Notably, diphtheria toxin-mediated ablation of Pdgfrα reporter+ cells resulted in significantly impaired calvarial bone healing. Our findings suggest that Pdgfrα-expressing cells within the calvarial niche play a critical role in the process of calvarial bone repair.
Collapse
Affiliation(s)
- Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Austin Z Chen
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Ziyi Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Manyu Zhu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Neelima Thottappillil
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Myles Zhou
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
34
|
Farmer DT, Dukov JE, Chen HJ, Arata C, Hernandez-Trejo J, Xu P, Teng CS, Maxson RE, Crump JG. Cellular transitions during cranial suture establishment in zebrafish. Nat Commun 2024; 15:6948. [PMID: 39138165 PMCID: PMC11322166 DOI: 10.1038/s41467-024-50780-5] [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: 03/17/2023] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
Cranial sutures separate neighboring skull bones and are sites of bone growth. A key question is how osteogenic activity is controlled to promote bone growth while preventing aberrant bone fusions during skull expansion. Using single-cell transcriptomics, lineage tracing, and mutant analysis in zebrafish, we uncover key developmental transitions regulating bone formation at sutures during skull expansion. In particular, we identify a subpopulation of mesenchyme cells in the mid-suture region that upregulate a suite of genes including BMP antagonists (e.g. grem1a) and pro-angiogenic factors. Lineage tracing with grem1a:nlsEOS reveals that this mid-suture subpopulation is largely non-osteogenic. Moreover, combinatorial mutation of BMP antagonists enriched in this mid-suture subpopulation results in increased BMP signaling in the suture, misregulated bone formation, and abnormal suture morphology. These data reveal establishment of a non-osteogenic mesenchyme population in the mid-suture region that restricts bone formation through local BMP antagonism, thus ensuring proper suture morphology.
Collapse
Affiliation(s)
- D'Juan T Farmer
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, 90095, USA.
| | - Jennifer E Dukov
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Hung-Jhen Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Claire Arata
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jose Hernandez-Trejo
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Pengfei Xu
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Camilla S Teng
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Robert E Maxson
- Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - J Gage Crump
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
35
|
Li Z, Shi B, Li N, Sun J, Zeng X, Huang R, Bok S, Chen X, Han J, Yallowitz AR, Debnath S, Cung M, Ling Z, Zhong CQ, Hong Y, Li G, Koenen M, Cohen P, Su X, Lu H, Greenblatt MB, Xu R. Bone controls browning of white adipose tissue and protects from diet-induced obesity through Schnurri-3-regulated SLIT2 secretion. Nat Commun 2024; 15:6697. [PMID: 39107299 PMCID: PMC11303806 DOI: 10.1038/s41467-024-51155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
The skeleton has been suggested to function as an endocrine organ controlling whole organism energy balance, however the mediators of this effect and their molecular links remain unclear. Here, utilizing Schnurri-3-/- (Shn3-/-) mice with augmented osteoblast activity, we show Shn3-/-mice display resistance against diet-induced obesity and enhanced white adipose tissue (WAT) browning. Conditional deletion of Shn3 in osteoblasts but not adipocytes recapitulates lean phenotype of Shn3-/-mice, indicating this phenotype is driven by skeleton. We further demonstrate osteoblasts lacking Shn3 can secrete cytokines to promote WAT browning. Among them, we identify a C-terminal fragment of SLIT2 (SLIT2-C), primarily secreted by osteoblasts, as a Shn3-regulated osteokine that mediates WAT browning. Lastly, AAV-mediated Shn3 silencing phenocopies the lean phenotype and augmented glucose metabolism. Altogether, our findings establish a novel bone-fat signaling axis via SHN3 regulated SLIT2-C production in osteoblasts, offering a potential therapeutic target to address both osteoporosis and metabolic syndrome.
