1
|
Houghtaling S, Gombart SK, Ho TH, Huang G, Beier DR. A conditional smoothened (smo) allele on an inbred C57BL/6J genetic background has a hypomorphic smo mutant phenotype. Dev Biol 2025; 518:71-76. [PMID: 39603584 PMCID: PMC11728190 DOI: 10.1016/j.ydbio.2024.11.010] [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: 09/05/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024]
Abstract
We have introduced the floxed allele of Smoothened (Smo) carried by the mouse line Smotm2Amc into the C57BL/6J strain by serial backcross. Recapitulation of the Smo null phenotype was confirmed by deleting the allele using E2a-cre and intercrossing heterozygous Smo ± mice. No homozygous mutant embryos were identified at E9.5, suggesting the null phenotype is at least as severe as that observed on a mixed genetic background. While healthy and fertile homozygous floxed mice were regularly obtained after intercrosses, their numbers at weaning were reduced relative to Mendelian expectation, suggesting the unrecombined allele is itself hypomorphic. This hypothesis is supported by characterization of transcription of the floxed allele, which revealed that its expression was variably reduced relative to wild-type Smo.
Collapse
Affiliation(s)
- Scott Houghtaling
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, USA
| | - Sean K Gombart
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, USA
| | - Tzu-Hua Ho
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, USA
| | - Grace Huang
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, USA
| | - David R Beier
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, USA; Department of Pediatrics, Division of Genetic Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
2
|
Song C, Sawall JK, Ji X, Song F, Liao X, Peng R, Ren H, Koyama E, Pacifici M, Long F. Tgfβ signaling stimulates glycolysis to promote the genesis of synovial joint interzone in developing mouse embryonic limbs. SCIENCE ADVANCES 2025; 11:eadq4991. [PMID: 39772668 PMCID: PMC11708888 DOI: 10.1126/sciadv.adq4991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/04/2024] [Indexed: 01/30/2025]
Abstract
The initial interzone cells for synovial joints originate from chondrocytes, but such critical transition is minimally understood. With single-cell RNA sequencing (scRNA-seq) of murine embryonic knee joint primordia, we discovered that heightened expression of glycolysis genes characterized developing interzone cells when compared to flanking chondrocytes. Conditional deletion of the glucose transporters Glut1 and/or Glut3, in either the incipient pre-skeletal mesenchyme with Prx1Cre or in chondrocytes with Col2Cre, disrupted interzone formation dose-dependently. In contrast, deletion of Glut1/3 in established interzone cells with Gdf5Cre did not have similar severe disruption of joint development. scRNA-seq revealed that Glut1/3 deletion by Prx1Cre impeded Tgfβ signaling in the developing interzone cells. Direct elimination of Tgfβ signaling with Prx1Cre partially phenocopied the deletion of Glut1/3 in impairing interzone formation. Tgfβ stimulated glycolysis in chondrocytes via activation of mTOR and Hif1α in vitro. The data support that the essential conversion of chondrocytes to interzone cells requires a transient elevation of glycolysis partly dependent on Tgfβ signaling.
Collapse
Affiliation(s)
- Chao Song
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Orthopedic Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jasmin Koehnken Sawall
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xing Ji
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fangfang Song
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xueyang Liao
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Renpeng Peng
- Department of Orthopedic Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Ren
- Department of Orthopedic Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Deaprtment of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Deaprtment of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Deaprtment of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
3
|
Zhang C, Zhao R, Dong Z, Liu Y, Liu M, Li H, Yin Y, Che X, Wu G, li Guo, Li P, Wei X, Yang Z. IHH-GLI-1-HIF-2α signalling influences hypertrophic chondrocytes to exacerbate osteoarthritis progression. J Orthop Translat 2024; 49:207-217. [PMID: 39498143 PMCID: PMC11532729 DOI: 10.1016/j.jot.2024.09.008] [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: 06/16/2024] [Revised: 09/09/2024] [Accepted: 09/25/2024] [Indexed: 11/07/2024] Open
Abstract
Background Chondrocyte hypertrophy is a potential target for osteoarthritis (OA) treatment, with Indian hedgehog (IHH), glioma-associated oncogene homolog (GLI), and hypoxia-inducible factor-2α (HIF-2α) being closely associated with chondrocyte hypertrophy during OA progression. Whereas IHH can modulate chondrocyte hypertrophy, interference with IHH signalling has not achieved the anticipated therapeutic effects and poses safety concerns, necessitating further clarification of the specific mechanisms by which IHH affects articular cartilage degeneration. Inhibition of the HIF-2α overexpression in cartilage slows the progression of early OA, but the mechanisms underlying HIF-2α accumulation in OA cartilage remain unclear. The aim of this study was to determine the function of Ihh, as well as its downstream factors, in chondrocytes, based on an early osteoarthritis (OA) mouse model and in vitro chondrocyte model. Methods Investigated the expression levels and locations of IHH-GLI-1 pathway in normal and early degenerated human cartilage, comparing them with HIF-2α and its downstream factors. RT-qPCR, Western blotting, Crystal violet staining, and EdU assays were used to evaluate the pecific regulatory mechanisms of the IHH-GLI-1-HIF-2α signalling axis in normal chondrocytes and in chondrocytes under inflammatory conditions. Validated the impact of IHH on early cartilage degeneration and the relationship between the IHH-GLI-1 pathway and the expression levels and expression locations of HIF-2α and its downstream factors in Col2a1-CreERT2;Ihhfl/fl mice. Results In early-stage degenerative joint cartilage, the GLI-1 pathway in hypertrophic chondrocytes exhibited similar changes in location and levels to HIF-2α and its downstream factor vascular endothelial growth factor (VEGF). In vitro, IHH-GLI-1-HIF-2α signalling activation in chondrocytes under physiological hypoxic conditions inhibited chondrocyte proliferation. In chondrocytes stimulated by inflammatory environments, IHH inhibited the degradation of HIF-2α via the GLI-1 pathway, thereby promoting HIF-2α protein expression. Elevated HIF-2α expression further enhanced intracellular IHH-GLI-1 levels, generating a positive feedback loop to collectively regulate the expression of downstream hypertrophic factors and matrix-degradation factors. In vivo, conditional Ihh knockout in mouse chondrocytes downregulated Hif-2α protein expression in early degenerative cartilage tissue and affected the expression of downstream Vegf and hypertrophic factors. Conclusions During OA progression, the IHH-GLI-1-HIF-2α axis mainly operates within hypertrophic chondrocytes, exacerbating cartilage degeneration by regulating hypertrophic chondrocyte functions, cartilage matrix degradation, and microvascular invasion. The translational potential of this article This study identifies the IHH-GLI-1-HIF-2α signalling axis and reveals its potential as a therapeutic target for OA.
Collapse
Affiliation(s)
- Chengming Zhang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Ruipeng Zhao
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Zhengquan Dong
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yang Liu
- Department of Laboratory Medicine, Handan Second Hospital, Hebei University of Engineering, Handan, 056000, PR China
| | - Mengrou Liu
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Haoqian Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yukun Yin
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xianda Che
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Gaige Wu
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - li Guo
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Pengcui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Ziquan Yang
- Department of Orthopedics, First Hospital of Shanxi Medical University, Taiyuan, 030000, PR China
| |
Collapse
|
4
|
Puviindran V, Shimada E, Huang Z, Ma X, Ban GI, Xiang Y, Zhang H, Ou J, Wei X, Nakagawa M, Martin J, Diao Y, Alman BA. Single cell analysis of Idh mutant growth plates identifies cell populations responsible for longitudinal bone growth and enchondroma formation. Sci Rep 2024; 14:26208. [PMID: 39482341 PMCID: PMC11527983 DOI: 10.1038/s41598-024-76539-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024] Open
Abstract
Enchondromas are a common tumor in bone that can occur as multiple lesions in enchondromatosis, which is associated with deformity of the affected bone. These lesions harbor somatic mutations in IDH and driving expression of a mutant Idh1 in Col2 expressing cells in mice causes an enchondromatosis phenotype. Here we compared growth plates from E18.5 mice expressing a mutant Idh1 with control littermates using single cell RNA sequencing. Data from Col2 expressing cells were analysed using UMAP and RNA pseudo-time analyses. A unique cluster of cells was identified in the mutant growth plates that expressed genes known to be upregulated in enchondromas. There was also a cluster of cells that was underrepresented in the mutant growth plates that expressed genes known to be important in longitudinal bone growth. Immunofluorescence showed that the genes from the unique cluster identified in the mutant growth plates were expressed in multiple growth plate anatomic zones, and pseudo-time analysis also suggested these cells could arise from multiple growth plate chondrocyte subpopulations. This data supports the notion that a subpopulation of chondrocytes become enchondromas at the expense of contributing to longitudinal growth.
Collapse
Affiliation(s)
- Vijitha Puviindran
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Eijiro Shimada
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Zeyu Huang
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Xinyi Ma
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
- Developmental and Stem Cell Biology Program, Duke University School of Medicine, Durham, NC, USA
| | - Ga I Ban
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Yu Xiang
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
- Regeneration Center, Duke University School of Medicine, Durham, NC, USA
| | - Hongyuan Zhang
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jianhong Ou
- Regeneration Center, Duke University School of Medicine, Durham, NC, USA
| | - Xiaolin Wei
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
- Regeneration Center, Duke University School of Medicine, Durham, NC, USA
| | - Makoto Nakagawa
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - John Martin
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Yarui Diao
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
- Regeneration Center, Duke University School of Medicine, Durham, NC, USA
| | - Benjamin A Alman
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA.
- Regeneration Center, Duke University School of Medicine, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
- Developmental and Stem Cell Biology Program, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
5
|
Bian F, Hansen V, Feng HC, He J, Chen Y, Feng K, Ebrahimi B, Gray RS, Chai Y, Wu CL, Liu Z. The G protein-coupled receptor ADGRG6 maintains mouse growth plate homeostasis through IHH signaling. J Bone Miner Res 2024; 39:1644-1658. [PMID: 39236220 PMCID: PMC11523133 DOI: 10.1093/jbmr/zjae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/31/2024] [Accepted: 09/04/2024] [Indexed: 09/07/2024]
Abstract
The cartilage growth plate is essential for maintaining skeletal growth; however, the mechanisms governing postnatal growth plate homeostasis are still poorly understood. Using approaches of molecular mouse genetics and spatial transcriptomics applied to formalin-fixed, paraffin-embedded tissues, we show that ADGRG6/GPR126, a cartilage-enriched adhesion G protein-coupled receptor (GPCR), is essential for maintaining slow-cycling resting zone cells, appropriate chondrocyte proliferation and differentiation, and growth plate homeostasis in mice. Constitutive ablation of Adgrg6 in osteochondral progenitor cells with Col2a1Cre leads to a shortened resting zone, formation of cell clusters within the proliferative zone, and an elongated hypertrophic growth plate, marked by limited expression of parathyroid hormone-related protein (PTHrP) but increased Indian Hedgehog (IHH) signaling throughout the growth plate. Attenuation of smoothened-dependent hedgehog signaling restored the Adgrg6 deficiency-induced expansion of hypertrophic chondrocytes, confirming that IHH signaling can promote chondrocyte hypertrophy in a PTHrP-independent manner. In contrast, postnatal ablation of Adgrg6 in mature chondrocytes with AcanCreERT2, induced after the formation of the resting zone, does not affect PTHrP expression but causes an overall reduction of growth plate thickness marked by increased cell death specifically in the resting zone cells and a general reduction of chondrocyte proliferation and differentiation. Spatial transcriptomics reveals that ADGRG6 is essential for maintaining chondrocyte homeostasis by regulating osteogenic and catabolic genes in all the zones of the postnatal growth plates, potentially through positive regulation of SOX9 expression. Our findings elucidate the essential role of a cartilage-enriched adhesion GPCR in regulating cell proliferation and hypertrophic differentiation by regulation of PTHrP/IHH signaling, maintenance of slow-cycle resting zone chondrocytes, and safeguarding chondrocyte homeostasis in postnatal mouse growth plates.
Collapse
Affiliation(s)
- Fangzhou Bian
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Victoria Hansen
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - Hong Colleen Feng
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Jingyu He
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Yanshi Chen
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, United States
- Department of Biology, University of Rochester, Rochester, NY 14642, United States
| | - Kaining Feng
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Brenda Ebrahimi
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Ryan S Gray
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78723, United States
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
| | - Chia-Lung Wu
- Center for Musculoskeletal Research, Department of Orthopedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY 14642, United States
| | - Zhaoyang Liu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, United States
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| |
Collapse
|
6
|
Wang L, Guo Q, Acharya S, Zheng X, Huynh V, Whitmore B, Yimit A, Malhotra M, Chatterji S, Rosin N, Labit E, Chipak C, Gorzo K, Haidey J, Elliott DA, Ram T, Zhang Q, Kuipers H, Gordon G, Biernaskie J, Guo J. Primary cilia signaling in astrocytes mediates development and regional-specific functional specification. Nat Neurosci 2024; 27:1708-1720. [PMID: 39103557 DOI: 10.1038/s41593-024-01726-z] [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: 07/16/2023] [Accepted: 07/09/2024] [Indexed: 08/07/2024]
Abstract
Astrocyte diversity is greatly influenced by local environmental modulation. Here we report that the majority of astrocytes across the mouse brain possess a singular primary cilium localized to the cell soma. Comparative single-cell transcriptomics reveals that primary cilia mediate canonical SHH signaling to modulate astrocyte subtype-specific core features in synaptic regulation, intracellular transport, energy and metabolism. Independent of canonical SHH signaling, primary cilia are important regulators of astrocyte morphology and intracellular signaling balance. Dendritic spine analysis and transcriptomics reveal that perturbation of astrocytic cilia leads to disruption of neuronal development and global intercellular connectomes in the brain. Mice with primary ciliary-deficient astrocytes show behavioral deficits in sensorimotor function, sociability, learning and memory. Our results uncover a critical role for primary cilia in transmitting local cues that drive the region-specific diversification of astrocytes within the developing brain.
