101
|
Yao JC, Oetjen KA, Wang T, Xu H, Abou-Ezzi G, Krambs JR, Uttarwar S, Duncavage EJ, Link DC. TGF-β signaling in myeloproliferative neoplasms contributes to myelofibrosis without disrupting the hematopoietic niche. J Clin Invest 2022; 132:154092. [PMID: 35439167 PMCID: PMC9151699 DOI: 10.1172/jci154092] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 04/14/2022] [Indexed: 12/31/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are associated with significant alterations in the bone marrow microenvironment that include decreased expression of key niche factors and myelofibrosis. Here, we explored the contribution of TGF-β to these alterations by abrogating TGF-β signaling in bone marrow mesenchymal stromal cells. Loss of TGF-β signaling in Osx-Cre-targeted MSCs prevented the development of myelofibrosis in both MPLW515L and Jak2V617F models of MPNs. In contrast, despite the absence of myelofibrosis, loss of TGF-β signaling in mesenchymal stromal cells did not rescue the defective hematopoietic niche induced by MPLW515L, as evidenced by decreased bone marrow cellularity, hematopoietic stem/progenitor cell number, and Cxcl12 and Kitlg expression, and the presence of splenic extramedullary hematopoiesis. Induction of myelofibrosis by MPLW515L was intact in Osx-Cre Smad4fl/fl recipients, demonstrating that SMAD4-independent TGF-β signaling mediates the myelofibrosis phenotype. Indeed, treatment with a c-Jun N-terminal kinase (JNK) inhibitor prevented the development of myelofibrosis induced by MPLW515L. Together, these data show that JNK-dependent TGF-β signaling in mesenchymal stromal cells is responsible for the development of myelofibrosis but not hematopoietic niche disruption in MPNs, suggesting that the signals that regulate niche gene expression in bone marrow mesenchymal stromal cells are distinct from those that induce a fibrogenic program.
Collapse
Affiliation(s)
- Juo-Chin Yao
- Division of Oncology, Department of Medicine and
| | | | - Tianjiao Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Haoliang Xu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | - Eric J. Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
102
|
Whole Aspect of Runx2 Functions in Skeletal Development. Int J Mol Sci 2022; 23:ijms23105776. [PMID: 35628587 PMCID: PMC9144571 DOI: 10.3390/ijms23105776] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022] Open
Abstract
Runt-related transcription factor 2 (Runx2) is a fundamental transcription factor for bone development. In endochondral ossification, Runx2 induces chondrocyte maturation, enhances chondrocyte proliferation through Indian hedgehog (Ihh) induction, and induces the expression of vascular endothelial growth factor A (Vegfa), secreted phosphoprotein 1 (Spp1), integrin-binding sialoprotein (Ibsp), and matrix metallopeptidase 13 (Mmp13) in the terminal hypertrophic chondrocytes. Runx2 inhibits the apoptosis of the terminal hypertrophic chondrocytes and induces their transdifferentiation into osteoblasts and osteoblast progenitors. The transdifferentiation is required for trabecular bone formation during embryonic and newborn stages but is dispensable for acquiring normal bone mass in young and adult mice. Runx2 enhances the proliferation of osteoblast progenitors and induces their commitment to osteoblast lineage cells through the direct regulation of the expressions of a hedgehog, fibroblast growth factor (Fgf), Wnt, and parathyroid hormone-like hormone (Pthlh) signaling pathway genes and distal-less homeobox 5 (Dlx5), which all regulate Runx2 expression and/or protein activity. Runx2, Sp7, and Wnt signaling further induce osteoblast differentiation. In immature osteoblasts, Runx2 regulates the expression of bone matrix protein genes, including Col1a1, Col1a2, Spp1, Ibsp, and bone gamma carboxyglutamate protein (Bglap)/Bglap2, and induces osteoblast maturation. Osteocalcin (Bglap/Bglap2) is required for the alignment of apatite crystals parallel to the collagen fibers; however, it does not physiologically work as a hormone that regulates glucose metabolism, testosterone synthesis, or muscle mass. Thus, Runx2 exerts multiple functions essential for skeletal development.
Collapse
|
103
|
Hojo H, Ohba S. Sp7 Action in the Skeleton: Its Mode of Action, Functions, and Relevance to Skeletal Diseases. Int J Mol Sci 2022; 23:5647. [PMID: 35628456 PMCID: PMC9143072 DOI: 10.3390/ijms23105647] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/14/2022] [Accepted: 05/16/2022] [Indexed: 02/01/2023] Open
Abstract
Osteoblast differentiation is a tightly regulated process in which key transcription factors (TFs) and their target genes constitute gene regulatory networks (GRNs) under the control of osteogenic signaling pathways. Among these TFs, Sp7 works as an osteoblast determinant critical for osteoblast differentiation. Following the identification of Sp7 and a large number of its functional studies, recent genome-scale analyses have made a major contribution to the identification of a "non-canonical" mode of Sp7 action as well as "canonical" ones. The analyses have not only confirmed known Sp7 targets but have also uncovered its additional targets and upstream factors. In addition, biochemical analyses have demonstrated that Sp7 actions are regulated by chemical modifications and protein-protein interaction with other transcriptional regulators. Sp7 is also involved in chondrocyte differentiation and osteocyte biology as well as postnatal bone metabolism. The critical role of SP7 in the skeleton is supported by its relevance to human skeletal diseases. This review aims to overview the Sp7 actions in skeletal development and maintenance, particularly focusing on recent advances in our understanding of how Sp7 functions in the skeleton under physiological and pathological conditions.