Collapse
Affiliation(s)
- Zan Li
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- PET Center, Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baohong Shi
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Na Li
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Xiangchen Zeng
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Rui Huang
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Xiaohui Chen
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Jie Han
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michelle Cung
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Zheng Ling
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Chuan-Qi Zhong
- College of Life Science, Xiamen University, Xiamen, China
| | - Yixang Hong
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Gang Li
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Mascha Koenen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Xinhui Su
- PET Center, Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Research Division, Hospital for Special Surgery, New York, NY, USA.
| | - Ren Xu
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China.
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
36
|
Otsuru S, Kodama J, Oichi T, Wilkinson K, Abzug J, Kaito T, Iwamoto-Enomoto M, Iwamoto M. Apolipoprotein E is a novel marker for chondrocytes in the growth plate resting zone. RESEARCH SQUARE 2024:rs.3.rs-4656728. [PMID: 39149484 PMCID: PMC11326366 DOI: 10.21203/rs.3.rs-4656728/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
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 Apoe-mCherry 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 both in vitro and in vivo. Moreover, APOE was detected in human growth plate RCs. These findings suggest that Apoe is a novel pan-RC marker in both mouse and human growth plates.
Collapse
|
37
|
Babey ME, Krause WC, Chen K, Herber CB, Torok Z, Nikkanen J, Rodriguez R, Zhang X, Castro-Navarro F, Wang Y, Wheeler EE, Villeda S, Leach JK, Lane NE, Scheller EL, Chan CKF, Ambrosi TH, Ingraham HA. A maternal brain hormone that builds bone. Nature 2024; 632:357-365. [PMID: 38987585 PMCID: PMC11306098 DOI: 10.1038/s41586-024-07634-3] [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: 09/16/2023] [Accepted: 05/30/2024] [Indexed: 07/12/2024]
Abstract
In lactating mothers, the high calcium (Ca2+) demand for milk production triggers significant bone loss1. Although oestrogen normally counteracts excessive bone resorption by promoting bone formation, this sex steroid drops precipitously during this postpartum period. Here we report that brain-derived cellular communication network factor 3 (CCN3) secreted from KISS1 neurons of the arcuate nucleus (ARCKISS1) fills this void and functions as a potent osteoanabolic factor to build bone in lactating females. We began by showing that our previously reported female-specific, dense bone phenotype2 originates from a humoral factor that promotes bone mass and acts on skeletal stem cells to increase their frequency and osteochondrogenic potential. This circulatory factor was then identified as CCN3, a brain-derived hormone from ARCKISS1 neurons that is able to stimulate mouse and human skeletal stem cell activity, increase bone remodelling and accelerate fracture repair in young and old mice of both sexes. The role of CCN3 in normal female physiology was revealed after detecting a burst of CCN3 expression in ARCKISS1 neurons coincident with lactation. After reducing CCN3 in ARCKISS1 neurons, lactating mothers lost bone and failed to sustain their progeny when challenged with a low-calcium diet. Our findings establish CCN3 as a potentially new therapeutic osteoanabolic hormone for both sexes and define a new maternal brain hormone for ensuring species survival in mammals.
Collapse
Affiliation(s)
- Muriel E Babey
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Francisco, San Francisco, CA, USA
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Kun Chen
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, CA, USA
| | - Candice B Herber
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Denali Therapeutics, South San Francisco, CA, USA
| | - Zsofia Torok
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Joni Nikkanen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ruben Rodriguez
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Carmot Therapeutics, Berkeley, CA, USA
| | - Xiao Zhang
- Department of Medicine, Washington University, St Louis, MO, USA
| | - Fernanda Castro-Navarro
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Yuting Wang
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Erika E Wheeler
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Saul Villeda
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Nancy E Lane
- Department of Medicine, Division of Rheumatology, University of California, Davis, Sacramento, CA, USA
| | - Erica L Scheller
- Department of Medicine, Washington University, St Louis, MO, USA
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas H Ambrosi
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, CA, USA.
| | - Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
38
|
Khan MP, Sabini E, Beigel K, Lanzolla G, Laslow B, Wang D, Merceron C, Giaccia A, Long F, Taylor D, Schipani E. HIF1 activation safeguards cortical bone formation against impaired oxidative phosphorylation. JCI Insight 2024; 9:e182330. [PMID: 39088272 PMCID: PMC11457864 DOI: 10.1172/jci.insight.182330] [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: 04/30/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024] Open
Abstract
Energy metabolism, through pathways such as oxidative phosphorylation (OxPhos) and glycolysis, plays a pivotal role in cellular differentiation and function. Our study investigates the impact of OxPhos disruption in cortical bone development by deleting mitochondrial transcription factor A (TFAM). TFAM controls OxPhos by regulating the transcription of mitochondrial genes. The cortical bone, constituting the long bones' rigid shell, is sheathed by the periosteum, a connective tissue layer populated with skeletal progenitors that spawn osteoblasts, the bone-forming cells. TFAM-deficient mice presented with thinner cortical bone, spontaneous midshaft fractures, and compromised periosteal cell bioenergetics, characterized by reduced ATP levels. Additionally, they exhibited an enlarged periosteal progenitor cell pool with impaired osteoblast differentiation. Increasing hypoxia-inducible factor 1a (HIF1) activity within periosteal cells substantially mitigated the detrimental effects induced by TFAM deletion. HIF1 is known to promote glycolysis in all cell types. Our findings underscore the indispensability of OxPhos for the proper accrual of cortical bone mass and indicate a compensatory mechanism between OxPhos and glycolysis in periosteal cells. The study opens new avenues for understanding the relationship between energy metabolism and skeletal health and suggests that modulating bioenergetic pathways may provide a therapeutic avenue for conditions characterized by bone fragility.
Collapse
Affiliation(s)
- Mohd P. Khan
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Elena Sabini
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Katherine Beigel
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Giulia Lanzolla
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Brittany Laslow
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dian Wang
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christophe Merceron
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Amato Giaccia
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Fanxin Long
- Department of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Deanne Taylor
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
39
|
Pi HJ, Huang B, Yuan Q, Jing JJ. Neural regulation of mesenchymal stem cells in craniofacial bone: development, homeostasis and repair. Front Physiol 2024; 15:1423539. [PMID: 39135707 PMCID: PMC11318092 DOI: 10.3389/fphys.2024.1423539] [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: 04/26/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Mesenchymal stem cells endow various functions, including proliferation, multipotency, migration, etc. Craniofacial bones originate from the cranial neural crest and are developed mainly through intramembranous ossification, which are different from long bones. There are varied mesenchymal stem cells existing in the craniofacial bone, including Gli1 + cells, Axin2 + cells, Prx1 + cells, etc. Nerves distributed in craniofacial area are also derived from the neural crest, and the trigeminal nerve is the major sensory nerve in craniofacial area. The nerves and the skeleton are tightly linked spatially, and the skeleton is broadly innervated by sensory and sympathetic nerves, which also participate in bone development, homeostasis and healing process. In this review, we summarize mesenchymal stem cells located in craniofacial bone or, to be more specific, in jaws, temporomandibular joint and cranial sutures. Then we discuss the research advance concerning neural regulation of mesenchymal stem cells in craniofacial bone, mainly focused on development, homeostasis and repair. Discovery of neural regulation of mesenchymal stem cells may assist in treatment in the craniofacial bone diseases or injuries.