Collapse
Affiliation(s)
- Lizheng Wang
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Qianqian Guo
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sandesh Acharya
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Xiao Zheng
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vanessa Huynh
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Brandon Whitmore
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Askar Yimit
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mehr Malhotra
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Siddharth Chatterji
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicole Rosin
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Elodie Labit
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Colten Chipak
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kelsea Gorzo
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Jordan Haidey
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - David A Elliott
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tina Ram
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Qingrun Zhang
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Mathematics and Statistics, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Hedwich Kuipers
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Grant Gordon
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jiami Guo
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
7
|
Nocera S, Marchena MA, Fernández-Gómez B, Gómez-Martín P, Sánchez-Jiménez E, Macías-Castellano A, Laó Y, Cordano C, Gómez-Torres Ó, Luján R, de Castro F. Activation of Shh/Smo is sufficient to maintain oligodendrocyte precursor cells in an undifferentiated state and is not necessary for myelin formation and (re)myelination. Glia 2024; 72:1469-1483. [PMID: 38771121 DOI: 10.1002/glia.24540] [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: 07/07/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/22/2024]
Abstract
Myelination is the terminal step in a complex and precisely timed program that orchestrates the proliferation, migration and differentiation of oligodendroglial cells. It is thought that Sonic Hedgehog (Shh) acting on Smoothened (Smo) participates in regulating this process, but that these effects are highly context dependent. Here, we investigate oligodendroglial development and remyelination from three specific transgenic lines: NG2-CreERT2 (control), Smofl/fl/NG2-CreERT2 (loss of function), and SmoM2/NG2-CreERT2 (gain of function), as well as pharmacological manipulation that enhance or inhibit the Smo pathway (Smoothened Agonist (SAG) or cyclopamine treatment, respectively). To explore the effects of Shh/Smo on differentiation and myelination in vivo, we developed a highly quantifiable model by transplanting oligodendrocyte precursor cells (OPCs) in the retina. We find that myelination is greatly enhanced upon cyclopamine treatment and hypothesize that Shh/Smo could promote OPC proliferation to subsequently inhibit differentiation. Consistent with this hypothesis, we find that the genetic activation of Smo significantly increased numbers of OPCs and decreased oligodendrocyte differentiation when we examined the corpus callosum during development and after cuprizone demyelination and remyelination. However, upon loss of function with the conditional ablation of Smo, myelination in the same scenarios are unchanged. Taken together, our present findings suggest that the Shh pathway is sufficient to maintain OPCs in an undifferentiated state, but is not necessary for myelination and remyelination.
Collapse
Affiliation(s)
- Sonia Nocera
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
| | - Miguel A Marchena
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
- Facultad HM de Ciencias de la Salud de la UCJC, Universidad Camilo José Cela, Madrid, Spain
- NeuroLab, Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | | | - Paula Gómez-Martín
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
| | | | | | - Yolanda Laó
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
| | - Christian Cordano
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, California, USA
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health. University of Genoa, Italy
- Department of Neuroscience, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Óscar Gómez-Torres
- Facultad de Ciencias Ambientales, Universidad de Castilla-La Mancha, Toledo, Spain
| | - Rafael Luján
- Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Madrid, Spain
| |
Collapse
|
8
|
Yoshida S, Kawamura A, Aoki K, Wiriyasermkul P, Sugimoto S, Tomiyoshi J, Tajima A, Ishida Y, Katoh Y, Tsukada T, Tsuneoka Y, Yamada K, Nagamori S, Nakayama K, Yoshida K. Positive regulation of Hedgehog signaling via phosphorylation of GLI2/GLI3 by DYRK2 kinase. Proc Natl Acad Sci U S A 2024; 121:e2320070121. [PMID: 38968120 PMCID: PMC11252808 DOI: 10.1073/pnas.2320070121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 06/02/2024] [Indexed: 07/07/2024] Open
Abstract
Hedgehog (Hh) signaling, an evolutionarily conserved pathway, plays an essential role in development and tumorigenesis, making it a promising drug target. Multiple negative regulators are known to govern Hh signaling; however, how activated Smoothened (SMO) participates in the activation of downstream GLI2 and GLI3 remains unclear. Herein, we identified the ciliary kinase DYRK2 as a positive regulator of the GLI2 and GLI3 transcription factors for Hh signaling. Transcriptome and interactome analyses demonstrated that DYRK2 phosphorylates GLI2 and GLI3 on evolutionarily conserved serine residues at the ciliary base, in response to activation of the Hh pathway. This phosphorylation induces the dissociation of GLI2/GLI3 from suppressor, SUFU, and their translocation into the nucleus. Loss of Dyrk2 in mice causes skeletal malformation, but neural tube development remains normal. Notably, DYRK2-mediated phosphorylation orchestrates limb development by controlling cell proliferation. Taken together, the ciliary kinase DYRK2 governs the activation of Hh signaling through the regulation of two processes: phosphorylation of GLI2 and GLI3 downstream of SMO and cilia formation. Thus, our findings of a unique regulatory mechanism of Hh signaling expand understanding of the control of Hh-associated diseases.
Collapse
Affiliation(s)
- Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Akira Kawamura
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Katsuhiko Aoki
- Radioisotope Research Facilities, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Pattama Wiriyasermkul
- Center for Stable Isotope Medical Research, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Shinya Sugimoto
- Department of Bacteriology, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Center for Biofilm Science and Technology, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Laboratory of Amyloid Regulation, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Junnosuke Tomiyoshi
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Ayasa Tajima
- Center for Stable Isotope Medical Research, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Department of Molecular Biology, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Yamato Ishida
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto606-8501, Japan
| | - Yohei Katoh
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto606-8501, Japan
| | - Takehiro Tsukada
- Department of Biomolecular Science, Toho University, Chiba274-8510, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo143-8540, Japan
| | - Kohji Yamada
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Shushi Nagamori
- Center for Stable Isotope Medical Research, The Jikei University School of Medicine, Tokyo105-8461, Japan
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo105-8461, Japan
| | - Kazuhisa Nakayama
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto606-8501, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo105-8461, Japan
| |
Collapse
|
9
|
Díaz-González F, Sentchordi-Montané L, Lucas-Castro E, Modamio-Høybjør S, Heath KE. Variants in both the N- or C-terminal domains of IHH lead to defective secretion causing short stature and skeletal defects. Eur J Endocrinol 2024; 191:38-46. [PMID: 38917024 DOI: 10.1093/ejendo/lvae072] [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: 04/02/2024] [Revised: 05/08/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Heterozygous Indian Hedgehog gene (IHH) variants are associated with brachydactyly type A1 (BDA1). However, in recent years, numerous variants have been identified in patients with short stature and more variable forms of brachydactyly. Many are located in the C-terminal domain of IHH (IHH-C), which lacks signaling activity but is critical for auto-cleavage and activation of the N-terminal (IHH-N) peptide. The absence of functional studies of IHH variants, particularly for those located in IHH-C, has led to these variants being classified as variants of uncertain significance (VUS). OBJECTIVE To establish a simple functional assay to determine the pathogenicity of IHH VUS and confirm that variants in the C-terminal domain affect protein function. DESIGN/METHODS In vitro studies were performed for 9 IHH heterozygous variants, to test their effect on secretion and IHH intracellular processing by western blot of cells expressing each variant. RESULTS IHH secretion was significantly reduced in all mutants, regardless of the location. Similarly, intracellular levels of N-terminal and C-terminal IHH peptides were severely reduced in comparison with the control. Two variants present at a relatively high frequency in the general population also reduced secretion but to a lesser degree in the heterozygous state. CONCLUSIONS These studies provide the first evidence that variants in the C-terminal domain affect the secretion capacity of IHH and thus, reduce availability of IHH ligand, resulting in short stature and mild skeletal defects. The secretion assay permits a relatively easy test to determine the pathogenicity of IHH variants. All studied variants affected secretion and interestingly, more frequent population variants appear to have a deleterious effect and thus contribute to height variation.
Collapse
Affiliation(s)
- Francisca Díaz-González
- Institute of Medical and Molecular Genetics (INGEMM), IdiPAZ, Hospital Universitario La Paz, UAM, 28046 Madrid, Spain
- Skeletal Dysplasia Multidisciplinary Unit (UMDE-ERN BOND), Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Lucía Sentchordi-Montané
- Institute of Medical and Molecular Genetics (INGEMM), IdiPAZ, Hospital Universitario La Paz, UAM, 28046 Madrid, Spain
- Skeletal Dysplasia Multidisciplinary Unit (UMDE-ERN BOND), Hospital Universitario La Paz, 28046 Madrid, Spain
- Department of Pediatrics, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
- Department of Pediatrics, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Elsa Lucas-Castro
- Institute of Medical and Molecular Genetics (INGEMM), IdiPAZ, Hospital Universitario La Paz, UAM, 28046 Madrid, Spain
- Skeletal Dysplasia Multidisciplinary Unit (UMDE-ERN BOND), Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Silvia Modamio-Høybjør
- Institute of Medical and Molecular Genetics (INGEMM), IdiPAZ, Hospital Universitario La Paz, UAM, 28046 Madrid, Spain
- Skeletal Dysplasia Multidisciplinary Unit (UMDE-ERN BOND), Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Karen E Heath
- Institute of Medical and Molecular Genetics (INGEMM), IdiPAZ, Hospital Universitario La Paz, UAM, 28046 Madrid, Spain
- Skeletal Dysplasia Multidisciplinary Unit (UMDE-ERN BOND), Hospital Universitario La Paz, 28046 Madrid, Spain
- CIBERER, ISCIII, 28029 Madrid, Spain
| |
Collapse
|
10
|
Li D, Cheng K, Zhu X. Construction and Identification of a Novel Mice Model of Microphthalmia. Transl Vis Sci Technol 2024; 13:11. [PMID: 39007834 PMCID: PMC467107 DOI: 10.1167/tvst.13.7.11] [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: 11/09/2023] [Accepted: 05/30/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose Microphthalmia is a rare developmental eye disease that affects 1 in 7000 births. Currently, there is no cure for this condition. This study aimed to construct a stable mouse model of microphthalmia, thus providing a new tool for the study of the etiology of microphthalmia. Methods The Hedgehog signaling pathway plays a crucial role in eye development. One of the key mechanisms of the Sonic Hedgehog signaling is the strong transcriptional activation ability of GLI3, a major mediator of this pathway. This study used CRISPR/Cas9 system to construct a novel TgGli3Ki/Ki lens-specific over-expression mouse line. To identify the ocular characteristics of this line, quantitative PCR, Western blot, hematoxylin and eosin staining, immunofluorescent staining, and RNA-seq were performed on the ocular tissues of this line and normal mice. Results The TgGli3Ki/Ki lens-specific over-expression mouse model exhibits the ocular phenotype of microphthalmia. In the TgGli3Ki/Ki mouse, Gli3 is over-expressed in the lens, and the size of the eyeball and lens is significantly smaller than the normal one. RNA-seq analysis using the lens and the retina samples from TgGli3Ki/Ki and normal mice indicates that the phototransduction pathway is ectopically activated in the lens. Immunofluorescent staining of the lens samples confirmed this activation. Conclusions The TgGli3Ki/Ki mouse model consistently manifests the stereotypical microphthalmia phenotype across generations, making it an excellent tool for studying this severe eye disease. Translational Relevance This study developed a novel animal model to facilitate clinical research on microphthalmia.
Collapse
Affiliation(s)
- Dan Li
- Eye Institute, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Kaiwen Cheng
- Eye Institute, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xiangjia Zhu
- Eye Institute, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
11
|
Puviindran V, Shimada E, Huang Z, Ma X, Ban GI, Xiang Y, Zhang H, Ou J, Wei X, Nakagawa M, Martin J, Diao Y, Alman BA. Single-cell transcriptomic analyses of mouse idh1 mutant growth plate chondrocytes reveal distinct cell populations responsible for longitudinal growth and enchondroma formation. RESEARCH SQUARE 2024:rs.3.rs-4451086. [PMID: 38883785 PMCID: PMC11178001 DOI: 10.21203/rs.3.rs-4451086/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Enchondromas are a common tumor in bone that can occur as multiple lesions in enchondromatosis, which is associated with deformity of the effected bone. These lesions harbor mutations in IDH and driving expression of a mutant Idh1 in Col2 expressing cells in mice causes an enchondromatosis phenotype. In this study we compared growth plates from E18.5 mice expressing a mutant Idh1 with control littermates using single cell RNA sequencing. Data from Col2 expressing cells were analyzed using UMAP and RNA pseudo-time analyses. A unique cluster of cells was identified in the mutant growth plates that expressed genes known to be upregulated in enchondromas. There was also a cluster of cells that was underrepresented in the mutant growth plates that expressed genes known to be important in longitudinal bone growth. Immunofluorescence showed that the genes from the unique cluster identified in the mutant growth plates were expressed in multiple growth plate anatomic zones, and pseudo-time analysis also suggested these cells could arise from multiple growth plate chondrocyte subpopulations. This data identifies subpopulations of cells in control and mutant growth plates, and supports the notion that a mutant Idh1 alters the subpopulations of growth plate chondrocytes, resulting a subpopulation of cells that become enchondromas at the expense of other populations that contribute to longitudinal growth.