Collapse
Affiliation(s)
- Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan;
| | - Shinsuke Ohba
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
- Department of Oral Anatomy and Developmental Biology, Osaka University Graduate School of Dentistry, Osaka 565-0871, Japan
| |
Collapse
|
104
|
Sedes L, Wondimu E, Crockett B, Hansen J, Cantalupo A, Asano K, Iyengar R, Rifkin DB, Smaldone S, Ramirez F. Fibrillin-1 deficiency in the outer perichondrium causes longitudinal bone overgrowth in mice with Marfan syndrome. Hum Mol Genet 2022; 31:3281-3289. [PMID: 35567544 PMCID: PMC9523555 DOI: 10.1093/hmg/ddac107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/19/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
A disproportionate tall stature is the most evident manifestation in Marfan syndrome (MFS), a multisystem condition caused by mutations in the extracellular protein and TGFβ modulator, fibrillin-1. Unlike cardiovascular manifestations, there has been little effort devoted to unravel the molecular mechanism responsible for long bone overgrowth in MFS. By combining the Cre-LoxP recombination system with metatarsal bone cultures, here we identify the outer layer of the perichondrium as the tissue responsible for long bone overgrowth in MFS mice. Analyses of differentially expressed genes in the fibrillin-1 deficient perichondrium predicted that loss of TGFβ signaling may influence chondrogenesis in the neighboring epiphyseal growth plate (GP). Immunohistochemistry revealed that fibrillin-1 deficiency in the outer perichondrium is associated with decreased accumulation of latent TGFβ-binding proteins (LTBPs)-3 and - 4, and reduced levels of phosphorylated (activated) Smad2. Consistent with these findings, mutant metatarsal bones grown in vitro were longer and released less TGFβ than the wild type counterparts. Moreover, addition of recombinant TGFβ1 normalized linear growth of mutant metatarsal bones. We conclude that longitudinal bone overgrowth in MFS is accounted for by diminished sequestration of LTBP-3 and LTBP-4 into the fibrillin-1 deficient matrix of the outer perichondrium, which results in less TGFβ signaling locally and improper GP differentiation distally.
Collapse
Affiliation(s)
- Lauriane Sedes
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| | - Elisa Wondimu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| | - Brittany Crockett
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| | - Jens Hansen
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| | - Anna Cantalupo
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| | - Keiichi Asano
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| | - Daniel B Rifkin
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Silvia Smaldone
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| | - Francesco Ramirez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10021
| |
Collapse
|
105
|
Omatsu Y, Aiba S, Maeta T, Higaki K, Aoki K, Watanabe H, Kondoh G, Nishimura R, Takeda S, Chung UI, Nagasawa T. Runx1 and Runx2 inhibit fibrotic conversion of cellular niches for hematopoietic stem cells. Nat Commun 2022; 13:2654. [PMID: 35551452 PMCID: PMC9098511 DOI: 10.1038/s41467-022-30266-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/08/2022] [Indexed: 01/01/2023] Open
Abstract
In bone marrow, special microenvironments, known as niches, are essential for the maintenance of hematopoietic stem cells (HSCs). A population of mesenchymal stem cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells or leptin receptor-expressing cells are the major cellular component of HSC niches. The molecular regulation of HSC niche properties is not fully understood. The role of Runx transcription factors, Runx1 and Runx2 in HSC cellular niches remains unclear. Here we show that Runx1 is predominantly expressed in CAR cells and that mice lacking both Runx1 and Runx2 in CAR cells display an increase in fibrosis and bone formation with markedly reduced hematopoietic stem and progenitor cells in bone marrow. In vitro, Runx1 is induced by the transcription factor Foxc1 and decreases fibrotic gene expression in CAR cells. Thus, HSC cellular niches require Runx1 or Runx2 to prevent their fibrotic conversion and maintain HSCs and hematopoiesis in adults. The transcription factors, Runx1 and Runx2 are critical embryonically for generation of HSCs and osteoblasts, respectively. Here the authors show that adult mice lacking Runx1 and Runx2 in HSC-supporting CAR cells displayed an increase in fibrosis with reduced HSCs in bone marrow.
Collapse
Affiliation(s)
- Yoshiki Omatsu
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shota Aiba
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Tomonori Maeta
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kei Higaki
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kazunari Aoki
- Laboratory of Stem Cell Genetics, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hitomi Watanabe
- Laboratory of Animal Experiments for Regeneration, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Gen Kondoh
- Laboratory of Animal Experiments for Regeneration, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shu Takeda
- Endocrinology Division, Toranomon Hospital, Minato-ku, Tokyo, 105-8470, Japan
| | - Ung-Il Chung
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan. .,Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan. .,Laboratory of Stem Cell Biology and Developmental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
106
|
Ren Y, Yang M, Wang X, Xu B, Xu Z, Su B. ELAV-like RNA binding protein 1 regulates osteogenesis in diabetic osteoporosis: Involvement of divalent metal transporter 1. Mol Cell Endocrinol 2022; 546:111559. [PMID: 35051552 DOI: 10.1016/j.mce.2022.111559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/23/2021] [Accepted: 01/10/2022] [Indexed: 12/18/2022]
Abstract
Diabetic osteoporosis (DOP) is a complication of diabetes mellitus (DM) and occurs due to alterations in bone metabolism under hyperglycemic condition. ELAV-like RNA binding protein 1 (ELAVL1) is abnormally up-regulated in diabetes-related diseases. Bioinformatics prediction indicates that divalent metal transporter 1 (DMT1) is a potential target of ELAVL1. To explore the role of ELAVL1 and the involvement of ELAVL1/DMT1 axis in DOP, we established a mouse model of DM by administration of high-fat diet and intraperitoneal injection with streptozotocin (STZ). The expression of ELAVL1 and DMT1 was increased in the bone tissues of DM mice. Knockdown of ELAVL1 reduced iron level and oxidative stress, promoted osteogensis, and prevented bone mass loss, thus mitigating DOP in DM mice. In vitro, mouse pre-osteoblast MC3T3-E1 cells were treated with high glucose (25 mM) and ferric ammonium citrate (FAC, 200 μM). The inhibitory effects of ELAVL1 knockdown on iron accumulation and oxidative stress were evidenced in MC3T3-E1 cells. Knockdown of ELAVL1 enhanced osteoblast viability, differentiation and mineralization. Notably, the expression of DMT1 was positively correlated with that of ELAVL1 in vivo and in vitro. Overexpression of DMT1 abolished the effect of ELAVL1 knockdown on the behaviors of MC3T3-E1 cells, suggesting that ELAVL1 might function through regulating DMT1. In conclusion, knockdown of ELAVL1 likely alleviated DOP by inhibiting iron overload and oxidative stress and promoting osteogenesis, and DMT1 might be involved in this process. These findings provide insights into the pathogenesis of DOP and suggest a potential therapeutic target for DOP treatment.