Collapse
Affiliation(s)
| | | | - Quan Yuan
- *Correspondence: Quan Yuan, ; Jun-Jun Jing,
| | | |
Collapse
|
40
|
Perrin S, Ethel M, Bretegnier V, Goachet C, Wotawa CA, Luka M, Coulpier F, Masson C, Ménager M, Colnot C. Single nuclei transcriptomics reveal the differentiation trajectories of periosteal skeletal/stem progenitor cells in bone regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.23.546220. [PMID: 39211065 PMCID: PMC11361009 DOI: 10.1101/2023.06.23.546220] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Bone regeneration is mediated by skeletal stem/progenitor cells (SSPCs) that are mainly recruited from the periosteum after bone injury. The composition of the periosteum and the steps of SSPC activation and differentiation remain poorly understood. Here, we generated a single-nuclei atlas of the periosteum at steady-state and of the fracture site during early stages of bone repair ( https://fracture-repair-atlas.cells.ucsc.edu ). We identified periosteal SSPCs expressing stemness markers ( Pi16 and Ly6a /SCA1) and responding to fracture by adopting an injury-induced fibrogenic cell (IIFC) fate, prior to undergoing osteogenesis or chondrogenesis. We identified distinct gene cores associated with IIFCs and their engagement into osteogenesis and chondrogenesis involving Notch, Wnt and the circadian clock signaling respectively. Finally, we show that IIFCs are the main source of paracrine signals in the fracture environment, suggesting a crucial paracrine role of this transient IIFC population during fracture healing. Overall, our study provides a complete temporal topography of the early stages of fracture healing and the dynamic response of periosteal SSPCs to injury, redefining our knowledge of bone regeneration.
Collapse
|
41
|
Marchand T, Akinnola KE, Takeishi S, Maryanovich M, Pinho S, Saint-Vanne J, Birbrair A, Lamy T, Tarte K, Frenette PS, Gritsman K. Periosteal skeletal stem cells can migrate into the bone marrow and support hematopoiesis after injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.12.523842. [PMID: 36711927 PMCID: PMC9882153 DOI: 10.1101/2023.01.12.523842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Skeletal stem cells have been isolated from various tissues, including periosteum and bone marrow, where they exhibit key functions in bone biology and hematopoiesis, respectively. The role of periosteal skeletal stem cells in bone regeneration and healing has been extensively studied, but their ability to contribute to the bone marrow stroma is still under debate. In the present study, we characterized a whole bone transplantation model that mimics the initial bone marrow necrosis and fatty infiltration seen after injury. Using this model and a lineage tracing approach, we observed the migration of periosteal skeletal stem cells into the bone marrow after transplantation. Once in the bone marrow, periosteal skeletal stem cells are phenotypically and functionally reprogrammed into bone marrow mesenchymal stem cells that express high levels of hematopoietic stem cell niche factors such as Cxcl12 and Kitl. In addition, using in-vitro and in-vivo approaches, we found that periosteal skeletal stem cells are more resistant to acute stress than bone marrow mesenchymal stem cells. These results highlight the plasticity of periosteal skeletal stem cells and their potential role in bone marrow regeneration after bone marrow injury.
Collapse
|
42
|
Trompet D, Melis S, Chagin AS, Maes C. Skeletal stem and progenitor cells in bone development and repair. J Bone Miner Res 2024; 39:633-654. [PMID: 38696703 DOI: 10.1093/jbmr/zjae069] [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: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/04/2024]
Abstract
Bone development, growth, and repair are complex processes involving various cell types and interactions, with central roles played by skeletal stem and progenitor cells. Recent research brought new insights into the skeletal precursor populations that mediate intramembranous and endochondral bone development. Later in life, many of the cellular and molecular mechanisms determining development are reactivated upon fracture, with powerful trauma-induced signaling cues triggering a variety of postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect. Interestingly, in this injury context, the current evidence suggests that the fates of both SSPCs and differentiated skeletal cells can be considerably flexible and dynamic, and that multiple cell sources can be activated to operate as functional progenitors generating chondrocytes and/or osteoblasts. The combined implementation of in vivo lineage tracing, cell surface marker-based cell selection, single-cell molecular analyses, and high-resolution in situ imaging has strongly improved our insights into the diversity and roles of developmental and reparative stem/progenitor subsets, while also unveiling the complexity of their dynamics, hierarchies, and relationships. Albeit incompletely understood at present, findings supporting lineage flexibility and possibly plasticity among sources of osteogenic cells challenge the classical dogma of a single primitive, self-renewing, multipotent stem cell driving bone tissue formation and regeneration from the apex of a hierarchical and strictly unidirectional differentiation tree. We here review the state of the field and the newest discoveries in the origin, identity, and fates of skeletal progenitor cells during bone development and growth, discuss the contributions of adult SSPC populations to fracture repair, and reflect on the dynamism and relationships among skeletal precursors and differentiated cell lineages. Further research directed at unraveling the heterogeneity and capacities of SSPCs, as well as the regulatory cues determining their fate and functioning, will offer vital new options for clinical translation toward compromised fracture healing and bone regenerative medicine.