Collapse
Affiliation(s)
| | | | | | - Xinyi Ma
- Duke University School of Medicine
| | - Ga I Ban
- Duke University School of Medicine
| | - Yu Xiang
- Duke University School of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Liang XG, Hoang K, Meyerink BL, Kc P, Paraiso K, Wang L, Jones IR, Zhang Y, Katzman S, Finn TS, Tsyporin J, Qu F, Chen Z, Visel A, Kriegstein A, Shen Y, Pilaz LJ, Chen B. A conserved molecular logic for neurogenesis to gliogenesis switch in the cerebral cortex. Proc Natl Acad Sci U S A 2024; 121:e2321711121. [PMID: 38713624 PMCID: PMC11098099 DOI: 10.1073/pnas.2321711121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/02/2024] [Indexed: 05/09/2024] Open
Abstract
During development, neural stem cells in the cerebral cortex, also known as radial glial cells (RGCs), generate excitatory neurons, followed by production of cortical macroglia and inhibitory neurons that migrate to the olfactory bulb (OB). Understanding the mechanisms for this lineage switch is fundamental for unraveling how proper numbers of diverse neuronal and glial cell types are controlled. We and others recently showed that Sonic Hedgehog (Shh) signaling promotes the cortical RGC lineage switch to generate cortical oligodendrocytes and OB interneurons. During this process, cortical RGCs generate intermediate progenitor cells that express critical gliogenesis genes Ascl1, Egfr, and Olig2. The increased Ascl1 expression and appearance of Egfr+ and Olig2+ cortical progenitors are concurrent with the switch from excitatory neurogenesis to gliogenesis and OB interneuron neurogenesis in the cortex. While Shh signaling promotes Olig2 expression in the developing spinal cord, the exact mechanism for this transcriptional regulation is not known. Furthermore, the transcriptional regulation of Olig2 and Egfr has not been explored. Here, we show that in cortical progenitor cells, multiple regulatory programs, including Pax6 and Gli3, prevent precocious expression of Olig2, a gene essential for production of cortical oligodendrocytes and astrocytes. We identify multiple enhancers that control Olig2 expression in cortical progenitors and show that the mechanisms for regulating Olig2 expression are conserved between the mouse and human. Our study reveals evolutionarily conserved regulatory logic controlling the lineage switch of cortical neural stem cells.
Collapse
Affiliation(s)
- Xiaoyi G. Liang
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| | - Kendy Hoang
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| | - Brandon L. Meyerink
- Division of Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD57104
- Department of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Sioux Falls, SD57105
| | - Pratiksha Kc
- Division of Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD57104
| | - Kitt Paraiso
- Environmental Genomics & System Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA94720
| | - Li Wang
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA94143
- Department of Neurology, University of California, San Francisco, CA94143
| | - Ian R. Jones
- Institute for Human Genetics, University of California, San Francisco, CA94143
| | - Yue Zhang
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| | - Sol Katzman
- Genome Institute, University of California, Santa Cruz, CA95064
| | - Thomas S. Finn
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| | - Jeremiah Tsyporin
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| | - Fangyuan Qu
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| | - Zhaoxu Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| | - Axel Visel
- Environmental Genomics & System Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA94720
- U.S. Department of Energy Joint Genome Institute, Berkeley, CA94720
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA95343
| | - Arnold Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA94143
- Department of Neurology, University of California, San Francisco, CA94143
| | - Yin Shen
- Department of Neurology, University of California, San Francisco, CA94143
- Institute for Human Genetics, University of California, San Francisco, CA94143
| | - Louis-Jan Pilaz
- Division of Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD57104
- Department of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Sioux Falls, SD57105
| | - Bin Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA95064
| |
Collapse
|
13
|
Xu M, Li Z, Liang X, Li J, Ye Y, Qi P, Yan X. Transcriptomic Analysis Provides Insights into Candidate Genes and Molecular Pathways Involved in Growth of Mytilus coruscus Larvae. Int J Mol Sci 2024; 25:1898. [PMID: 38339176 PMCID: PMC10855951 DOI: 10.3390/ijms25031898] [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: 01/04/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Growth is a fundamental aspect of aquaculture breeding programs, pivotal for successful cultivation. Understanding the mechanisms that govern growth and development differences across various stages can significantly boost seedling production of economically valuable species, thereby enhancing aquaculture efficiency and advancing the aquaculture industry. Mytilus coruscus, a commercially vital marine bivalve, underscores this importance. To decipher the intricate molecular mechanisms dictating growth and developmental disparities in marine shellfish, we conducted transcriptome sequencing and meticulously analyzed gene expression variations and molecular pathways linked to growth traits in M. coruscus. This study delved into the molecular and gene expression variations across five larval development stages, with a specific focus on scrutinizing the differential expression patterns of growth-associated genes using RNA sequencing and quantitative real-time PCR analysis. A substantial number of genes-36,044 differentially expressed genes (DEGs)-exhibited significant differential expression between consecutive developmental stages. These DEGs were then categorized into multiple pathways (Q value < 0.05), including crucial pathways such as the spliceosome, vascular smooth muscle contraction, DNA replication, and apoptosis, among others. In addition, we identified two pivotal signaling pathways-the Hedgehog (Hh) signaling pathway and the TGF-beta (TGF-β) signaling pathway-associated with the growth and development of M. coruscus larvae. Ten key growth-related genes were pinpointed, each playing crucial roles in molecular function and the regulation of growth traits in M. coruscus. These genes and pathways associated with growth provide deep insights into the molecular basis of physiological adaptation, metabolic processes, and growth variability in marine bivalves.
Collapse
Affiliation(s)
| | | | | | | | - Yingying Ye
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China; (M.X.); (Z.L.); (X.L.); (J.L.); (X.Y.)
| | - Pengzhi Qi
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China; (M.X.); (Z.L.); (X.L.); (J.L.); (X.Y.)
| | | |
Collapse
|
14
|
Zhang L, Hu S, Xiu C, Li M, Zheng Y, Zhang R, Li B, Chen J. Intervertebral disc-intrinsic Hedgehog signaling maintains disc cell phenotypes and prevents disc degeneration through both cell autonomous and non-autonomous mechanisms. Cell Mol Life Sci 2024; 81:74. [PMID: 38308696 PMCID: PMC10838248 DOI: 10.1007/s00018-023-05106-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: 11/13/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 02/05/2024]
Abstract
Intervertebral disc degeneration is closely related to abnormal phenotypic changes in disc cells. However, the mechanism by which disc cell phenotypes are maintained remains poorly understood. Here, Hedgehog-responsive cells were found to be specifically localized in the inner annulus fibrosus and cartilaginous endplate of postnatal discs, likely activated by Indian Hedgehog. Global inhibition of Hedgehog signaling using a pharmacological inhibitor or Agc1-CreERT2-mediated deletion of Smo in disc cells of juvenile mice led to spontaneous degenerative changes in annulus fibrosus and cartilaginous endplate accompanied by aberrant disc cell differentiation in adult mice. In contrast, Krt19-CreER-mediated deletion of Smo specifically in nucleus pulposus cells led to healthy discs and normal disc cell phenotypes. Similarly, age-related degeneration of nucleus pulposus was accelerated by genetic inactivation of Hedgehog signaling in all disc cells, but not in nucleus pulposus cells. Furthermore, inactivation of Gli2 in disc cells resulted in partial loss of the vertebral growth plate but otherwise healthy discs, whereas deletion of Gli3 in disc cells largely corrected disc defects caused by Smo ablation in mice. Taken together, our findings not only revealed for the first time a direct role of Hedgehog-Gli3 signaling in maintaining homeostasis and cell phenotypes of annuls fibrosus and cartilaginous endplate, but also identified disc-intrinsic Hedgehog signaling as a novel non-cell-autonomous mechanism to regulate nucleus pulposus cell phenotype and protect mice from age-dependent nucleus pulposus degeneration. Thus, targeting Hedgehog signaling may represent a potential therapeutic strategy for the prevention and treatment of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Siyuan Hu
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Chunmei Xiu
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
| | - Meng Li
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yixin Zheng
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Rui Zhang
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Bin Li
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China.
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Jianquan Chen
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China.
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
15
|
Singh S, Budiman T, Redmond D, Gupta V. Modulation of canonical Wnt signaling regulates peribiliary mesenchymal identity during homeostasis and injury. Hepatol Commun 2024; 8:e0368. [PMID: 38251878 PMCID: PMC10805418 DOI: 10.1097/hc9.0000000000000368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/10/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND The matrix and associated mesenchyme of the extrahepatic bile ducts are distinct, which could drive diseases with a predilection for these ducts, such as primary sclerosing cholangitis. We aimed to understand the molecular drivers of peribiliary mesenchymal cell (PMC) identity in the extrahepatic bile ducts and dissect how this changed in the context of injury using an entirely in vivo approach with transcriptomic analysis. METHODS AND RESULTS Single-cell sequencing with a receptor-ligand analysis showed that PMCs had the most interactions with surrounding cells. Wnt4, Wnt5a, and Wnt7b were identified as the major ligands secreted from PMCs and cholangiocytes that interacted in both paracrine and autocrine fashion. Bile duct ligation caused an increase in all 3 Wingless/Integrated ligands and Axin2 with an associated increase in the transcription factors T-box transcription factor (Tbx)2 and Tbx3. Conversely, Indian hedgehog secretion decreased without an associated decrease in hedgehog signaling effectors. Loss of smoothened within PMCs did not impact hedgehog signaling effectors or cellular identity, whereas smoothened gain of function led to myofibroblast transdifferentiation with upregulation of Tbx2 and Tbx3 without injury. Loss of β-catenin caused a decrease in expression of all 3 Gli transcription factors and associated mesenchymal gene expression, which was phenocopied with compound Gli2 and Gli3 loss in uninjured PMCs. With injury, loss of β-catenin resulted in decreased myofibroblast transdifferentiation with reduced Tbx2 and Tbx3 expression. CONCLUSIONS Our results show how modulation of canonical Wingless/Integrated signaling in PMCs is important for regulating basal mesenchymal gene expression and initiating a myogenic gene transcriptional program during injury. They also highlight reciprocating interactions between the hedgehog and Wingless/Integrated signaling pathways within PMCs.
Collapse
Affiliation(s)
- Serrena Singh
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tifanny Budiman
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - David Redmond
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Vikas Gupta
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
16
|
Yu H, Khanshour AM, Ushiki A, Otomo N, Koike Y, Einarsdottir E, Fan Y, Antunes L, Kidane YH, Cornelia R, Sheng RR, Zhang Y, Pei J, Grishin NV, Evers BM, Cheung JPY, Herring JA, Terao C, Song YQ, Gurnett CA, Gerdhem P, Ikegawa S, Rios JJ, Ahituv N, Wise CA. Association of genetic variation in COL11A1 with adolescent idiopathic scoliosis. eLife 2024; 12:RP89762. [PMID: 38277211 PMCID: PMC10945706 DOI: 10.7554/elife.89762] [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: 01/27/2024] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a common and progressive spinal deformity in children that exhibits striking sexual dimorphism, with girls at more than fivefold greater risk of severe disease compared to boys. Despite its medical impact, the molecular mechanisms that drive AIS are largely unknown. We previously defined a female-specific AIS genetic risk locus in an enhancer near the PAX1 gene. Here, we sought to define the roles of PAX1 and newly identified AIS-associated genes in the developmental mechanism of AIS. In a genetic study of 10,519 individuals with AIS and 93,238 unaffected controls, significant association was identified with a variant in COL11A1 encoding collagen (α1) XI (rs3753841; NM_080629.2_c.4004C>T; p.(Pro1335Leu); p=7.07E-11, OR = 1.118). Using CRISPR mutagenesis we generated Pax1 knockout mice (Pax1-/-). In postnatal spines we found that PAX1 and collagen (α1) XI protein both localize within the intervertebral disc-vertebral junction region encompassing the growth plate, with less collagen (α1) XI detected in Pax1-/- spines compared to wild-type. By genetic targeting we found that wild-type Col11a1 expression in costal chondrocytes suppresses expression of Pax1 and of Mmp3, encoding the matrix metalloproteinase 3 enzyme implicated in matrix remodeling. However, the latter suppression was abrogated in the presence of the AIS-associated COL11A1P1335L mutant. Further, we found that either knockdown of the estrogen receptor gene Esr2 or tamoxifen treatment significantly altered Col11a1 and Mmp3 expression in chondrocytes. We propose a new molecular model of AIS pathogenesis wherein genetic variation and estrogen signaling increase disease susceptibility by altering a PAX1-COL11a1-MMP3 signaling axis in spinal chondrocytes.
Collapse
Affiliation(s)
- Hao Yu
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Anas M Khanshour
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Aki Ushiki
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Nao Otomo
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
| | - Yoshinao Koike
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical SciencesYokohamaJapan
| | - Elisabet Einarsdottir
- Science for Life Laboratory, Department of Gene Technology, KTH-Royal Institute of TechnologySolnaSweden
| | - Yanhui Fan
- School of Biomedical Sciences, The University of Hong KongHong Kong SARChina
| | - Lilian Antunes
- Department of Neurology, Washington University in St. LouisSt. LouisUnited States
| | - Yared H Kidane
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Reuel Cornelia
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Rory R Sheng
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Yichi Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
- School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| | - Jimin Pei
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Nick V Grishin
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Ophthalmology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology LKS Faculty of Medicine, The University of Hong KongHong Kong SARChina
| | - John A Herring
- Department of Orthopedic Surgery, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical SciencesYokohamaJapan
| | - You-qiang Song
- School of Biomedical Sciences, The University of Hong KongHong Kong SARChina
| | - Christina A Gurnett
- Department of Neurology, Washington University in St. LouisSt. LouisUnited States
| | - Paul Gerdhem
- Department of Surgical Sciences, Uppsala UniversityUppsalaSweden
- Department of Orthopaedics and Hand Surgery, Uppsala University HospitalUppsalaSweden
- Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet, Stockholm, Uppsala UniversityUppsalaSweden
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
| | - Jonathan J Rios
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pediatrics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Carol A Wise
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pediatrics, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
17
|
Orikasa S, Matsushita Y, Manabe H, Fogge M, Lee Z, Mizuhashi K, Sakagami N, Ono W, Ono N. Hedgehog activation promotes osteogenic fates of growth plate resting zone chondrocytes through transient clonal competency. JCI Insight 2024; 9:e165619. [PMID: 38051593 PMCID: PMC10906233 DOI: 10.1172/jci.insight.165619] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/05/2023] [Indexed: 12/07/2023] Open
Abstract
The resting zone of the postnatal growth plate is organized by slow-cycling chondrocytes expressing parathyroid hormone-related protein (PTHrP), which include a subgroup of skeletal stem cells that contribute to the formation of columnar chondrocytes. The PTHrP-Indian hedgehog feedback regulation is essential for sustaining growth plate activities; however, molecular mechanisms regulating cell fates of PTHrP+ resting chondrocytes and their eventual transformation into osteoblasts remain largely undefined. Here, in a mouse model, we specifically activated Hedgehog signaling in PTHrP+ resting chondrocytes and traced the fate of their descendants using a tamoxifen-inducible Pthrp-creER line with patched-1-floxed and tdTomato reporter alleles. Hedgehog-activated PTHrP+ chondrocytes formed large, concentric, clonally expanded cell populations within the resting zone ("patched roses") and generated significantly wider columns of chondrocytes, resulting in hyperplasia of the growth plate. Interestingly, Hedgehog-activated PTHrP+ cell descendants migrated away from the growth plate and transformed into trabecular osteoblasts in the diaphyseal marrow space in the long term. Therefore, Hedgehog activation drives resting zone chondrocytes into transit-amplifying states as proliferating chondrocytes and eventually converts these cells into osteoblasts, unraveling a potentially novel Hedgehog-mediated mechanism that facilitates osteogenic cell fates of PTHrP+ skeletal stem cells.