Collapse
Affiliation(s)
- Yuanfei Ren
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China; The First Department of Hand and Foot Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China
| | - Maowei Yang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Xindong Wang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Buxuan Xu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zerong Xu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bo Su
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
107
|
Liu C, Xiong Q, Li Q, Lin W, Jiang S, Zhang D, Wang Y, Duan X, Gong P, Kang N. CHD7 regulates bone-fat balance by suppressing PPAR-γ signaling. Nat Commun 2022; 13:1989. [PMID: 35418650 PMCID: PMC9007978 DOI: 10.1038/s41467-022-29633-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 03/23/2022] [Indexed: 02/08/2023] Open
Abstract
Chromodomain helicase DNA-binding protein 7 (CHD7), an ATP-dependent eukaryotic chromatin remodeling enzyme, is essential for the development of organs. The mutation of CHD7 is the main cause of CHARGE syndrome, but its function and mechanism in skeletal system remain unclear. Here, we show conditional knockout of Chd7 in bone marrow mesenchymal stem cells (MSCs) and preosteoblasts leads to a pathological phenotype manifested as low bone mass and severely high marrow adiposity. Mechanistically, we identify enhancement of the peroxisome proliferator-activated receptor (PPAR) signaling in Chd7-deficient MSCs. Loss of Chd7 reduces the restriction of PPAR-γ and then PPAR-γ associates with trimethylated histone H3 at lysine 4 (H3K4me3), which subsequently activates the transcription of downstream adipogenic genes and disrupts the balance between osteogenic and adipogenic differentiation. Our data illustrate the pathological manifestations of Chd7 mutation in MSCs and reveal an epigenetic mechanism in skeletal health and diseases. CHD7 is chromatin remodeler and mutations of CHD7 are the main cause of CHARGE syndrome. Here the authors show that conditional knockout of Chd7 in bone marrow mesenchymal stem cells (MSCs) and pre-osteoblasts leads to a skeletal system development disorder in mice and upregulated PPAR signaling, disrupting the balance between osteogenic and adipogenic differentiation.
Collapse
Affiliation(s)
- Caojie Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Qiuchan Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Qiwen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Shuang Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Danting Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yuan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Xiaobo Duan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.
| | - Ning Kang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
108
|
Sadeghzadeh H, Mehdipour A, Dianat-Moghadam H, Salehi R, Khoshfetrat AB, Hassani A, Mohammadnejad D. PCL/Col I-based magnetic nanocomposite scaffold provides an osteoinductive environment for ADSCs in osteogenic cues-free media conditions. Stem Cell Res Ther 2022; 13:143. [PMID: 35379318 PMCID: PMC8981929 DOI: 10.1186/s13287-022-02816-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/19/2022] [Indexed: 12/20/2022] Open
Abstract
Background The bone tissue engineering (BTE) approach has been introduced as an alternative to conventional treatments for large non-healing bone defects. Magnetism promotes stem cells' adherence to biocompatible scaffolds toward osteoblast differentiation. Furthermore, osteogenic differentiation media are expensive and any changes in its composition affect stem cells differentiation. Moreover, media growth factors possess a short half-life resulting in the rapid loss of their functions in vivo. With the above in mind, we fabricated a multilayered nanocomposite scaffold containing the wild type of Type I collagen (Col I) with endogenous magnetic property to promote osteogenesis in rat ADSCs with the minimum requirement of osteogenic differentiation medium.
Methods Fe3O4 NPs were synthesized by co-precipitation method and characterized using SEM, VSM, and FTIR. Then, a PCL/Col I nanocomposite scaffold entrapping Fe3O4 NPs was fabricated by electrospinning and characterized using SEM, TEM, AFM, VSM, Contact Angle, tensile stretching, and FTIR. ADSCs were isolated from rat adipose tissue and identified by flow cytometry. ADSCs were loaded onto PCL/Col I and PCL/Col I/Fe3O4-scaffolds for 1–3 weeks with/without osteogenic media conditions. The cell viability, cell adhesion, and osteogenic differentiation were evaluated using MTT assay, SEM, DAPI staining, ALP/ARS staining, RT-PCR, and western blotting, respectively. Results SEM, VSM, and FTIR results indicated that Fe3O4 was synthesized in nano-sized (15–30 nm) particles with spherical-shaped morphology and superparamagnetic properties with approved chemical structure as FTIR revealed. According to SEM images, the fabricated magnetic scaffolds consisted of nanofiber (500–700 nm). TEM images have shown the Fe3O4 NPs entrapped in the scaffold's fiber without bead formation. FTIR spectra analysis confirmed the maintenance of the natural structure of Col I, PCL, and Fe3O4 upon electrospinning. AFM data have shown that MNPs incorporation introduced stripe-like topography to nanofibers, while the depth of the grooves has decreased from 800 to 500 nm. Flow cytometry confirmed the phenotype of ADSCs according to their surface markers (i.e., CD29 and CD105). Additionally, Fe3O4 NP improved nanocomposite scaffold strength, wettability, porosity, biocompatibility and also facilitates the ALP activity, calcium-mineralization. Finally, magnetic nanocomposite scaffolds upregulated osteogenic-related genes or proteins’ expression (e.g., Col I, Runx2, OCN, ON, BMP2) in seeded ADSCs with/without osteo-differentiation media conditions. Conclusions Together, these results indicate that Fe3O4 NPs within the natural structure of Col I increase osteogenic differentiation in osteogenic cues-free media conditions. This effect could be translated in vivo toward bone defects healing. These findings support the use of natural ECM materials alongside magnetic particles as composite scaffolds to achieve their full therapeutic potential in BTE treatments. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Hadi Sadeghzadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mehdipour
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Ayla Hassani
- Chemical Engineering Faculty, Sahand University of Technology, 51335-1996, Tabriz, Iran
| | - Daryush Mohammadnejad
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
109
|
Galán-Díez M, Borot F, Ali AM, Zhao J, Gil-Iturbe E, Shan X, Luo N, Liu Y, Huang XP, Bisikirska B, Labella R, Kurland I, Roth BL, Quick M, Mukherjee S, Rabadán R, Carroll M, Raza A, Kousteni S. Subversion of Serotonin Receptor Signaling in Osteoblasts by Kynurenine Drives Acute Myeloid Leukemia. Cancer Discov 2022; 12:1106-1127. [PMID: 35046097 PMCID: PMC8983599 DOI: 10.1158/2159-8290.cd-21-0692] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/08/2021] [Accepted: 01/04/2022] [Indexed: 01/09/2023]