Collapse
Affiliation(s)
- Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Seppe Melis
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Andrei S Chagin
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
43
|
Yuan G, Lin X, Liu Y, Greenblatt MB, Xu R. Skeletal stem cells in bone development, homeostasis, and disease. Protein Cell 2024; 15:559-574. [PMID: 38442300 PMCID: PMC11259547 DOI: 10.1093/procel/pwae008] [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: 12/18/2023] [Accepted: 02/19/2024] [Indexed: 03/07/2024] Open
Abstract
Tissue-resident stem cells are essential for development and repair, and in the skeleton, this function is fulfilled by recently identified skeletal stem cells (SSCs). However, recent work has identified that SSCs are not monolithic, with long bones, craniofacial sites, and the spine being formed by distinct stem cells. Recent studies have utilized techniques such as fluorescence-activated cell sorting, lineage tracing, and single-cell sequencing to investigate the involvement of SSCs in bone development, homeostasis, and disease. These investigations have allowed researchers to map the lineage commitment trajectory of SSCs in different parts of the body and at different time points. Furthermore, recent studies have shed light on the characteristics of SSCs in both physiological and pathological conditions. This review focuses on discussing the spatiotemporal distribution of SSCs and enhancing our understanding of the diversity and plasticity of SSCs by summarizing recent discoveries.
Collapse
Affiliation(s)
- Guixin Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xixi Lin
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Ying Liu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Research Division, Hospital for Special Surgery, New York, NY 10065, USA
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen 361102, China
| |
Collapse
|
44
|
Zhao L, Xiao J, Chan CKF. New evidence for the epigenetic regulation of skeletal stem cells. Sci Bull (Beijing) 2024; 69:2004-2006. [PMID: 38664096 DOI: 10.1016/j.scib.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Affiliation(s)
- Liming Zhao
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford CA 94305, USA; Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford CA 94305, USA; Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford CA 94305, USA; Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford CA 94305, USA.
| |
Collapse
|
45
|
Ruoss S, Nasamran CA, Ball ST, Chen JL, Halter KN, Bruno KA, Whisenant TC, Parekh JN, Dorn SN, Esparza MC, Bremner SN, Fisch KM, Engler AJ, Ward SR. Comparative single-cell transcriptional and proteomic atlas of clinical-grade injectable mesenchymal source tissues. SCIENCE ADVANCES 2024; 10:eadn2831. [PMID: 38996032 PMCID: PMC11244553 DOI: 10.1126/sciadv.adn2831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/06/2024] [Indexed: 07/14/2024]
Abstract
Bone marrow aspirate concentrate (BMAC) and adipose-derived stromal vascular fraction (ADSVF) are the most marketed stem cell therapies to treat a variety of conditions in the general population and elite athletes. Both tissues have been used interchangeably clinically even though their detailed composition, heterogeneity, and mechanisms of action have neither been rigorously inventoried nor compared. This lack of information has prevented investigations into ideal dosages and has facilitated anecdata and misinformation. Here, we analyzed single-cell transcriptomes, proteomes, and flow cytometry profiles from paired clinical-grade BMAC and ADSVF. This comparative transcriptional atlas challenges the prevalent notion that there is one therapeutic cell type present in both tissues. We also provide data of surface markers that may enable isolation and investigation of cell (sub)populations. Furthermore, the proteome atlas highlights intertissue and interpatient heterogeneity of injected proteins with potentially regenerative or immunomodulatory capacities. An interactive webtool is available online.