Collapse
Affiliation(s)
- Shion Orikasa
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Yuki Matsushita
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroaki Manabe
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Michael Fogge
- University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Zachary Lee
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Koji Mizuhashi
- University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Naoko Sakagami
- University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Wanida Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| |
Collapse
|
18
|
Li Z, Liu G, Yang L, Sun M, Zhang Z, Xu Z, Gao Y, Jiang X, Su Z, Li X, Yang Z. BMP7 expression in mammalian cortical radial glial cells increases the length of the neurogenic period. Protein Cell 2024; 15:21-35. [PMID: 37300483 PMCID: PMC10762677 DOI: 10.1093/procel/pwad036] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
The seat of human intelligence is the human cerebral cortex, which is responsible for our exceptional cognitive abilities. Identifying principles that lead to the development of the large-sized human cerebral cortex will shed light on what makes the human brain and species so special. The remarkable increase in the number of human cortical pyramidal neurons and the size of the human cerebral cortex is mainly because human cortical radial glial cells, primary neural stem cells in the cortex, generate cortical pyramidal neurons for more than 130 days, whereas the same process takes only about 7 days in mice. The molecular mechanisms underlying this difference are largely unknown. Here, we found that bone morphogenic protein 7 (BMP7) is expressed by increasing the number of cortical radial glial cells during mammalian evolution (mouse, ferret, monkey, and human). BMP7 expression in cortical radial glial cells promotes neurogenesis, inhibits gliogenesis, and thereby increases the length of the neurogenic period, whereas Sonic Hedgehog (SHH) signaling promotes cortical gliogenesis. We demonstrate that BMP7 signaling and SHH signaling mutually inhibit each other through regulation of GLI3 repressor formation. We propose that BMP7 drives the evolutionary expansion of the mammalian cortex by increasing the length of the neurogenic period.
Collapse
Affiliation(s)
- Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Lin Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Mengge Sun
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Zhejun Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Yanjing Gao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Xin Jiang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Zihao Su
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Xiaosu Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| |
Collapse
|
19
|
Matsushita Y, Noguchi A, Ono W, Ono N. Multi-omics analysis in developmental bone biology. JAPANESE DENTAL SCIENCE REVIEW 2023; 59:412-420. [PMID: 38022387 PMCID: PMC10665596 DOI: 10.1016/j.jdsr.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Single-cell omics and multi-omics have revolutionized our understanding of molecular and cellular biological processes at a single-cell level. In bone biology, the combination of single-cell RNA-sequencing analyses and in vivo lineage-tracing approaches has successfully identified multi-cellular diversity and dynamics of skeletal cells. This established a new concept that bone growth and regeneration are regulated by concerted actions of multiple types of skeletal stem cells, which reside in spatiotemporally distinct niches. One important subtype is endosteal stem cells that are particularly abundant in young bone marrow. The discovery of this new skeletal stem cell type has been facilitated by single-cell multi-omics, which simultaneously measures gene expression and chromatin accessibility. Using single-cell omics, it is now possible to computationally predict the immediate future state of individual cells and their differentiation potential. In vivo validation using histological approaches is the key to interpret the computational prediction. The emerging spatial omics, such as spatial transcriptomics and epigenomics, have major advantage in retaining the location of individual cells within highly complex tissue architecture. Spatial omics can be integrated with other omics to further obtain in-depth insights. Single-cell multi-omics are now becoming an essential tool to unravel intricate multicellular dynamics and intercellular interactions of skeletal cells.
Collapse
Affiliation(s)
- Yuki Matsushita
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Azumi Noguchi
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Wanida Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77054, USA
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77054, USA
| |
Collapse
|
20
|
Vaghjiani VG, Cochrane CR, Jayasekara WSN, Chong WC, Szczepny A, Kumar B, Martelotto LG, McCaw A, Carey K, Kansara M, Thomas DM, Walkley C, Mudge S, Gough DJ, Downie PA, Peacock CD, Matsui W, Watkins DN, Cain JE. Ligand-dependent hedgehog signaling maintains an undifferentiated, malignant osteosarcoma phenotype. Oncogene 2023; 42:3529-3541. [PMID: 37845394 PMCID: PMC10656285 DOI: 10.1038/s41388-023-02864-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023]
Abstract
TP53 and RB1 loss-of-function mutations are common in osteosarcoma. During development, combined loss of TP53 and RB1 function leads to downregulation of autophagy and the aberrant formation of primary cilia, cellular organelles essential for the transmission of canonical Hedgehog (Hh) signaling. Excess cilia formation then leads to hypersensitivity to Hedgehog (Hh) ligand signaling. In mouse and human models, we now show that osteosarcomas with mutations in TP53 and RB1 exhibit enhanced ligand-dependent Hh pathway activation through Smoothened (SMO), a transmembrane signaling molecule required for activation of the canonical Hh pathway. This dependence is mediated by hypersensitivity to Hh ligand and is accompanied by impaired autophagy and increased primary cilia formation and expression of Hh ligand in vivo. Using a conditional genetic mouse model of Trp53 and Rb1 inactivation in osteoblast progenitors, we further show that deletion of Smo converts the highly malignant osteosarcoma phenotype to benign, well differentiated bone tumors. Conversely, conditional overexpression of SHH ligand, or a gain-of-function SMO mutant in committed osteoblast progenitors during development blocks terminal bone differentiation. Finally, we demonstrate that the SMO antagonist sonidegib (LDE225) induces growth arrest and terminal differentiation in vivo in osteosarcomas that express primary cilia and Hh ligand combined with mutations in TP53. These results provide a mechanistic framework for aberrant Hh signaling in osteosarcoma based on defining mutations in the tumor suppressor, TP53.
Collapse
Affiliation(s)
| | - Catherine R Cochrane
- Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia
| | | | - Wai Chin Chong
- Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Anette Szczepny
- Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
| | - Beena Kumar
- Department of Pathology, Monash Medical Centre, Clayton, VIC, 3168, Australia
| | - Luciano G Martelotto
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Andrew McCaw
- Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
| | - Kirstyn Carey
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Maya Kansara
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - David M Thomas
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St.Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, NSW, 1466, Australia
| | - Carl Walkley
- St. Vincent's Institute, Fitzroy, VIC, 3065, Australia
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Stuart Mudge
- Mayne Pharma International Pty Ltd, Salisbury Sth, SA, 5106, Australia
| | - Daniel J Gough
- Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Peter A Downie
- Monash Children's Cancer Centre, Monash Children's Hospital, Monash Health, Clayton, VIC, 3168, Australia
- Department of Paediatrics, Monash University, Clayton, VIC, 3168, Australia
| | - Craig D Peacock
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH, 44106, USA
| | - William Matsui
- Department of Oncology and Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, 78712, USA
| | - D Neil Watkins
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB, R3E-0V9, Canada.
- Department of Internal Medicine, Rady Faculty of Heath Sciences, University of Manitoba, Winnipeg, MB, R3A-1R9, Canada.
| | - Jason E Cain
- Hudson Institute of Medical Research, Clayton, VIC, 3168, Australia.
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3800, Australia.
- Department of Paediatrics, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
21
|
Roberson EC, Tran NK, Godambe AN, Mark H, Nguimtsop M, Rust T, Ung E, Barker LJ, Fitch RD, Wallingford JB. Hedgehog signaling is required for endometrial remodeling and myometrial homeostasis in the cycling mouse uterus. iScience 2023; 26:107993. [PMID: 37810243 PMCID: PMC10551904 DOI: 10.1016/j.isci.2023.107993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/24/2023] [Accepted: 09/16/2023] [Indexed: 10/10/2023] Open
Abstract
Decades of work demonstrate that the mammalian estrous cycle is controlled by cycling steroid hormones. However, the signaling mechanisms that act downstream, linking hormonal action to the physical remodeling of the cycling uterus, remain unclear. To address this issue, we analyzed gene expression at all stages of the mouse estrous cycle. Strikingly, we found that several genetic programs well-known to control tissue morphogenesis in developing embryos displayed cyclical patterns of expression. We find that most of the genetic architectures of Hedgehog signaling (ligands, receptors, effectors, and transcription factors) are transcribed cyclically in the uterus, and that conditional disruption of the Hedgehog receptor smoothened not only elicits a failure of normal cyclical thickening of the endometrial lining but also induces aberrant deformation of the uterine smooth muscle. Together, our data shed light on the mechanisms underlying normal uterine remodeling specifically and cyclical gene expression generally.
Collapse
Affiliation(s)
- Elle C. Roberson
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - Ngan Kim Tran
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Anushka N. Godambe
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Harrison Mark
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Michelle Nguimtsop
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Trinity Rust
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Elizabeth Ung
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - LeCaine J. Barker
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical School, Aurora, CO 80045, USA
| | - Rebecca D. Fitch
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - John B. Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
22
|
Gurdita A, Pham Truong VQB, Dolati P, Juric M, Tachibana N, Liu ZC, Ortín-Martínez A, Ibrahimi M, Pokrajac NT, Comanita L, Pacal M, Huang M, Sugita S, Bremner R, Wallace VA. Progenitor division and cell autonomous neurosecretion are required for rod photoreceptor sublaminar positioning. Proc Natl Acad Sci U S A 2023; 120:e2308204120. [PMID: 37812728 PMCID: PMC10589646 DOI: 10.1073/pnas.2308204120] [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: 06/13/2023] [Accepted: 09/06/2023] [Indexed: 10/11/2023] Open
Abstract
Migration is essential for the laminar stratification and connectivity of neurons in the central nervous system. In the retina, photoreceptors (PRs) migrate to positions according to birthdate, with early-born cells localizing to the basal-most side of the outer nuclear layer. It was proposed that apical progenitor mitoses physically drive these basal translocations non-cell autonomously, but direct evidence is lacking, and whether other mechanisms participate is unknown. Here, combining loss- or gain-of-function assays to manipulate cell cycle regulators (Sonic hedgehog, Cdkn1a/p21) with an in vivo lentiviral labelling strategy, we demonstrate that progenitor division is one of two forces driving basal translocation of rod soma. Indeed, replacing Shh activity rescues abnormal rod translocation in retinal explants. Unexpectedly, we show that rod differentiation also promotes rod soma translocation. While outer segment function or formation is dispensable, Crx and SNARE-dependent synaptic function are essential. Thus, both non-cell and cell autonomous mechanisms underpin PR soma sublaminar positioning in the mammalian retina.
Collapse
Affiliation(s)
- Akshay Gurdita
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S 1A8, Canada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Victor Q. B. Pham Truong
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S 1A8, Canada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Parnian Dolati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S 1A8, Canada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Matey Juric
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Nobuhiko Tachibana
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Zhongda C. Liu
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Arturo Ortín-Martínez
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Mostafa Ibrahimi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S 1A8, Canada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Nenad T. Pokrajac
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S 1A8, Canada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Lacrimioara Comanita
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
| | - Marek Pacal
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ONM5G 1X5, Canada
| | - Mengjia Huang
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ONM5T 2S8, Canada
- Department of Physiology, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Shuzo Sugita
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute, University Health Network, Toronto, ONM5T 2S8, Canada
- Department of Physiology, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Rod Bremner
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S 1A8, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ONM5G 1X5, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ONM5T 3A9, Canada
| | - Valerie A. Wallace
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S 1A8, Canada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ONM5T 2S8, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ONM5T 3A9, Canada
| |
Collapse
|
23
|
Michalski MN, Williams BO. The Past, Present, and Future of Genetically Engineered Mouse Models for Skeletal Biology. Biomolecules 2023; 13:1311. [PMID: 37759711 PMCID: PMC10526739 DOI: 10.3390/biom13091311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The ability to create genetically engineered mouse models (GEMMs) has exponentially increased our understanding of many areas of biology. Musculoskeletal biology is no exception. In this review, we will first discuss the historical development of GEMMs and how these developments have influenced musculoskeletal disease research. This review will also update our 2008 review that appeared in BONEKey, a journal that is no longer readily available online. We will first review the historical development of GEMMs in general, followed by a particular emphasis on the ability to perform tissue-specific (conditional) knockouts focusing on musculoskeletal tissues. We will then discuss how the development of CRISPR/Cas-based technologies during the last decade has revolutionized the generation of GEMMs.