Abstract
Remodeling of the microenvironment by tumor cells can activate pathways that favor cancer growth. Molecular delineation and targeting of such malignant-cell nonautonomous pathways may help overcome resistance to targeted therapies. Herein we leverage genetic mouse models, patient-derived xenografts, and patient samples to show that acute myeloid leukemia (AML) exploits peripheral serotonin signaling to remodel the endosteal niche to its advantage. AML progression requires the presence of serotonin receptor 1B (HTR1B) in osteoblasts and is driven by AML-secreted kynurenine, which acts as an oncometabolite and HTR1B ligand. AML cells utilize kynurenine to induce a proinflammatory state in osteoblasts that, through the acute-phase protein serum amyloid A (SAA), acts in a positive feedback loop on leukemia cells by increasing expression of IDO1-the rate-limiting enzyme for kynurenine synthesis-thereby enabling AML progression. This leukemia-osteoblast cross-talk, conferred by the kynurenine-HTR1B-SAA-IDO1 axis, could be exploited as a niche-focused therapeutic approach against AML, opening new avenues for cancer treatment. SIGNIFICANCE AML remains recalcitrant to treatments due to the emergence of resistant clones. We show a leukemia-cell nonautonomous progression mechanism that involves activation of a kynurenine-HTR1B-SAA-IDO1 axis between AML cells and osteoblasts. Targeting the niche by interrupting this axis can be pharmacologically harnessed to hamper AML progression and overcome therapy resistance. This article is highlighted in the In This Issue feature, p. 873.
Collapse
Affiliation(s)
- Marta Galán-Díez
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York.,Corresponding Authors: Stavroula Kousteni, Phone: 212-305-2068; E-mail: ; and Marta Galán-Díez, Department of Physiology and Cellular Biophysics, Columbia University, 650 W. 168th Street, New York, NY 10032. Phone: 212-305-2481; E-mail:
| | - Florence Borot
- Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, New York
| | - Abdullah Mahmood Ali
- Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, New York.,Myelodysplastic Syndromes Center, Columbia University, New York, New York
| | - Junfei Zhao
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, New York.,Edward P. Evans Center for Myelodysplastic Syndromes at Columbia University, New York, New York
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University, New York, New York
| | - Xiaochuan Shan
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Na Luo
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York
| | - Yongfeng Liu
- NIMH Psychoactive Drug Screening Program, Department of Pharmacology, University of North Carolina Chapel Hill Medical School, Chapel Hill, North Carolina
| | - Xi-Ping Huang
- NIMH Psychoactive Drug Screening Program, Department of Pharmacology, University of North Carolina Chapel Hill Medical School, Chapel Hill, North Carolina
| | - Brygida Bisikirska
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York
| | - Rossella Labella
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York
| | - Irwin Kurland
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Bryan L. Roth
- NIMH Psychoactive Drug Screening Program, Department of Pharmacology, University of North Carolina Chapel Hill Medical School, Chapel Hill, North Carolina.,Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Matthias Quick
- Department of Psychiatry, Columbia University, New York, New York.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York
| | - Siddhartha Mukherjee
- Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, New York.,Myelodysplastic Syndromes Center, Columbia University, New York, New York.,Edward P. Evans Center for Myelodysplastic Syndromes at Columbia University, New York, New York
| | - Raul Rabadán
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, New York.,Department of Biomedical Informatics, Columbia University, New York, New York
| | - Martin Carroll
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Azra Raza
- Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, New York.,Myelodysplastic Syndromes Center, Columbia University, New York, New York.,Edward P. Evans Center for Myelodysplastic Syndromes at Columbia University, New York, New York
| | - Stavroula Kousteni
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York.,Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, New York.,Edward P. Evans Center for Myelodysplastic Syndromes at Columbia University, New York, New York.,Columbia Stem Cell Initiative, Columbia University, New York, New York.,Corresponding Authors: Stavroula Kousteni, Phone: 212-305-2068; E-mail: ; and Marta Galán-Díez, Department of Physiology and Cellular Biophysics, Columbia University, 650 W. 168th Street, New York, NY 10032. Phone: 212-305-2481; E-mail:
| |
Collapse
|
110
|
Palmieri M, Joseph TE, O’Brien CA, Gomez-Acevedo H, Manolagas SC, Ambrogini E. Deletion of the scavenger receptor Scarb1 in osteoblast progenitors does not affect bone mass. PLoS One 2022; 17:e0265893. [PMID: 35349600 PMCID: PMC8963559 DOI: 10.1371/journal.pone.0265893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/09/2022] [Indexed: 11/18/2022] Open
Abstract
The scavenger receptor class B member 1 (SR-B1 or Scarb1) is a cell surface receptor for high density lipoproteins. It also binds oxidized low density lipoproteins and phosphocholine-containing oxidized phospholipids (PC-OxPL), which adversely affect bone homeostasis. Overexpression of a single chain form of the antigen-binding domain of E06 IgM-a natural antibody that recognizes PC-OxPL-increases trabecular and cortical bone mass in female and male mice by stimulating bone formation. We have previously reported that Scarb1 is the most abundant scavenger receptor for PC-OxPL in calvaria-derived osteoblastic cells. Additionally, bone marrow- and calvaria-derived osteoblasts from Scarb1 knockout mice (Scarb1 KO) are protected from the pro-apoptotic and anti-differentiating effects of OxPL. Previous skeletal analysis of Scarb1 KO mice has produced contradictory results, with some studies reporting elevated bone mass but another study reporting low bone mass. To clarify the role of Scarb1 in osteoblasts, we deleted Scarb1 specifically in cells of the osteoblast lineage using Osx1-Cre transgenic mice. We observed no difference in bone mineral density measured by DXA in either female or male Osx1-Cre;Scarb1fl/fl mice compared to wild type (WT), Osx1-Cre, or Scarb1fl/fl littermate controls. Additionally, microCT analysis of 6-month-old females and 7-month-old males did not detect any difference in trabecular or cortical bone mass between genotypes. These results indicate that expression of Scarb1 in cells of the osteoblast lineage does not play an important role in bone homeostasis and, therefore, it is not essential for the effects of PC-OxPL on these cells.