Collapse
Affiliation(s)
- Severin Ruoss
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Chanond A. Nasamran
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
| | - Scott T. Ball
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Jeffrey L. Chen
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Kenneth N. Halter
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Kelly A. Bruno
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Thomas C. Whisenant
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
| | - Jesal N. Parekh
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Shanelle N. Dorn
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Mary C. Esparza
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | | | - Kathleen M. Fisch
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, UC San Diego, La Jolla, CA, USA
| | - Adam J. Engler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Samuel R. Ward
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Department of Radiology, UC San Diego, La Jolla, CA, USA
| |
Collapse
|
46
|
Jin M, An Y, Wang Z, Wang G, Lin Z, Ding P, Lu E, Zhao Z, Bi H. Distraction force promotes the osteogenic differentiation of Gli1 + cells in facial sutures via primary cilia-mediated Hedgehog signaling pathway. Stem Cell Res Ther 2024; 15:198. [PMID: 38971766 PMCID: PMC11227703 DOI: 10.1186/s13287-024-03811-3] [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: 10/07/2023] [Accepted: 06/21/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Trans-sutural distraction osteogenesis (TSDO) involves the application of distraction force to facial sutures to stimulate osteogenesis. Gli1+ cells in the cranial sutures play an important role in bone growth. However, whether Gli1+ cells in facial sutures differentiate into bone under distraction force is unknown. METHODS 4-week-old Gli1ER/Td and C57BL/6 mice were used to establish a TSDO model to explore osteogenesis of zygomaticomaxillary sutures. A Gli1+ cell lineage tracing model was used to observe the distribution of Gli1+ cells and explore the role of Gli1+ cells in facial bone remodeling. RESULTS Distraction force promoted bone remodeling during TSDO. Fluorescence and two-photon scanning images revealed the distribution of Gli1+ cells. Under distraction force, Gli1-lineage cells proliferated significantly and co-localized with Runx2+ cells. Hedgehog signaling was upregulated in Gli1+ cells. Inhibition of Hedgehog signaling suppresses the proliferation and osteogenesis of Gli1+ cells induced by distraction force. Subsequently, the stem cell characteristics of Gli1+ cells were identified. Cell-stretching experiments verified that mechanical force promoted the osteogenic differentiation of Gli1+ cells through Hh signaling. Furthermore, immunofluorescence staining and RT-qPCR experiments demonstrated that the primary cilia in Gli1+ cells exhibit Hedgehog-independent mechanosensitivity, which was required for the osteogenic differentiation induced by mechanical force. CONCLUSIONS Our study indicates that the primary cilia of Gli1+ cells sense mechanical stimuli, mediate Hedgehog signaling activation, and promote the osteogenic differentiation of Gli1+ cells in zygomaticomaxillary sutures.