Collapse
Affiliation(s)
- Megan N. Michalski
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA;
| | - Bart O. Williams
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI 49503, USA;
- Core Technologies and Services, Van Andel Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
24
|
Pathmanapan S, Poon R, De Renshaw TB, Nadesan P, Nakagawa M, Seesankar GA, Ho Loe AK, Zhang HH, Guinovart JJ, Duran J, Newgard CB, Wunder JS, Alman BA. Mutant IDH regulates glycogen metabolism from early cartilage development to malignant chondrosarcoma formation. Cell Rep 2023; 42:112578. [PMID: 37267108 PMCID: PMC10592452 DOI: 10.1016/j.celrep.2023.112578] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/22/2023] [Accepted: 05/16/2023] [Indexed: 06/04/2023] Open
Abstract
Chondrosarcomas are the most common malignancy of cartilage and are associated with somatic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 genes. Somatic IDH mutations are also found in its benign precursor lesion, enchondromas, suggesting that IDH mutations are early events in malignant transformation. Human mutant IDH chondrosarcomas and mutant Idh mice that develop enchondromas investigated in our studies display glycogen deposition exclusively in mutant cells from IDH mutant chondrosarcomas and Idh1 mutant murine growth plates. Pharmacologic blockade of glycogen utilization induces changes in tumor cell behavior, downstream energetic pathways, and tumor burden in vitro and in vivo. Mutant IDH1 interacts with hypoxia-inducible factor 1α (HIF1α) to regulate expression of key enzymes in glycogen metabolism. Here, we show a critical role for glycogen in enchondromas and chondrosarcomas, which is likely mediated through an interaction with mutant IDH1 and HIF1α.
Collapse
Affiliation(s)
- Sinthu Pathmanapan
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Raymond Poon
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | | | | | - Makoto Nakagawa
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Gireesh A Seesankar
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Adrian Kwan Ho Loe
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Hongyuan H Zhang
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona) Barcelona, Barcelona, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona) Barcelona, Barcelona, Spain
| | - Christopher B Newgard
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, USA; Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Jay S Wunder
- Lunenfeld-Tanenbaum Research Institute and the University Musculoskeletal Oncology Unit, Mount Sinai Hospital, Toronto, ON, Canada
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA.
| |
Collapse
|
25
|
Shigley C, Trivedi J, Meghani O, Owens BD, Jayasuriya CT. Suppressing Chondrocyte Hypertrophy to Build Better Cartilage. Bioengineering (Basel) 2023; 10:741. [PMID: 37370672 DOI: 10.3390/bioengineering10060741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Current clinical strategies for restoring cartilage defects do not adequately consider taking the necessary steps to prevent the formation of hypertrophic tissue at injury sites. Chondrocyte hypertrophy inevitably causes both macroscopic and microscopic level changes in cartilage, resulting in adverse long-term outcomes following attempted restoration. Repairing/restoring articular cartilage while minimizing the risk of hypertrophic neo tissue formation represents an unmet clinical challenge. Previous investigations have extensively identified and characterized the biological mechanisms that regulate cartilage hypertrophy with preclinical studies now beginning to leverage this knowledge to help build better cartilage. In this comprehensive article, we will provide a summary of these biological mechanisms and systematically review the most cutting-edge strategies for circumventing this pathological hallmark of osteoarthritis.
Collapse
Affiliation(s)
- Christian Shigley
- The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Jay Trivedi
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Ozair Meghani
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Brett D Owens
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Sports Surgery, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
26
|
Taieb M, Ghannoum D, Barré L, Ouzzine M. Xylosyltransferase I mediates the synthesis of proteoglycans with long glycosaminoglycan chains and controls chondrocyte hypertrophy and collagen fibers organization of in the growth plate. Cell Death Dis 2023; 14:355. [PMID: 37296099 PMCID: PMC10256685 DOI: 10.1038/s41419-023-05875-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/06/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Genetic mutations in the Xylt1 gene are associated with Desbuquois dysplasia type II syndrome characterized by sever prenatal and postnatal short stature. However, the specific role of XylT-I in the growth plate is not completely understood. Here, we show that XylT-I is expressed and critical for the synthesis of proteoglycans in resting and proliferative but not in hypertrophic chondrocytes in the growth plate. We found that loss of XylT-I induces hypertrophic phenotype-like of chondrocytes associated with reduced interterritorial matrix. Mechanistically, deletion of XylT-I impairs the synthesis of long glycosaminoglycan chains leading to the formation of proteoglycans with shorter glycosaminoglycan chains. Histological and Second Harmonic Generation microscopy analysis revealed that deletion of XylT-I accelerated chondrocyte maturation and prevents chondrocytes columnar organization and arrangement in parallel of collagen fibers in the growth plate, suggesting that XylT-I controls chondrocyte maturation and matrix organization. Intriguingly, loss of XylT-I induced at embryonic stage E18.5 the migration of progenitor cells from the perichondrium next to the groove of Ranvier into the central part of epiphysis of E18.5 embryos. These cells characterized by higher expression of glycosaminoglycans exhibit circular organization then undergo hypertrophy and death creating a circular structure at the secondary ossification center location. Our study revealed an uncovered role of XylT-I in the synthesis of proteoglycans and provides evidence that the structure of glycosaminoglycan chains of proteoglycans controls chondrocyte maturation and matrix organization.
Collapse
Affiliation(s)
- Mahdia Taieb
- UMR 7365 CNRS-University of Lorraine, Biopôle, Faculty of Medicine, BP 20199, 54505, Vandoeuvre-lès-Nancy, CEDEX, France
| | - Dima Ghannoum
- UMR 7365 CNRS-University of Lorraine, Biopôle, Faculty of Medicine, BP 20199, 54505, Vandoeuvre-lès-Nancy, CEDEX, France
| | - Lydia Barré
- UMR 7365 CNRS-University of Lorraine, Biopôle, Faculty of Medicine, BP 20199, 54505, Vandoeuvre-lès-Nancy, CEDEX, France
| | - Mohamed Ouzzine
- UMR 7365 CNRS-University of Lorraine, Biopôle, Faculty of Medicine, BP 20199, 54505, Vandoeuvre-lès-Nancy, CEDEX, France.
| |
Collapse
|
27
|
Fan M, Qiang L, Wang Y, Liu Y, Zhuang H, Guo R, Ben Y, Li Q, Zheng P. 3D bioprinted hydrogel/polymer scaffold with factor delivery and mechanical support for growth plate injury repair. Front Bioeng Biotechnol 2023; 11:1210786. [PMID: 37324424 PMCID: PMC10265638 DOI: 10.3389/fbioe.2023.1210786] [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: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Growth plate injury is a significant challenge in clinical practice, as it could severely affect the limb development of children, leading to limb deformity. Tissue engineering and 3D bioprinting technology have great potential in the repair and regeneration of injured growth plate, but there are still challenges associated with achieving successful repair outcomes. Methods: In this study, GelMA hydrogel containing PLGA microspheres loaded with chondrogenic factor PTH(1-34) was combined with BMSCs and Polycaprolactone (PCL) to develop the PTH(1-34)@PLGA/BMSCs/GelMA-PCL scaffold using bio-3D printing technology. Results: The scaffold exhibited a three-dimensional interconnected porous network structure, good mechanical properties, biocompatibility, and was suitable for cellchondrogenic differentiation. And a rabbit model of growth plate injury was appliedto validate the effect of scaffold on the repair of injured growth plate. The resultsshowed that the scaffold was more effective than injectable hydrogel in promotingcartilage regeneration and reducing bone bridge formation. Moreover, the addition ofPCL to the scaffold provided good mechanical support, significantly reducing limbdeformities after growth plate injury compared with directly injected hydrogel. Discussion: Accordingly, our study demonstrates the feasibility of using 3D printed scaffolds for treating growth plate injuries and could offer a new strategy for the development of growth plate tissue engineering therapy.
Collapse
Affiliation(s)
- Minjie Fan
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Qiang
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Advanced Technologies of Materials (MOE), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Yiwei Wang
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yihao Liu
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanjie Zhuang
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruoyi Guo
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yulong Ben
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiang Li
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pengfei Zheng
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
28
|
Lin YC, Sahoo BK, Gau SS, Yang RB. The biology of SCUBE. J Biomed Sci 2023; 30:33. [PMID: 37237303 PMCID: PMC10214685 DOI: 10.1186/s12929-023-00925-3] [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: 02/20/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
The SCUBE [Signal peptide-Complement C1r/C1s, Uegf, Bmp1 (CUB)-Epithelial growth factor domain-containing protein] family consists of three proteins in vertebrates, SCUBE1, 2 and 3, which are highly conserved in zebrafish, mice and humans. Each SCUBE gene encodes a polypeptide of approximately 1000 amino acids that is organized into five modular domains: (1) an N-terminal signal peptide sequence, (2) nine tandem epidermal growth factor (EGF)-like repeats, (3) a large spacer region, (4) three cysteine-rich (CR) motifs, and (5) a CUB domain at the C-terminus. Murine Scube genes are expressed individually or in combination during the development of various tissues, including those in the central nervous system and the axial skeleton. The cDNAs of human SCUBE orthologs were originally cloned from vascular endothelial cells, but SCUBE expression has also been found in platelets, mammary ductal epithelium and osteoblasts. Both soluble and membrane-associated SCUBEs have been shown to play important roles in physiology and pathology. For instance, upregulation of SCUBEs has been reported in acute myeloid leukemia, breast cancer and lung cancer. In addition, soluble SCUBE1 is released from activated platelets and can be used as a clinical biomarker for acute coronary syndrome and ischemic stroke. Soluble SCUBE2 enhances distal signaling by facilitating the secretion of dual-lipidated hedgehog from nearby ligand-producing cells in a paracrine manner. Interestingly, the spacer regions and CR motifs can increase or enable SCUBE binding to cell surfaces via electrostatic or glycan-lectin interactions. As such, membrane-associated SCUBEs can function as coreceptors that enhance the signaling activity of various serine/threonine kinase or tyrosine kinase receptors. For example, membrane-associated SCUBE3 functions as a coreceptor that promotes signaling in bone morphogenesis. In humans, SCUBE3 mutations are linked to abnormalities in growth and differentiation of both bones and teeth. In addition to studies on human SCUBE function, experimental results from genetically modified mouse models have yielded important insights in the field of systems biology. In this review, we highlight novel molecular discoveries and critical directions for future research on SCUBE proteins in the context of cancer, skeletal disease and cardiovascular disease.
Collapse
Affiliation(s)
- Yuh-Charn Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Binay K Sahoo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shiang-Shin Gau
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan.
- Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
29
|
Richard D, Pregizer S, Venkatasubramanian D, Raftery RM, Muthuirulan P, Liu Z, Capellini TD, Craft AM. Lineage-specific differences and regulatory networks governing human chondrocyte development. eLife 2023; 12:e79925. [PMID: 36920035 PMCID: PMC10069868 DOI: 10.7554/elife.79925] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 03/14/2023] [Indexed: 03/16/2023] Open
Abstract
To address large gaps in our understanding of the molecular regulation of articular and growth plate cartilage development in humans, we used our directed differentiation approach to generate these distinct cartilage tissues from human embryonic stem cells. The resulting transcriptomic profiles of hESC-derived articular and growth plate chondrocytes were similar to fetal epiphyseal and growth plate chondrocytes, with respect to genes both known and previously unknown to cartilage biology. With the goal to characterize the regulatory landscapes accompanying these respective transcriptomes, we mapped chromatin accessibility in hESC-derived chondrocyte lineages, and mouse embryonic chondrocytes, using ATAC-sequencing. Integration of the expression dataset with the differentially accessible genomic regions revealed lineage-specific gene regulatory networks. We validated functional interactions of two transcription factors (TFs) (RUNX2 in growth plate chondrocytes and RELA in articular chondrocytes) with their predicted genomic targets. The maps we provide thus represent a framework for probing regulatory interactions governing chondrocyte differentiation. This work constitutes a substantial step towards comprehensive and comparative molecular characterizations of distinct chondrogenic lineages and sheds new light on human cartilage development and biology.
Collapse
Affiliation(s)
- Daniel Richard
- Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Steven Pregizer
- Department of Orthopedic Research, Boston Children’s HospitalBostonUnited States
- Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
| | - Divya Venkatasubramanian
- Department of Orthopedic Research, Boston Children’s HospitalBostonUnited States
- Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
- Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Rosanne M Raftery
- Department of Orthopedic Research, Boston Children’s HospitalBostonUnited States
- Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
| | | | - Zun Liu
- Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Terence D Capellini
- Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Broad Institute of MIT and HarvardCambridgeUnited States
| | - April M Craft
- Department of Orthopedic Research, Boston Children’s HospitalBostonUnited States
- Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| |
Collapse
|
30
|
Ishihara S, Usumi-Fujita R, Kasahara Y, Oishi S, Shibata K, Shimizu Y, Ishida Y, Kaneko S, Sugiura-Nakazato M, Tabata MJ, Hosomichi J, Taniyama Y, Ono T. Periostin splice variants affect craniofacial growth by influencing chondrocyte hypertrophy. J Bone Miner Metab 2023; 41:171-181. [PMID: 36859617 DOI: 10.1007/s00774-023-01409-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/30/2023] [Indexed: 03/03/2023]
Abstract
INTRODUCTION Periostin, an extracellular matrix protein, plays an important role in osteogenesis and is also known to activate several signals that contribute to chondrogenesis. The absence of periostin in periostin knockout mice leads to several disorders such as craniosynostosis and periostitis. There are several splice variants with different roles in heart disease and myocardial infarction. However, little is known about each variant's role in chondrogenesis, followed by bone formation. Therefore, the aim of this study is to investigate the role of several variants in chondrogenesis differentiation and bone formation in the craniofacial region. Periostin splice variants included a full-length variant (Control), a variant lacking exon 17 (ΔEx17), a variant lacking exon 21 (ΔEx21), and another variant lacking both exon 17 and 21 ***(ΔEx17&21). MATERIALS AND METHODS We used C56BL6/N mice (n = 6) for the wild type (Control)*** and the three variant type mice (n = 6 each) to identify the effect of each variant morphologically and histologically. Micro-computed tomography demonstrated a smaller craniofacial skeleton in ΔEx17s, ΔEx21s, and ΔEx17&21s compared to Controls, especially the mandibular bone. We, thus, focused on the mandibular condyle. RESULTS The most distinctive histological observation was that each defected mouse appeared to have more hypertrophic chondrocytes than Controls. Real-time PCR demonstrated the differences among the group. Moreover, the lack of exon 17 or exon 21 in periostin leads to inadequate chondrocyte differentiation and presents in a diminutive craniofacial skeleton. DISCUSSION Therefore, these findings suggested that each variant has a significant role in chondrocyte hypertrophy, leading to suppression of bone formation.