Collapse
Affiliation(s)
- Michela Palmieri
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| | - Teenamol E. Joseph
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| | - Charles A. O’Brien
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| | - Horacio Gomez-Acevedo
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Stavros C. Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
| |
Collapse
|
111
|
Davis JL, Pokhrel NK, Cox L, Rohatgi N, Faccio R, Veis DJ. Conditional loss of IKKα in Osterix + cells has no effect on bone but leads to age-related loss of peripheral fat. Sci Rep 2022; 12:4915. [PMID: 35318397 PMCID: PMC8940989 DOI: 10.1038/s41598-022-08914-6] [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: 11/30/2021] [Accepted: 03/10/2022] [Indexed: 11/09/2022] Open
Abstract
NF-κB has been reported to both promote and inhibit bone formation. To explore its role in osteolineage cells, we conditionally deleted IKKα, an upstream kinase required for non-canonical NF-κB activation, using Osterix (Osx)-Cre. Surprisingly, we found no effect on either cancellous or cortical bone, even following mechanical loading. However, we noted that IKKα conditional knockout (cKO) mice began to lose body weight after 6 months of age with severe reductions in fat mass and lower adipocyte size in geriatric animals. qPCR analysis of adipogenic markers in fat pads of cKO mice indicated no difference in early differentiation, but instead markedly lower leptin with age. We challenged young mice with a high fat diet finding that cKO mice gained less weight and showed improved glucose metabolism. Low levels of recombination at the IKKα locus were detected in fat pads isolated from old cKO mice. To determine whether recombination occurs in adipocytes, we examined fat pads in Osx-Cre;TdT reporter mice; these showed increasing Osx-Cre-mediated expression in peripheral adipocytes from 6 weeks to 18 months. Since Osx-Cre drives recombination in peripheral adipocytes with age, we conclude that fat loss in cKO mice is most likely caused by progressive deficits of IKKα in adipocytes.
Collapse
Affiliation(s)
- Jennifer L Davis
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nitin Kumar Pokhrel
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Linda Cox
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nidhi Rohatgi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Roberta Faccio
- Musculoskeletal Research Center, Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Shriners Hospitals for Children, St. Louis, MO, 63110, USA
| | - Deborah J Veis
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Shriners Hospitals for Children, St. Louis, MO, 63110, USA.
| |
Collapse
|
112
|
Shen L, Yu Y, Zhou Y, Pruett-Miller SM, Zhang GF, Karner CM. SLC38A2 provides proline to fulfil unique synthetic demands arising during osteoblast differentiation and bone formation. eLife 2022; 11:76963. [PMID: 35261338 PMCID: PMC9007586 DOI: 10.7554/elife.76963] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular differentiation is associated with the acquisition of a unique protein signature which is essential to attain the ultimate cellular function and activity of the differentiated cell. This is predicted to result in unique biosynthetic demands that arise during differentiation. Using a bioinformatic approach, we discovered osteoblast differentiation is associated with increased demand for the amino acid proline. When compared to other differentiated cells, osteoblast-associated proteins including RUNX2, OSX, OCN and COL1A1 are significantly enriched in proline. Using a genetic and metabolomic approach, we demonstrate that the neutral amino acid transporter SLC38A2 acts cell autonomously to provide proline to facilitate the efficient synthesis of proline-rich osteoblast proteins. Genetic ablation of SLC38A2 in osteoblasts limits both osteoblast differentiation and bone formation in mice. Mechanistically, proline is primarily incorporated into nascent protein with little metabolism observed. Collectively, these data highlight a requirement for proline in fulfilling the unique biosynthetic requirements that arise during osteoblast differentiation and bone formation.
Collapse
Affiliation(s)
- Leyao Shen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yilin Yu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yunji Zhou
- Department of Biostatistics and Bioinformatics, Duke University, Durham, United States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis, United States
| | - Guo-Fang Zhang
- Sarah W Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, United States
| | - Courtney M Karner
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
113
|
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: 24] [Impact Index Per Article: 8.0] [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
|
114
|
Peredo N, Valle-Tenney R, Melis S, Mesnieres M, Nefyodova E, Maes C. Visualization and quantification of the stromal-vascular compartment in fetal or adult mouse bones: From sampling to high-resolution 3D image analysis. STAR Protoc 2022. [DOI: 10.1016/j.xpro.2022.101222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
115
|
Song L, Li H, Liu Y, Zhang X, Wen Y, Zhang K, Zhang M. Postnatal deletion of β-catenin in preosteoblasts regulates global energy metabolism through increasing bone resorption and adipose tissue fibrosis. Bone 2022; 156:116320. [PMID: 34973494 DOI: 10.1016/j.bone.2021.116320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 12/12/2021] [Accepted: 12/22/2021] [Indexed: 11/02/2022]
Abstract
Many studies revealed bone can regulate global energy metabolism and our previous study also showed that Wnt/β-catenin pathway is involved in this process. To better understand the participation of canonical Wnt pathway in energy metabolism, we examined the β-catenin knock-out (β-cat KO) mice by crossing the osterix-cre transgenic mice with β-cateninflox/flox mice. First, we identified that postnatal deletion of β-catenin in preosteoblasts led to decreased fat mass and increased energy expenditure in mice. Osteoprotegerin administration largely rescued the decreased fat mass and partly normalized the energy expenditure accompanied by the inhibition of bone resorption. Anti-resorption with alendronate or RANKL-antibody could also partly rescued the decreased bone mass, decreased fat mass and increased energy expenditure in β-cat KO mice. We further found that the adipose cells in the inguinal fat tissue were smaller and the extracellular matrix components around adipocytes accumulated more in β-cat KO mice than their controls by histomorphology. Gene analysis by RT-PCR showed that the expression of collagen VI is 4.8 folds in adipose tissue of the β-cat KO mice compared with the control mice. We further detected the expression of cytokines which were related to fibrosis and the data showed that the level of TGF-beta1 was elevated in both of bone marrow serum and adipose tissue derived from the β-cat KO mice. After administration of TGF-beta1 neutralizing antibody, the impaired energy metabolism was partly rescued in β-cat KO mice. Besides, anti-resorption treatment and TGF-beta1 antibody could partly suppress the increased expression of genes related to fat tissue fibrosis. These results indicate that the abnormal global energy metabolism in β-cat KO mice may be attributed to increasing bone resorption and adipose tissue fibrosis.