Collapse
Affiliation(s)
- Mengying Jin
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
- Department of Plastic and Cosmetic Surgery, Henan Provincial People's Hospital, Henan, China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Zheng Wang
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Guanhuier Wang
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Zhiyu Lin
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Pengbing Ding
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Enhang Lu
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Zhenmin Zhao
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
| | - Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
47
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [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: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
48
|
Xu R, Sheng R, Lin W, Jiang S, Zhang D, Liu L, Lei K, Li X, Liu Z, Zhang X, Wang Y, Seriwatanachai D, Zhou X, Yuan Q. METTL3 Modulates Ctsk + Lineage Supporting Cranial Osteogenesis via Hedgehog. J Dent Res 2024; 103:734-744. [PMID: 38752256 DOI: 10.1177/00220345241245033] [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] [Indexed: 06/21/2024] Open
Abstract
N6-methyladenosine (m6A) modification, a eukaryotic messenger RNA modification catalyzed by methyltransferase-like 3 (METTL3), plays a pivotal role in stem cell fate determination. Calvarial bone development and maintenance are orchestrated by the cranial sutures. Cathepsin K (CTSK)-positive calvarial stem cells (CSCs) contribute to mice calvarial ossification. However, the role of m6A modification in regulating Ctsk+ lineage cells during calvarial development remains elusive. Here, we showed that METTL3 was colocalized with cranial nonosteoclastic Ctsk+ lineage cells, which were also associated with GLI1 expression. During neonatal development, depletion of Mettl3 in the Ctsk+ lineage cells delayed suture formation and decreased mineralization. During adulthood maintenance, loss of Mettl3 in the Ctsk+ lineage cells impaired calvarial bone formation, which was featured by the increased bone porosity, enhanced bone marrow cavity, and decreased number of osteocytes with the less-developed cellular outline. The analysis of methylated RNA immunoprecipitation sequencing and RNA sequencing data indicated that loss of METTL3 reduced Hedgehog (Hh) signaling pathway. Restoration of Hh signaling pathway by crossing Sufufl/+ alleles or by local administration of SAG21 partially rescued the abnormity. Our data indicate that METTL3 modulates Ctsk+ lineage cells supporting calvarial bone formation by regulating the Hh signaling pathway, providing new insights for clinical treatment of skull vault osseous diseases.
Collapse
Affiliation(s)
- R Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - R Sheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - W Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - S Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - D Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - L Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - K Lei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - X Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Z Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - X Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - D Seriwatanachai
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - X Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Q Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
49
|
Liu Y, Lin R, Fang H, Li L, Zhang M, Lu L, Gao X, Song J, Wei J, Xiao Q, Zhang F, Wu K, Cui L. Sargassum polysaccharide attenuates osteoarthritis in rats and is associated with the up-regulation of the ITGβ1-PI3K-AKT signaling pathway. J Orthop Translat 2024; 47:176-190. [PMID: 39040490 PMCID: PMC11260896 DOI: 10.1016/j.jot.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Background Osteoarthritis (OA) presents a formidable challenge, characterized by as-yet-unclear mechanical intricacies within cartilage and the dysregulation of bone homeostasis. Our preliminary data revealed the encouraging potential of a Sargassum polysaccharide (SP), in promoting chondrogenesis. The aim of our study is to comprehensively assess the therapeutic effects of SP on OA models and further elucidate its potential mechanism. Methods The protective effects of SP were initially evaluated in an inflammation-induced human chondrocyte (C28) cell model. CCK-8 assays, Alcian blue staining, RT-qPCR and Western blotting were used to verify the chondrogenesis of SP in vitro. To assess the efficacy of SP in vivo, surgically induced medial meniscus destabilization (DMM) OA rats underwent an 8-week SP treatment. The therapeutic effects of SP in OA rats were comprehensively evaluated using X-ray imaging, micro-computed tomography (μ-CT), histopathological analysis, as well as immunohistochemical and immunofluorescent staining. Following these assessments, we delved into the potential signaling pathways of SP in inflammatory chondrocytes utilizing RNA-seq analysis. Validation of these findings was conducted through RT-qPCR and western blotting techniques. Results SP significantly enhance the viability of C28 chondrocytes, and increased the secretion of acidic