Collapse
Affiliation(s)
- Seiko Ishihara
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan
| | - Risa Usumi-Fujita
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan.
| | - Yuki Kasahara
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan
| | - Shuji Oishi
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan
| | - Kana Shibata
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasuhiro Shimizu
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan
| | - Yuji Ishida
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan
| | - Sawa Kaneko
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan
| | - Makoto Sugiura-Nakazato
- Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Makoto J Tabata
- Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Jun Hosomichi
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan
| | - Yoshiaki Taniyama
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Yushima 1-5-45, Tokyo, Japan
| |
Collapse
|
31
|
Jung HJ, Yeo S, Jang J, Pleasure S, Choe Y. Brain heterotopia formation by ciliopathic breakdown of neuroepithelial and blood-cerebrospinal fluid barriers. Brain Pathol 2023:e13148. [PMID: 36623505 DOI: 10.1111/bpa.13148] [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: 09/22/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
The developmental functions of primary cilia and the downstream signaling pathways have been widely studied; however, the roles of primary cilia in the developing neurovascular system are not clearly understood. In this study, we found that ablation of genes encoding ciliary transport proteins such as intraflagellar transport homolog 88 (Ift88) and kinesin family member 3a (Kif3a) in cortical radial progenitors led to periventricular heterotopia during late mouse embryogenesis. Conditional mutation of primary cilia unexpectedly caused breakdown of both the neuroepithelial lining and the blood-choroid plexus barrier. Choroidal leakage was partially caused by enlargement of the choroid plexus in the cilia mutants. We found that the choroid plexus expressed platelet-derived growth factor A (Pdgf-A) and that Pdgf-A expression was ectopically increased in cilia-mutant embryos. Cortices obtained from embryos in utero electroporated with Pdgfa mimicked periventricular heterotopic nodules of the cilia mutant. These results suggest that defective ciliogenesis in both cortical progenitors and the choroid plexus leads to breakdown of cortical and choroidal barriers causing forebrain neuronal dysplasia, which may be related to developmental cortical malformation.
Collapse
Affiliation(s)
| | - Seungeun Yeo
- Korea Brain Research Institute, Daegu, South Korea
| | | | - Samuel Pleasure
- Department of Neurology, Program in Neuroscience, Developmental Stem Cell Biology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and University of California, San Francisco, California, USA
| | | |
Collapse
|
32
|
Hasan MR, Koskenranta A, Alakurtti K, Takatalo M, Rice DP. RAB23 regulates musculoskeletal development and patterning. Front Cell Dev Biol 2023; 11:1049131. [PMID: 36910145 PMCID: PMC9995984 DOI: 10.3389/fcell.2023.1049131] [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: 09/20/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
RAB23 is a small GTPase which functions at the plasma membrane to regulate growth factor signaling. Mutations in RAB23 cause Carpenter syndrome, a condition that affects normal organogenesis and patterning. In this study, we investigate the role of RAB23 in musculoskeletal development and show that it is required for patella bone formation and for the maintenance of tendon progenitors. The patella is the largest sesamoid bone in mammals and plays a critical role during movement by providing structural and mechanical support to the knee. Rab23 -/- mice fail to form a patella and normal knee joint. The patella is formed from Sox9 and scleraxis (Scx) double-positive chondroprogenitor cells. We show that RAB23 is required for the specification of SOX9 and scleraxis double-positive patella chondroprogenitors during the formation of patella anlagen and the subsequent establishment of patellofemoral joint. We find that scleraxis and SOX9 expression are disrupted in Rab23 -/- mice, and as a result, development of the quadriceps tendons, cruciate ligaments, patella tendons, and entheses is either abnormal or lost. TGFβ-BMP signaling is known to regulate patella initiation and patella progenitor differentiation and growth. We find that the expression of TGFβR2, BMPR1, BMP4, and pSmad are barely detectable in the future patella site and in the rudimentary tendons and ligaments around the patellofemoral joint in Rab23 -/- mice. Also, we show that GLI1, SOX9, and scleraxis, which regulate entheses establishment and maturation, are weakly expressed in Rab23 -/- mice. Further analysis of the skeletal phenotype of Rab23 -/- mice showed a close resemblance to that of Tgfβ2 -/- mice, highlighting a possible role for RAB23 in regulating TGFβ superfamily signaling.
Collapse
Affiliation(s)
- Md Rakibul Hasan
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Anna Koskenranta
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Kirsi Alakurtti
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Maarit Takatalo
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - David P Rice
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
33
|
Chen T, Dalton G, Oh SH, Maeso-Diaz R, Du K, Meyers RA, Guy C, Abdelmalek MF, Henao R, Guarnieri P, Pullen SS, Gregory S, Locker J, Brown JM, Diehl AM. Hepatocyte Smoothened Activity Controls Susceptibility to Insulin Resistance and Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2022; 15:949-970. [PMID: 36535507 PMCID: PMC9957752 DOI: 10.1016/j.jcmgh.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH), a leading cause of cirrhosis, strongly associates with the metabolic syndrome, an insulin-resistant proinflammatory state that disrupts energy balance and promotes progressive liver degeneration. We aimed to define the role of Smoothened (Smo), an obligatory component of the Hedgehog signaling pathway, in controlling hepatocyte metabolic homeostasis and, thereby, susceptibility to NASH. METHODS We conditionally deleted Smo in hepatocytes of healthy chow-fed mice and performed metabolic phenotyping, coupled with single-cell RNA sequencing (RNA-seq), to characterize the role of hepatocyte Smo in regulating basal hepatic and systemic metabolic homeostasis. Liver RNA-seq datasets from 2 large human cohorts were also analyzed to define the relationship between Smo and NASH susceptibility in people. RESULTS Hepatocyte Smo deletion inhibited the Hedgehog pathway and promoted fatty liver, hyperinsulinemia, and insulin resistance. We identified a plausible mechanism whereby inactivation of Smo stimulated the mTORC1-SREBP1c signaling axis, which promoted lipogenesis while inhibiting the hepatic insulin cascade. Transcriptomics of bulk and single Smo-deficient hepatocytes supported suppression of insulin signaling and also revealed molecular abnormalities associated with oxidative stress and mitochondrial dysfunction. Analysis of human bulk RNA-seq data revealed that Smo expression was (1) highest in healthy livers, (2) lower in livers with NASH than in those with simple steatosis, (3) negatively correlated with markers of insulin resistance and liver injury, and (4) declined progressively as fibrosis severity worsened. CONCLUSIONS The Hedgehog pathway controls insulin sensitivity and energy homeostasis in adult livers. Loss of hepatocyte Hedgehog activity induces hepatic and systemic metabolic stress and enhances susceptibility to NASH by promoting hepatic lipoxicity and insulin resistance.
Collapse
Affiliation(s)
- Tianyi Chen
- Department of Medicine, Duke University, Durham, North Carolina
| | - George Dalton
- Department of Medicine, Duke University, Durham, North Carolina
| | - Seh-Hoon Oh
- Department of Medicine, Duke University, Durham, North Carolina
| | | | - Kuo Du
- Department of Medicine, Duke University, Durham, North Carolina
| | - Rachel A Meyers
- Department of Medicine, Duke University, Durham, North Carolina
| | - Cynthia Guy
- Department of Medicine, Duke University, Durham, North Carolina
| | | | - Ricardo Henao
- Department of Medicine, Duke University, Durham, North Carolina
| | - Paolo Guarnieri
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Steven S Pullen
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Simon Gregory
- Department of Medicine, Duke University, Durham, North Carolina
| | - Joseph Locker
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - J Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, North Carolina.
| |
Collapse
|
34
|
Effect of Sonic Hedgehog on the Regeneration of Epidermal Texture Patterns. Biomedicines 2022; 10:biomedicines10123099. [PMID: 36551853 PMCID: PMC9776110 DOI: 10.3390/biomedicines10123099] [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: 10/01/2022] [Revised: 11/15/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Wounds on embryonic mouse fetuses regenerate up to embryonic day (E) 13, but after E14, the pattern is lost and a visible scar remains. We hypothesized that the sonic hedgehog (Shh), which is involved in patterning during development, is involved in the regeneration of texture. Embryos of ICR mice were surgically injured at E13, E14, and E15 and analyzed for the expression of Shh. For external Shh administration, recombinant Shh-containing slow-release beads were implanted in the wounds of mice. In contrast, cyclopamine was administered to wounds of adult mice to inhibit Shh. The expression of Shh was unaltered at E13, whereas it was upregulated in the epidermis of the wound from E14 onward. Implantation of recombinant Shh-containing beads into E13 wounds inhibited skin texture regeneration. Cyclopamine treatment inhibited epithelialization and thickening of the epidermis in the wounds of adult mice. In vitro, Shh promoted proliferation and inhibited the migration of epidermal keratinocytes through the activation of cyclin D proteins. Thus, our results suggested that the expression of Shh is involved in the regeneration of texture during wound healing, especially in epidermal keratinocyte migration and division, and could inhibit skin texture regeneration after E14.
Collapse
|
35
|
Xu J, Liu H, Lan Y, Jiang R. The transcription factors Foxf1 and Foxf2 integrate the SHH, HGF and TGFβ signaling pathways to drive tongue organogenesis. Development 2022; 149:dev200667. [PMID: 36227576 PMCID: PMC10655918 DOI: 10.1242/dev.200667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 09/26/2022] [Indexed: 11/19/2023]
Abstract
The tongue is a highly specialized muscular organ with diverse cellular origins, which provides an excellent model for understanding mechanisms controlling tissue-tissue interactions during organogenesis. Previous studies showed that SHH signaling is required for tongue morphogenesis and tongue muscle organization, but little is known about the underlying mechanisms. Here we demonstrate that the Foxf1/Foxf2 transcription factors act in the cranial neural crest cell (CNCC)-derived mandibular mesenchyme to control myoblast migration into the tongue primordium during tongue initiation, and thereafter continue to regulate intrinsic tongue muscle assembly and lingual tendon formation. We performed chromatin immunoprecipitation sequencing analysis and identified Hgf, Tgfb2 and Tgfb3 among the target genes of Foxf2 in the embryonic tongue. Through genetic analyses of mice with CNCC-specific inactivation of Smo or both Foxf1 and Foxf2, we show that Foxf1 and Foxf2 mediate hedgehog signaling-mediated regulation of myoblast migration during tongue initiation and intrinsic tongue muscle formation by regulating the activation of the HGF and TGFβ signaling pathways. These data uncover the molecular network integrating the SHH, HGF and TGFβ signaling pathways in regulating tongue organogenesis.
Collapse
Affiliation(s)
- Jingyue Xu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Han Liu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yu Lan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Departments of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Departments of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
36
|
MPSI Manifestations and Treatment Outcome: Skeletal Focus. Int J Mol Sci 2022; 23:ijms231911168. [PMID: 36232472 PMCID: PMC9569890 DOI: 10.3390/ijms231911168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/23/2022] Open
Abstract
Mucopolysaccharidosis type I (MPSI) (OMIM #252800) is an autosomal recessive disorder caused by pathogenic variants in the IDUA gene encoding for the lysosomal alpha-L-iduronidase enzyme. The deficiency of this enzyme causes systemic accumulation of glycosaminoglycans (GAGs). Although disease manifestations are typically not apparent at birth, they can present early in life, are progressive, and include a wide spectrum of phenotypic findings. Among these, the storage of GAGs within the lysosomes disrupts cell function and metabolism in the cartilage, thus impairing normal bone development and ossification. Skeletal manifestations of MPSI are often refractory to treatment and severely affect patients’ quality of life. This review discusses the pathological and molecular processes leading to impaired endochondral ossification in MPSI patients and the limitations of current therapeutic approaches. Understanding the underlying mechanisms responsible for the skeletal phenotype in MPSI patients is crucial, as it could lead to the development of new therapeutic strategies targeting the skeletal abnormalities of MPSI in the early stages of the disease.
Collapse
|
37
|
Zhu J, Patel R, Trofka A, Harfe BD, Mackem S. Sonic hedgehog is not a limb morphogen but acts as a trigger to specify all digits in mice. Dev Cell 2022; 57:2048-2062.e4. [PMID: 35977544 PMCID: PMC9709693 DOI: 10.1016/j.devcel.2022.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/03/2022] [Accepted: 07/26/2022] [Indexed: 11/03/2022]
Abstract
Limb patterning by Sonic hedgehog (Shh), via either graded spatial or temporal signal integration, is a paradigm for "morphogen" function, yet how Shh instructs distinct digit identities remains controversial. Here, we bypass the Shh requirement in cell survival during outgrowth and demonstrate that a transient, early Shh pulse is both necessary and sufficient for normal mouse limb development. Shh response is only short range and is limited to the Shh-expressing zone during this time window. Shh patterns digits 1-3, anterior to this zone, by an indirect mechanism rather than direct spatial or temporal signal integration. Using a genetic relay-signaling assay, we discover that Shh also specifies digit 1/thumb (thought to be exclusively Shh independent) indirectly, and this finding implicates Shh in a unique regulatory hierarchy for digit 1 evolutionary adaptations such as opposable thumbs. This study illuminates Shh as a trigger for an indirect downstream network that becomes rapidly self-sustaining, with mechanistic relevance for limb development, regeneration, and evolution.