Collapse
Affiliation(s)
- Lige Song
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Huijuan Li
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yichen Liu
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiaoya Zhang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yuhua Wen
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Keqin Zhang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Institute of Osteoporosis and Metabolic Bone Diseases, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Mingzhu Zhang
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 1 Dongjiao Minxiang, West Area, Beijing, 100730, China.
| |
Collapse
|
116
|
Xu R, Liu Y, Zhou Y, Lin W, Yuan Q, Zhou X, Yang Y. Gnas Loss Causes Chondrocyte Fate Conversion in Cranial Suture Formation. J Dent Res 2022; 101:931-941. [PMID: 35220829 DOI: 10.1177/00220345221075215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Calvaria development is distinct from limb formation. Craniosynostosis is a skull deformity characterized by premature cranial suture fusion due to the loss of the GNAS gene and, consequently, its encoded protein Gαs. This birth defect requires surgery, with potential lethal consequences. So far, hardly any early-stage nonsurgical interventions for GNAS loss-related craniosynostosis are available. Here, we investigated the role of the Gnas gene in mice in guarding the distinctiveness of intramembranous ossification and how loss of Gnas triggered endochondral-like ossification within the cranial sutures. Single-cell RNA sequencing (scRNA-seq) of normal neonatal mice cranial suture chondrocytes showed a Hedgehog (Hh) inactivation pattern, which was associated with Gαs signaling activation. Loss of Gnas evoked chondrocyte-to-osteoblast fate conversion and resulted in cartilage heterotopic ossification (HO) within cranial sutures and fontanels of the mouse model, leading to a skull deformity resembling craniosynostosis in patients with loss of GNAS. Activation of ectopic Hh signaling within cranial chondrocytes stimulated the conversion of cell identity through a hypertrophy-like stage, which shared features of endochondral ossification in vivo. Reduction of Gli transcription activity by crossing with a loss-of-function Gli2 allele or injecting GLI1/2 antagonist hindered the progression of cartilage HO in neonatal stage mice. Our study uncovered the role of Gαs in maintaining cranial chondrocyte identity during neonatal calvaria development in mice and how reduction of Hh signaling could be a nonsurgical intervention to reduce skull deformity in craniosynostosis due to loss of GNAS.
Collapse
Affiliation(s)
- R. Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y. Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA, USA
| | - Y. Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA, USA
| | - W. Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q. Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X. Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y. Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA, USA
| |
Collapse
|
117
|
Stanwick M, Barkley C, Serra R, Kruggel A, Webb A, Zhao Y, Pietrzak M, Ashman C, Staats A, Shahid S, Peters SB. Tgfbr2 in Dental Pulp Cells Guides Neurite Outgrowth in Developing Teeth. Front Cell Dev Biol 2022; 10:834815. [PMID: 35265620 PMCID: PMC8901236 DOI: 10.3389/fcell.2022.834815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor β (TGFβ) plays an important role in tooth morphogenesis and mineralization. During postnatal development, the dental pulp (DP) mesenchyme secretes neurotrophic factors that guide trigeminal nerve fibers into and throughout the DP. This process is tightly linked with dentin formation and mineralization. Our laboratory established a mouse model in which Tgfbr2 was conditionally deleted in DP mesenchyme using an Osterix promoter-driven Cre recombinase (Tgfbr2 cko ). These mice survived postnatally with significant defects in bones and teeth, including reduced mineralization and short roots. Hematoxylin and eosin staining revealed reduced axon-like structures in the mutant mice. Reporter imaging demonstrated that Osterix-Cre activity within the tooth was active in the DP and derivatives, but not in neuronal afferents. Immunofluorescence staining for β3 tubulin (neuronal marker) was performed on serial cryosections from control and mutant molars on postnatal days 7 and 24 (P7, P24). Confocal imaging and pixel quantification demonstrated reduced innervation in Tgfbr2 cko first molars at both stages compared to controls, indicating that signals necessary to promote neurite outgrowth were disrupted by Tgfbr2 deletion. We performed mRNA-Sequence (RNA-Seq) and gene onotology analyses using RNA from the DP of P7 control and mutant mice to investigate the pathways involved in Tgfbr2-mediated tooth development. These analyses identified downregulation of several mineralization-related and neuronal genes in the Tgfbr2 cko DP compared to controls. Select gene expression patterns were confirmed by quantitative real-time PCR and immunofluorescence imaging. Lastly, trigeminal neurons were co-cultured atop Transwell filters overlying primary Tgfbr2 f/f DP cells. Tgfbr2 in the DP was deleted via Adenovirus-expressed Cre recombinase. Confocal imaging of axons through the filter pores showed increased axonal sprouting from neurons cultured with Tgfbr2-positive DP cells compared to neurons cultured alone. Axon sprouting was reduced when Tgfbr2 was knocked down in the DP cells. Immunofluorescence of dentin sialophosphoprotein in co-cultured DP cells confirmed reduced mineralization potential in cells with Tgfbr2 deletion. Both our proteomics and RNA-Seq analyses indicate that axonal guidance cues, particularly semaphorin signaling, were disrupted by Tgfbr2 deletion. Thus, Tgfbr2 in the DP mesenchyme appears to regulate differentiation and the cells' ability to guide neurite outgrowth during tooth mineralization and innervation.