glycoproteins. Moreover, SP stimulated the expression of chondrogenic genes (Aggrecan, Sox9, Col2a1) and facilitated the synthesis of Collagen II protein in C28 inflammatory chondrocytes. In vivo experiments revealed that SP markedly ameliorated knee joint stenosis, alleviated bone and cartilage injuries, and reduced the histopathological scores in the OA rats. μ-CT analysis confirmed that SP lessened bone impairments in the medial femoral condyle and the subchondral bone of the tibial plateau, significantly improving the microarchitectural parameters of the subchondral bone. Histopathological analyses indicated that SP notably enhanced cartilage quality on the surface of the tibial plateau, leading to increased cartilage thickness and area. Immunohistochemistry staining and immunofluorescence staining corroborated these findings by showing a significant promotion of Collagen II expression in OA joints treated with SP. RNA-seq analysis suggest that SP's effects were mediated through the regulation of the ITGβ1-PI3K-AKT signaling axis, thereby stimulating chondrogenesis. Verification through RT-qPCR and Western blot analyses confirmed that SP significantly upregulated the expression of ITGβ1, p110δ, AKT1, ACAN, and Col2a1. Notably, knock-down of ITGβ1 using siRNA in C28 chondrocytes inhibited the expression of ITGβ1, p110δ, AKT1, and ACAN. However, these inhibitory effects were not completely reversed by supplemental SP intervention. Conclusions In summary, our findings reveal that SP significantly enhances chondrogenesis both in vitro and in vivo, alleviating OA progression both in bone and cartilage. The observed beneficial effects are intricately linked to the activation of the ITGβ1-PI3K-AKT signaling axis. The translational potential of this article Our research marks the first instance unveiling the advantageous effects and underlying mechanisms of SP in OA treatment. With its clinical prospects, SP presents compelling new evidence for the advancement of a next-generation polysaccharide drug for OA therapy.
Collapse
Affiliation(s)
- Yanzhi Liu
- Corresponding author. Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524045, China.
| | | | | | - Lixian Li
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Min Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Lujiao Lu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiang Gao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Jintong Song
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Jinsong Wei
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Qixian Xiao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Fucheng Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Kefeng Wu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
50
|
Yang K, Lei S, Qin X, Mai X, Xie W, Yang S, Wang J. Biodegradable polyvinyl alcohol/nano-hydroxyapatite composite membrane enhanced by MXene nanosheets for guided bone regeneration. J Mech Behav Biomed Mater 2024; 155:106540. [PMID: 38615407 DOI: 10.1016/j.jmbbm.2024.106540] [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: 12/26/2023] [Revised: 03/21/2024] [Accepted: 04/07/2024] [Indexed: 04/16/2024]
Abstract
MXene, as a new category of two-dimensional nanomaterials, exhibits a promising prospect in biomedical applications due to its ultrathin structure and morphology, as well as a range of remarkable properties such as biological, chemical, electronic, and optical properties. In this work, different concentrations of MXene (M) were added to polyvinyl alcohol (PVA, P)/nano-hydroxyapatite (n-HA, H) mixed solution, and series of PVA/n-HA/MXene (PHM) composite membranes were obtained by combining sol-gel and freeze-drying processes. Morphology, chemical composition, surface, and mechanical properties of the prepared PHM membranes were characterized by various techniques. Subsequently, the swelling and degradation performances of the composite membranes were tested by swelling and degradation tests. In addition, in vitro studies like cell adhesion, cytotoxicity, proliferation, osteogenic differentiation, and antibacterial properties of MC3T3-E1 were also evaluated. The results showed that the addition of MXene could apparently improve the composite membranes' physicochemical properties, bioactivity, and osteogenic differentiation. Specially, PHM membrane had the best comprehensive properties when the concentration of MXene was set as 2.0% w/v. In a word, the addition of MXene has a positive effect on improving the mechanical properties, osteogenic induction, and antibacterial properties of PH composite membranes, and the prepared PHM composite membranes possess potential applications for guided bone regeneration.
Collapse
Affiliation(s)
- Kefan Yang
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Siqi Lei
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Xiaoli Qin
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Xiaoxue Mai
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China; State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Weibo Xie
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; Lanzhou University Second Hospital, Lanzhou, 730030, China.
| | - Shengrong Yang
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinqing Wang
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|