Collapse
Affiliation(s)
- Jianjian Zhu
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI, Frederick, MD, USA
| | - Rashmi Patel
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI, Frederick, MD, USA
| | - Anna Trofka
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI, Frederick, MD, USA
| | - Brian D Harfe
- College of Medicine, Department of Molecular Genetics and Microbiology and the Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Susan Mackem
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI, Frederick, MD, USA.
| |
Collapse
|
38
|
Xiu C, Gong T, Luo N, Ma L, Zhang L, Chen J. Suppressor of fused-restrained Hedgehog signaling in chondrocytes is critical for epiphyseal growth plate maintenance and limb elongation in juvenile mice. Front Cell Dev Biol 2022; 10:997838. [PMID: 36120578 PMCID: PMC9479194 DOI: 10.3389/fcell.2022.997838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
Hedgehog (Hh) signaling plays multiple critical roles in regulating chondrocyte proliferation and differentiation during epiphyseal cartilage development. However, it is still unclear whether Hh signaling in chondrocytes is required for growth plate maintenance during juvenile growth, and whether sustained activation of Hh signaling in chondrocytes promotes limb elongation. In this study, we first utilized Hh reporter mice to reveal that Hh signaling was activated in resting and columnar chondrocytes in growth plates of juvenile and adult mice. Next, we genetically modulated Hh signaling by conditionally deleting Smo or Sufu in all or a subpopulation of growth plate chondrocytes, and found that ablation of either Smo or Sufu in chondrocytes of juvenile mice caused premature closure of growth plates and shorter limbs, whereas Osx-Cre-mediated deletion of either of these two genes in prehypertrophic chondrocytes did not lead to obvious growth plate defects, indicating that Hh signaling mainly functions in resting and/or columnar chondrocytes to maintain growth plates at the juvenile stage. At the cellular level, we found that chondrocyte-specific ablation of Smo or Sufu accelerated or suppressed chondrocyte hypertrophy, respectively, whereas both decreased chondrocyte proliferation and survival. Thus, our study provided the first genetic evidence to establish the essential cell-autonomous roles for tightly-regulated Hh signaling in epiphyseal growth plate maintenance and limb elongation during juvenile growth.
Collapse
Affiliation(s)
- Chunmei Xiu
- Orthopedic Institute, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Tingting Gong
- Orthopedic Institute, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Na Luo
- Orthopedic Institute, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Linghui Ma
- Orthopedic Institute, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Lei Zhang
- Orthopedic Institute, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- *Correspondence: Jianquan Chen, ; Lei Zhang,
| | - Jianquan Chen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
- Department of Orthopaedics, First Affiliated Hospital of Soochow University, Orthopedic Institute, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- *Correspondence: Jianquan Chen, ; Lei Zhang,
| |
Collapse
|
39
|
Kurz J, Weiss AC, Lüdtke THW, Deuper L, Trowe MO, Thiesler H, Hildebrandt H, Heineke J, Duncan SA, Kispert A. GATA6 is a crucial factor for Myocd expression in the visceral smooth muscle cell differentiation program of the murine ureter. Development 2022; 149:dev200522. [PMID: 35905011 PMCID: PMC10656427 DOI: 10.1242/dev.200522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2023]
Abstract
Smooth muscle cells (SMCs) are a crucial component of the mesenchymal wall of the ureter, as they account for the efficient removal of the urine from the renal pelvis to the bladder by means of their contractile activity. Here, we show that the zinc-finger transcription factor gene Gata6 is expressed in mesenchymal precursors of ureteric SMCs under the control of BMP4 signaling. Mice with a conditional loss of Gata6 in these precursors exhibit a delayed onset and reduced level of SMC differentiation and peristaltic activity, as well as dilatation of the ureter and renal pelvis (hydroureternephrosis) at birth and at postnatal stages. Molecular profiling revealed a delayed and reduced expression of the myogenic driver gene Myocd, but the activation of signaling pathways and transcription factors previously implicated in activation of the visceral SMC program in the ureter was unchanged. Additional gain-of-function experiments suggest that GATA6 cooperates with FOXF1 in Myocd activation and SMC differentiation, possibly as pioneer and lineage-determining factors, respectively.
Collapse
Affiliation(s)
- Jennifer Kurz
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Anna-Carina Weiss
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Timo H.-W. Lüdtke
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Lena Deuper
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Hauke Thiesler
- Institut für Klinische Biochemie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institut für Klinische Biochemie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Joerg Heineke
- Abteilung für Kardiovaskuläre Physiologie, European Center for Angioscience, Medizinische Fakultät Mannheim, Universität Heidelberg, D-68167 Mannheim, Germany
| | - Stephen A. Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| |
Collapse
|
40
|
Bell N, Bhagat S, Muruganandan S, Kim R, Ho K, Pierce R, Kozhemyakina E, Lassar AB, Gamer L, Rosen V, Ionescu AM. Overexpression of transcription factor FoxA2 in the developing skeleton causes an enlargement of the cartilage hypertrophic zone, but it does not trigger ectopic differentiation in immature chondrocytes. Bone 2022; 160:116418. [PMID: 35398294 PMCID: PMC9133231 DOI: 10.1016/j.bone.2022.116418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 01/29/2023]
Abstract
We previously found that FoxA factors are necessary for chondrocyte differentiation. To investigate whether FoxA factors alone are sufficient to drive chondrocyte hypertrophy, we build a FoxA2 transgenic mouse in which FoxA2 cDNA is driven by a reiterated Tetracycline Response Element (TRE) and a minimal CMV promoter. This transgenic line was crossed with a col2CRE;Rosa26rtTA/+ mouse line to generate col2CRE;Rosa26rtTA/+;TgFoxA2+/- mice for inducible expression of FoxA2 in cartilage using doxycycline treatment. Ectopic expression of FoxA2 in the developing skeleton reveals skeletal defects and shorter skeletal elements in E17.5 mice. The chondro-osseous border was frequently mis-shaped in mutant mice, with small islands of col.10+ hypertrophic cells extending in the metaphyseal bone. Even though overexpression of FoxA2 causes an accumulation of hypertrophic chondrocytes, it did not trigger ectopic hypertrophy in the immature chondrocytes. This suggests that FoxA2 may need transcriptional co-factors (such as Runx2), whose expression is restricted to the hypertrophic zone, and absent in the immature chondrocytes. To investigate a potential FoxA2/Runx2 interaction in immature chondrocytes versus hypertrophic cells, we separated these two subpopulations by FACS to obtain CD24+CD200+ hypertrophic chondrocytes and CD24+CD200- immature chondrocytes and we ectopically expressed FoxA2 alone or in combination with Runx2 via lentiviral gene delivery. In CD24+CD200+ hypertrophic chondrocytes, FoxA2 enhanced the expression of chondrocyte hypertrophic markers collagen 10, MMP13, and alkaline phosphatase. In contrast, in the CD24+CD200- immature chondrocytes, neither FoxA2 nor Runx2 overexpression could induce ectopic expression of hypertrophic markers MMP13, alkaline phosphatase, or PTH/PTHrP receptor. Overall these findings mirror our in vivo data, and suggest that induction of chondrocyte hypertrophy by FoxA2 may require other factors in addition to Runx2 (i.e., Hif2α, MEF2C, or perhaps unknown factors), whose expression/activity is rate-limiting in immature chondrocytes.
Collapse
Affiliation(s)
- Nicole Bell
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, United States of America
| | - Sanket Bhagat
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, United States of America.
| | - Shanmugam Muruganandan
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, United States of America; Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA 02115, United States of America.
| | - Ryunhyung Kim
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, United States of America.
| | - Kailing Ho
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, United States of America.
| | - Rachel Pierce
- Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA 02115, United States of America.
| | - Elena Kozhemyakina
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Ave, Boston, MA 02115, United States of America.
| | - Andrew B Lassar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Ave, Boston, MA 02115, United States of America.
| | - Laura Gamer
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, United States of America.
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, United States of America.
| | - Andreia M Ionescu
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, United States of America; Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA 02115, United States of America.
| |
Collapse
|
41
|
Lei J, Chen S, Jing J, Guo T, Feng J, Ho T, Chai Y. Inhibiting Hh Signaling in Gli1 + Osteogenic Progenitors Alleviates TMJOA. J Dent Res 2022; 101:664-674. [PMID: 35045740 PMCID: PMC9124909 DOI: 10.1177/00220345211059079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023] Open
Abstract
The increased prevalence of temporomandibular joint osteoarthritis (TMJOA) in children and adolescents has drawn considerable attention as it may interfere with mandibular condyle growth, resulting in dento-maxillofacial deformities. However, treatments for osteoarthritis have been ineffective at restoring the damaged bone and cartilage structures due to poor understanding of the underlying degenerative mechanism. In this study, we demonstrate that Gli1+ cells residing in the subchondral bone contribute to bone formation and homeostasis in the mandibular condyle, identifying them as osteogenic progenitors in vivo. Furthermore, we show that, in a TMJOA mouse model, derivatives of Gli1+ cells undergo excessive expansion along with increased but uneven distribution of osteogenic differentiation in the subchondral bone, which leads to abnormal subchondral bone remodeling via Hedgehog (Hh) signaling activation and to the development of TMJOA. The selective pharmacological inhibition and specific genetic inhibition of Hh signaling in Gli1+ osteogenic progenitors result in improved subchondral bone microstructure, attenuated local immune inflammatory response in the subchondral bone, and reduced degeneration of the articular cartilage, providing in vivo functional evidence that targeting Hh signaling in Gli1+ osteogenic progenitors can modulate bone homeostasis in osteoarthritis and provide a potential approach for treating TMJOA.
Collapse
Affiliation(s)
- J. Lei
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Center for TMD & Orofacial Pain, Peking University School and Hospital of Stomatology, Beijing, China
| | - S. Chen
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
| | - J. Jing
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
| | - T. Guo
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
| | - J. Feng
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
| | - T.V. Ho
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
| | - Y. Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
42
|
The Emerging Role of Cell Transdifferentiation in Skeletal Development and Diseases. Int J Mol Sci 2022; 23:ijms23115974. [PMID: 35682655 PMCID: PMC9180549 DOI: 10.3390/ijms23115974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The vertebrate musculoskeletal system is known to be formed by mesenchymal stem cells condensing into tissue elements, which then differentiate into cartilage, bone, tendon/ligament, and muscle cells. These lineage-committed cells mature into end-stage differentiated cells, like hypertrophic chondrocytes and osteocytes, which are expected to expire and to be replaced by newly differentiated cells arising from the same lineage pathway. However, there is emerging evidence of the role of cell transdifferentiation in bone development and disease. Although the concept of cell transdifferentiation is not new, a breakthrough in cell lineage tracing allowed scientists to trace cell fates in vivo. Using this powerful tool, new theories have been established: (1) hypertrophic chondrocytes can transdifferentiate into bone cells during endochondral bone formation, fracture repair, and some bone diseases, and (2) tendon cells, beyond their conventional role in joint movement, directly participate in normal bone and cartilage formation, and ectopic ossification. The goal of this review is to obtain a better understanding of the key roles of cell transdifferentiation in skeletal development and diseases. We will first review the transdifferentiation of chondrocytes to bone cells during endochondral bone formation. Specifically, we will include the history of the debate on the fate of chondrocytes during bone formation, the key findings obtained in recent years on the critical factors and molecules that regulate this cell fate change, and the role of chondrocyte transdifferentiation in skeletal trauma and diseases. In addition, we will also summarize the latest discoveries on the novel roles of tendon cells and adipocytes on skeletal formation and diseases.
Collapse
|
43
|
Serowoky MA, Kuwahara ST, Liu S, Vakhshori V, Lieberman JR, Mariani FV. A murine model of large-scale bone regeneration reveals a selective requirement for Sonic Hedgehog. NPJ Regen Med 2022; 7:30. [PMID: 35581202 PMCID: PMC9114339 DOI: 10.1038/s41536-022-00225-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/25/2022] [Indexed: 11/21/2022] Open
Abstract
Building and maintaining skeletal tissue requires the activity of skeletal stem and progenitor cells (SSPCs). Following injury, local pools of these SSPCs become active and coordinate to build new cartilage and bone tissues. While recent studies have identified specific markers for these SSPCs, how they become activated in different injury contexts is not well-understood. Here, using a model of large-scale rib bone regeneration in mice, we demonstrate that the growth factor, Sonic Hedgehog (SHH), is an early and essential driver of large-scale bone healing. Shh expression is broadly upregulated in the first few days following rib bone resection, and conditional knockout of Shh at early but not late post-injury stages severely inhibits cartilage callus formation and later bone regeneration. Whereas Smoothened (Smo), a key transmembrane component of the Hh pathway, is required in Sox9+ lineage cells for rib regeneration, we find that Shh is required in a Prrx1-expressing, Sox9-negative mesenchymal population. Intriguingly, upregulation of Shh expression and requirements for Shh and Smo may be unique to large-scale injuries, as they are dispensable for both complete rib and femur fracture repair. In addition, single-cell RNA sequencing of callus tissue from animals with deficient Hedgehog signaling reveals a depletion of Cxcl12-expressing cells, which may indicate failed recruitment of Cxcl12-expressing SSPCs during the regenerative response. These results reveal a mechanism by which Shh expression in the local injury environment unleashes large-scale regenerative abilities in the murine rib.