Collapse
Affiliation(s)
- Monica Stanwick
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Courtney Barkley
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rosa Serra
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew Kruggel
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Amy Webb
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Yue Zhao
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Chandler Ashman
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Allie Staats
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Shifa Shahid
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Sarah B. Peters
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States,Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: Sarah B. Peters,
| |
Collapse
|
118
|
New Function of Cholesterol Oxidation Products Involved in Osteoporosis Pathogenesis. Int J Mol Sci 2022; 23:ijms23042020. [PMID: 35216140 PMCID: PMC8876989 DOI: 10.3390/ijms23042020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/05/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis (OP) is a systemic bone disease characterized by decreased bone strength, microarchitectural changes in bone tissues, and increased risk of fracture. Its occurrence is closely related to various factors such as aging, genetic factors, living habits, and nutritional deficiencies as well as the disturbance of bone homeostasis. The dysregulation of bone metabolism is regarded as one of the key influencing factors causing OP. Cholesterol oxidation products (COPs) are important compounds in the maintenance of bone metabolic homeostasis by participating in several important biological processes such as the differentiation of mesenchymal stem cells, bone formation in osteoblasts, and bone resorption in osteoclasts. The effects of specific COPs on mesenchymal stem cells are mainly manifested by promoting osteoblast genesis and inhibiting adipocyte genesis. This review aims to elucidate the biological roles of COPs in OP development, starting from the molecular mechanisms of OP, pointing out opportunities and challenges in current research, and providing new ideas and perspectives for further studies of OP pathogenesis.
Collapse
|
119
|
Autophagy in mesenchymal progenitors protects mice against bone marrow failure after severe intermittent stress. Blood 2022; 139:690-703. [PMID: 34657154 PMCID: PMC8814682 DOI: 10.1182/blood.2021011775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 10/01/2021] [Indexed: 11/26/2022] Open
Abstract
The cellular mechanisms required to ensure homeostasis of the hematopoietic niche and the ability of this niche to support hematopoiesis upon stress remain elusive. We here identify Wnt5a in Osterix+ mesenchymal progenitor and stem cells (MSPCs) as a critical factor for niche-dependent hematopoiesis. Mice lacking Wnt5a in MSPCs suffer from stress-related bone marrow (BM) failure and increased mortality. Niche cells devoid of Wnt5a show defective actin stress fiber orientation due to an elevated activity of the small GTPase CDC42. This results in incorrect positioning of autophagosomes and lysosomes, thus reducing autophagy and increasing oxidative stress. In MSPCs from patients from BM failure states which share features of peripheral cytopenia and hypocellular BM, we find similar defects in actin stress fiber orientation, reduced and incorrect colocalization of autophagosomes and lysosomes, and CDC42 activation. Strikingly, a short pharmacological intervention to attenuate elevated CDC42 activation in vivo in mice prevents defective actin-anchored autophagy in MSPCs, salvages hematopoiesis and protects against lethal cytopenia upon stress. In summary, our study identifies Wnt5a as a restriction factor for niche homeostasis by affecting CDC42-regulated actin stress-fiber orientation and autophagy upon stress. Our data further imply a critical role for autophagy in MSPCs for adequate support of hematopoiesis by the niche upon stress and in human diseases characterized by peripheral cytopenias and hypocellular BM.
Collapse
|
120
|
Hopkins C, de Castro LF, Corsi A, Boyce A, Collins MT, Riminucci M, Heegaard AM. Fibrous dysplasia animal models: A systematic review. Bone 2022; 155:116270. [PMID: 34875396 DOI: 10.1016/j.bone.2021.116270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Fibrous dysplasia (FD) is a rare genetic bone disorder resulting in an overproduction of cAMP leading to a structurally unsound tissue, caused by a genetic mutation in the guanine nucleotide-binding protein gene (GNAS). In order to better understand this disease, several animal models have been developed with different strategies and features. OBJECTIVE Conduct a systematic review to analyze and compare animal models with the causative mutation and features of FD. METHODS A PRISMA search was conducted in Scopus, PubMed, and Web of Science. Studies reporting an in vivo model of FD that expressed the causative mutation were included for analysis. Models without the causative mutation, but developed an FD phenotype and models of FD cell implantation were included for subanalysis. RESULTS Seven unique models were identified. The models were assessed and compared for their face validity, construct validity, mosaicism, and induction methods. This was based on the features of clinical FD that were reported within the categories of: macroscopic features, imaging, histology and histomorphometry, histochemical and cellular markers, and blood/urine markers. LIMITATIONS None of the models reported all features of FD and some features were only reported in one model. This made comparing models a challenge, but indicates areas where further research is necessary. CONCLUSION The benefits and disadvantages of every model were assessed from a practical and scientific standpoint. While all published reports lacked complete data, the models have nonetheless informed our understanding of FD and provided meaningful information to guide researchers in bench and clinical research.