Collapse
Affiliation(s)
- Maxwell A Serowoky
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, 1425 San Pablo Street, Los Angeles, CA, 90089, USA
| | - Stephanie T Kuwahara
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, 1425 San Pablo Street, Los Angeles, CA, 90089, USA
| | - Shuwan Liu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, 1425 San Pablo Street, Los Angeles, CA, 90089, USA
| | - Venus Vakhshori
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1520 San Pablo Street, Los Angeles, CA, 90089, USA
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1520 San Pablo Street, Los Angeles, CA, 90089, USA
| | - Francesca V Mariani
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, 1425 San Pablo Street, Los Angeles, CA, 90089, USA.
| |
Collapse
|
44
|
Guo Y, Wu D, Xu Q, Chen W. Inhibition of smoothened receptor by vismodegib leads to micrognathia during embryogenesis. Differentiation 2022; 125:27-34. [DOI: 10.1016/j.diff.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/03/2022] [Accepted: 04/07/2022] [Indexed: 11/03/2022]
|
45
|
Zhang H, Puviindran V, Nadesan P, Ding X, Shen L, Tang YJ, Tsushima H, Yahara Y, Ban GI, Zhang GF, Karner CM, Alman BA. Distinct Roles of Glutamine Metabolism in Benign and Malignant Cartilage Tumors With IDH Mutations. J Bone Miner Res 2022; 37:983-996. [PMID: 35220602 PMCID: PMC9314601 DOI: 10.1002/jbmr.4532] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/02/2022] [Accepted: 02/21/2022] [Indexed: 12/04/2022]
Abstract
Enchondromas and chondrosarcomas are common cartilage neoplasms that are either benign or malignant, respectively. The majority of these tumors harbor mutations in either IDH1 or IDH2. Glutamine metabolism has been implicated as a critical regulator of tumors with IDH mutations. Using genetic and pharmacological approaches, we demonstrated that glutaminase-mediated glutamine metabolism played distinct roles in enchondromas and chondrosarcomas with IDH1 or IDH2 mutations. Glutamine affected cell differentiation and viability in these tumors differently through different downstream metabolites. During murine enchondroma-like lesion development, glutamine-derived α-ketoglutarate promoted hypertrophic chondrocyte differentiation and regulated chondrocyte proliferation. Deletion of glutaminase in chondrocytes with Idh1 mutation increased the number and size of enchondroma-like lesions. In contrast, pharmacological inhibition of glutaminase in chondrosarcoma xenografts reduced overall tumor burden partially because glutamine-derived non-essential amino acids played an important role in preventing cell apoptosis. This study demonstrates that glutamine metabolism plays different roles in tumor initiation and cancer maintenance. Supplementation of α-ketoglutarate and inhibiting GLS may provide a therapeutic approach to suppress enchondroma and chondrosarcoma tumor growth, respectively. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Hongyuan Zhang
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | | | | | - Xiruo Ding
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - Leyao Shen
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuning J Tang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Yasuhito Yahara
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Ga I Ban
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.,Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, NC, USA
| | - Courtney M Karner
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin A Alman
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
46
|
Qu R, Gupta K, Dong D, Jiang Y, Landa B, Saez C, Strickland G, Levinsohn J, Weng PL, Taketo MM, Kluger Y, Myung P. Decomposing a deterministic path to mesenchymal niche formation by two intersecting morphogen gradients. Dev Cell 2022; 57:1053-1067.e5. [PMID: 35421372 PMCID: PMC9050909 DOI: 10.1016/j.devcel.2022.03.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/03/2022] [Accepted: 03/17/2022] [Indexed: 01/09/2023]
Abstract
Organ formation requires integrating signals to coordinate proliferation, specify cell fates, and shape tissue. Tracing these events and signals remains a challenge, as intermediate states across many critical transitions are unresolvable over real time and space. Here, we designed a unique computational approach to decompose a non-linear differentiation process into key components to resolve the signals and cell behaviors that drive a rapid transition, using the hair follicle dermal condensate as a model. Combining scRNA sequencing with genetic perturbation, we reveal that proliferative Dkk1+ progenitors transiently amplify to become quiescent dermal condensate cells by the mere spatiotemporal patterning of Wnt/β-catenin and SHH signaling gradients. Together, they deterministically coordinate a rapid transition from proliferation to quiescence, cell fate specification, and morphogenesis. Moreover, genetically repatterning these gradients reproduces these events autonomously in "slow motion" across more intermediates that resolve the process. This analysis unravels two morphogen gradients that intersect to coordinate events of organogenesis.
Collapse
Affiliation(s)
- Rihao Qu
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale University, New Haven, CT 06520, USA; Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Khusali Gupta
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Danni Dong
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | - Yiqun Jiang
- Department of Dermatology, Yale University, New Haven, CT 06520, USA; Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Boris Landa
- Applied Mathematics Program, Yale University, New Haven, CT 06511, USA
| | - Charles Saez
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | | | - Jonathan Levinsohn
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Pei-Lun Weng
- Department of Dermatology, Yale University, New Haven, CT 06520, USA
| | - M Mark Taketo
- Colon Cancer Project, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto 606-8501, Japan
| | - Yuval Kluger
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale University, New Haven, CT 06520, USA; Applied Mathematics Program, Yale University, New Haven, CT 06511, USA; Yale Cancer Center, New Haven, CT 06520, USA
| | - Peggy Myung
- Department of Pathology, Yale University, New Haven, CT 06520, USA; Department of Dermatology, Yale University, New Haven, CT 06520, USA; Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA; Yale Cancer Center, New Haven, CT 06520, USA; Yale Stem Cell Center, New Haven, CT 06520, USA.
| |
Collapse
|
47
|
Hedgehog signaling orchestrates cartilage-to-bone transition independently of Smoothened. Matrix Biol 2022; 110:76-90. [PMID: 35472633 DOI: 10.1016/j.matbio.2022.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 02/06/2022] [Accepted: 04/19/2022] [Indexed: 12/29/2022]
Abstract
Although recent lineage studies strongly support a chondrocyte-to-osteoblast differentiation continuum, the biological significance and molecular basis remain undetermined. In silico analysis at a single-cell level indicates a transient shutdown of Hedgehog-related transcriptome during simulated cartilage-to-bone transition. Prompted by this, we genetically induce gain- and loss-of function to probe the role of Hedgehog signaling in cartilage-to-bone transition. Ablating Smo in hypertrophic chondrocytes (HCs) does not result in any phenotypic outcome, whereas deleting Ptch1 in HCs leads to disrupted formation of primary spongiosa and actively proliferating HCs-derived osteogenic cells that contribute to bony bulges seen in adult mutant mice. In HCs-derived osteoblasts, constitutive activation of Hedgehog signaling blocks their further differentiation to osteocytes. Moreover, ablation of both Smo and Ptch1 in HCs reverses neither persistent Hedgehog signaling nor bone overgrowths. These results establish a functional contribution of extended chondrocyte lineage to bone homeostasis and diseases, governed by an unanticipated mode of regulation for Hedgehog signaling independently of Smo.
Collapse
|
48
|
Deng H, Xiao X, Chilufya MM, Qiao L, Lv Y, Guo Z, Lei J, Liu J, Zhao Y, Zhang J, Wang W, Han J, Qu C. Altered Expression of the Hedgehog Pathway Proteins BMP2, BMP4, SHH, and IHH Involved in Knee Cartilage Damage of Patients With Osteoarthritis and Kashin-Beck Disease. Cartilage 2022; 13:19476035221087706. [PMID: 35313742 PMCID: PMC9137319 DOI: 10.1177/19476035221087706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To investigate the expression of Hedgehog (HH) signaling pathway proteins in knee articular cartilage from Kashin-Beck disease (KBD) and osteoarthritis (OA) patients. METHODS Knee articular cartilage samples were collected from normal (N), OA, and KBD adults (aged 38-60 years) and divided into 3 groups with 6 subjects in each group. The localization of the HH pathway proteins bone morphogenetic protein 2 (BMP2), bone morphogenetic protein 4 (BMP4), Sonic hedgehog (SHH), and Indian hedgehog (IHH) was observed with the microscope after immunohistochemical (IHC) staining. Positive staining cell rates of each proteins were compared. RESULTS The strongest stainings of all proteins were observed in the middle zones of all 3 groups. The positive staining rates of BMP4 and IHH were significantly lower in the OA and KBD groups than those in the N group in all 3 zones. The positive staining rates of BMP2 and SHH tend to be lower in the OA and KBD groups than those in the N group in the deep zone, while higher in the OA and KBD groups than those in the N group in superficial and middle zones. CONCLUSIONS Altered expression of the HH pathway proteins BMP2, BMP4, SHH, and IHH was found in OA and KBD articular cartilage. There seemed to be a compensatory effect between SHH and IHH in cartilage damage. Further studies on the pathogenesis of OA and KBD may be carried out from these aspects in the future.
Collapse
Affiliation(s)
- Huan Deng
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China,Huan Deng, Xiang Xiao, and Mumba Mulutula Chilufya are co-first authors
| | - Xiang Xiao
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China,Huan Deng, Xiang Xiao, and Mumba Mulutula Chilufya are co-first authors
| | - Mumba Mulutula Chilufya
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China,Huan Deng, Xiang Xiao, and Mumba Mulutula Chilufya are co-first authors
| | - Lichun Qiao
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Yizhen Lv
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Ziwei Guo
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Jian Lei
- School of Public Health, Shanghai Medical College of Fudan University, Shanghai, P.R. China
| | - Jiaxin Liu
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Yan Zhao
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Jiaheng Zhang
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Wenyue Wang
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Jing Han
- School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China,Jing Han, School of Public Health, Health Science Center, Xi’an Jiaotong University, No. 76 West Yanta Road, Xi’an, Shaanxi 710061, P.R. China.
| | - Chengjuan Qu
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
49
|
Zhang L, Fu X, Ni L, Liu C, Zheng Y, You H, Li M, Xiu C, Zhang L, Gong T, Luo N, Zhang Z, He G, Hu S, Yang H, Chen D, Chen J. Hedgehog Signaling Controls Bone Homeostasis by Regulating Osteogenic/Adipogenic Fate of Skeletal Stem/Progenitor Cells in Mice. J Bone Miner Res 2022; 37:559-576. [PMID: 34870341 DOI: 10.1002/jbmr.4485] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 11/22/2021] [Accepted: 11/28/2021] [Indexed: 12/14/2022]
Abstract
Skeletal stem/progenitor cells (SSPCs) can differentiate into osteogenic or adipogenic lineage. The mechanism governing lineage allocation of SSPCs is still not completely understood. Hedgehog (Hh) signaling plays an essential role in specifying osteogenic fate of mesenchymal progenitors during embryogenesis. However, it is still unclear whether Hh signaling is required for lineage allocation of SSPCs in postnatal skeleton, and whether its dysregulation is related to age-related osteoporosis. Here, we demonstrated that Hh signaling was activated in metaphyseal SSPCs during osteogenic differentiation in the adult skeleton, and its activity decreased with aging. Inactivation of Hh signaling by genetic ablation of Smo, a key molecule in Hh signaling, in Osx-Cre-targeted SSPCs and hypertrophic chondrocytes led to decreased bone formation and increased bone marrow adiposity, two key pathological features of age-related osteoporosis. Moreover, we found that the bone-fat imbalance phenotype caused by Smo deletion mainly resulted from aberrant allocation of SSPCs toward adipogenic lineage at the expense of osteogenic differentiation, but not due to accelerated transdifferentiation of chondrocytes into adipocytes. Mechanistically, we found that Hh signaling regulated osteoblast versus adipocyte fate of SSPCs partly through upregulating Wnt signaling. Thus, our results indicate that Hh signaling regulates bone homeostasis and age-related osteoporosis by acting as a critical switch of cell fate decisions of Osx-Cre-targeted SSPCs in mice and suggest that Hh signaling may serve as a potential therapeutic target for the treatment of osteoporosis and other metabolic bone diseases. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Liwei Zhang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang, China.,Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Xuejie Fu
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Li Ni
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cunchang Liu
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Yixin Zheng
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Hongji You
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Meng Li
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Chunmei Xiu
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Lei Zhang
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Tingting Gong
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Na Luo
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Zunyi Zhang
- Key Laboratory of Mammalian Organogenesis and Regeneration, Hangzhou Normal University, Zhejiang, China
| | - Guangxu He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shijun Hu
- Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, China
| | - Huilin Yang
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China.,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Di Chen
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jianquan Chen
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China.,Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
50
|
Long JT, Leinroth A, Liao Y, Ren Y, Mirando AJ, Nguyen T, Guo W, Sharma D, Rouse D, Wu C, Cheah KSE, Karner CM, Hilton MJ. Hypertrophic chondrocytes serve as a reservoir for marrow-associated skeletal stem and progenitor cells, osteoblasts, and adipocytes during skeletal development. eLife 2022; 11:e76932. [PMID: 35179487 PMCID: PMC8893718 DOI: 10.7554/elife.76932] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/13/2022] [Indexed: 11/26/2022] Open
Abstract
Hypertrophic chondrocytes give rise to osteoblasts during skeletal development; however, the process by which these non-mitotic cells make this transition is not well understood. Prior studies have also suggested that skeletal stem and progenitor cells (SSPCs) localize to the surrounding periosteum and serve as a major source of marrow-associated SSPCs, osteoblasts, osteocytes, and adipocytes during skeletal development. To further understand the cell transition process by which hypertrophic chondrocytes contribute to osteoblasts or other marrow associated cells, we utilized inducible and constitutive hypertrophic chondrocyte lineage tracing and reporter mouse models (Col10a1CreERT2; Rosa26fs-tdTomato and Col10a1Cre; Rosa26fs-tdTomato) in combination with a PDGFRaH2B-GFP transgenic line, single-cell RNA-sequencing, bulk RNA-sequencing, immunofluorescence staining, and cell transplantation assays. Our data demonstrate that hypertrophic chondrocytes undergo a process of dedifferentiation to generate marrow-associated SSPCs that serve as a primary source of osteoblasts during skeletal development. These hypertrophic chondrocyte-derived SSPCs commit to a CXCL12-abundant reticular (CAR) cell phenotype during skeletal development and demonstrate unique abilities to recruit vasculature and promote bone marrow establishment, while also contributing to the adipogenic lineage.
Collapse
Affiliation(s)
- Jason T Long
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
| | - Abigail Leinroth
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
| | - Yihan Liao
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
- Department of Pharmacology and Cancer Biology, Duke University School of MedicineDurhamUnited States
| | - Yinshi Ren
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
| | - Anthony J Mirando
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
| | - Tuyet Nguyen
- Program of Developmental and Stem Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Wendi Guo
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
- Department of Pharmacology and Cancer Biology, Duke University School of MedicineDurhamUnited States
| | - Deepika Sharma
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
| | - Douglas Rouse
- Division of Laboratory Animal Resources, Duke University School of MedicineDurhamUnited States
| | - Colleen Wu
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
- Department of Pharmacology and Cancer Biology, Duke University School of MedicineDurhamUnited States
| | | | - Courtney M Karner
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
| | - Matthew J Hilton
- Department of Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Orthopaedic Surgery, Duke University School of MedicineDurhamUnited States
| |
Collapse
|