Collapse
Affiliation(s)
- Chelsea Hopkins
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Luis Fernandez de Castro
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alison Boyce
- Metabolic Bone Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Michael T Collins
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
121
|
Pierce JL, Sharma AK, Roberts RL, Yu K, Irsik DL, Choudhary V, Dorn JS, Bensreti H, Benson RD, Kaiser H, Khayrullin A, Davis C, Wehrle CJ, Johnson MH, Bollag WB, Hamrick MW, Shi X, Isales CM, McGee-Lawrence ME. The Glucocorticoid Receptor in Osterix-Expressing Cells Regulates Bone Mass, Bone Marrow Adipose Tissue, and Systemic Metabolism in Female Mice During Aging. J Bone Miner Res 2022; 37:285-302. [PMID: 34747055 PMCID: PMC9976194 DOI: 10.1002/jbmr.4468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 09/23/2021] [Accepted: 10/17/2021] [Indexed: 12/18/2022]
Abstract
Hallmarks of aging-associated osteoporosis include bone loss, bone marrow adipose tissue (BMAT) expansion, and impaired osteoblast function. Endogenous glucocorticoid levels increase with age, and elevated glucocorticoid signaling, associated with chronic stress and dysregulated metabolism, can have a deleterious effect on bone mass. Canonical glucocorticoid signaling through the glucocorticoid receptor (GR) was recently investigated as a mediator of osteoporosis during the stress of chronic caloric restriction. To address the role of the GR in an aging-associated osteoporotic phenotype, the current study utilized female GR conditional knockout (GR-CKO; GRfl/fl :Osx-Cre+) mice and control littermates on the C57BL/6 background aged to 21 months and studied in comparison to young (3- and 6-month-old) mice. GR deficiency in Osx-expressing cells led to low bone mass and BMAT accumulation that persisted with aging. Surprisingly, however, GR-CKO mice also exhibited alterations in muscle mass (reduced % lean mass and soleus fiber size), accompanied by reduced voluntary physical activity, and also exhibited higher whole-body metabolic rate and elevated blood pressure. Moreover, increased lipid storage was observed in GR-CKO osteoblastic cultures in a glucocorticoid-dependent fashion despite genetic deletion of the GR, and could be reversed via pharmacological inhibition of the mineralocorticoid receptor (MR). These findings provide evidence of a role for the GR (and possibly the MR) in facilitating healthy bone maintenance with aging in females. The effects of GR-deficient bone on whole-body physiology also demonstrate the importance of bone as an endocrine organ and suggest evidence for compensatory mechanisms that facilitate glucocorticoid signaling in the absence of osteoblastic GR function; these represent new avenues of research that may improve understanding of glucocorticoid signaling in bone toward the development of novel osteogenic agents. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Jessica L Pierce
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Anuj K Sharma
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Rachel L Roberts
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Debra L Irsik
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, USA.,Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Vivek Choudhary
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Jennifer S Dorn
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Husam Bensreti
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Reginald D Benson
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Helen Kaiser
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Andrew Khayrullin
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Colleen Davis
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Chase J Wehrle
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Maribeth H Johnson
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Department of Physiology, Augusta University, Augusta, GA, USA
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Xingming Shi
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Carlos M Isales
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA.,Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA
| |
Collapse
|
122
|
FAM20C plays a critical role in the development of mouse vertebra. Spine J 2022; 22:337-348. [PMID: 34343663 DOI: 10.1016/j.spinee.2021.07.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Family with sequence similarity 20-member C (FAM20C) is a protein kinase that is responsible for the phosphorylation of many secretory proteins; however, its roles in spine or vertebra development have not be studied. PURPOSE The aim of this investigation is to analyze the roles of FAM20C in vertebra development. STUDY DESIGN/SETTING A mouse study of the Fam20c gene using conditional knockout to assess the effects of its inactivation on vertebra development. METHODS By breeding Sox2-Cre mice with Fam20cflox/flox mice, Sox2-Cre;Fam20cflox/flox mice (abbreviated as cKO mice) are created. X-ray radiography, resin-casted scanning electron microscopy, Hematoxylin and Eosin staining, safranin O staining, Goldner's Masson trichrome staining, Von Kossa staining, tartrate-resistant alkaline phosphatase staining, immunohistochemistry staining, Western Immunoblotting and real-time PCR were employed to characterize the vertebrae of cKO mice compared to the normal control mice. RESULTS Inactivation of Fam20c in mice results in remarkable spine deformity, severe morphology and mineralization defects, altered levels of osteoblast differentiation markers, reduction of activity of the Wnt/β-catenin signaling pathway and reduced level of osteoclastogenesis in the vertebrae. CONCLUSIONS FAM20C plays an essential role in vertebral development; it may regulate vertebral formation through the Wnt/β-catenin signaling pathway. CLINICAL SIGNIFICANCE Mutations in the human FAM20C gene are associated with Raine syndrome. The findings of this study provide valuable clues for the clinical management of Raine syndrome regarding spine manifestations in patients.
Collapse
|
123
|
Ma C, Liu H, Wei Y, Li H, Miao D, Ren Y. Exogenous PTH 1-34 Attenuates Impaired Fracture Healing in Endogenous PTH Deficiency Mice via Activating Indian Hedgehog Signaling Pathway and Accelerating Endochondral Ossification. Front Cell Dev Biol 2022; 9:750878. [PMID: 35071224 PMCID: PMC8766796 DOI: 10.3389/fcell.2021.750878] [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] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Fracture healing is a complicated, long-term, and multistage repair process. Intermittent administration of parathyroid hormone (PTH) has been proven effective on intramembranous and endochondral bone formation during the fracture healing process, however, the mechanism is unclear. In this study, we investigated the role of exogenous PTH and endogenous PTH deficiency in bone fracture healing and explored the mechanism by using PTH knockout (PTH-/-) mice and ATDC5 cells. In a mouse femur fracture model, endogenous PTH deficiency could delay endochondral ossification whereas exogenous PTH promotes accumulation of endochondral bone, accelerates cartilaginous callus conversion to bony callus, enhances maturity of bony callus, and attenuates impaired fracture healing resulting from endogenous PTH deficiency. In fracture callus tissue, endogenous PTH deficiency could inhibit chondrocyte proliferation and differentiation whereas exogenous PTH could activate the IHH signaling pathway to accelerate endochondral ossification and rescue impaired fracture healing resulting from endogenous PTH deficiency. In vitro, exogenous PTH promotes cell proliferation by activating IHH signaling pathway on ATDC5 cells. In mechanistic studies, by using ChIP and luciferase reporter assays, we showed that PTH could phosphorylate CREB, and subsequently bind to the promoter of IHH, causing the activation of IHH gene expression. Therefore, results from this study support the concept that exogenous PTH 1-34 attenuates impaired fracture healing in endogenous PTH deficiency mice via activating the IHH pathway and accelerating endochondral ossification. Hence, the investigation of the mechanism underlying the effects of PTH treatment on fracture repair might guide the exploration of effective therapeutic targets for fracture.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huan Liu
- Department of Orthopaedics, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Yifan Wei
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - He Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- Nanjing Medical University, Affiliated Friendship Plastic Surgery Hospital, Nanjing, China
| | - Yongxin Ren
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
124
|
Wang X, Tian Y, Liang X, Yin C, Huai Y, Zhao Y, Huang Q, Chu X, Wang W, Qian AR. Bergamottin promotes osteoblast differentiation and bone formation via activating Wnt/β-catenin signaling pathway. Food Funct 2022; 13:2913-2924. [DOI: 10.1039/d1fo02755g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Osteoporosis is one of the most common bone disorders that seriously affect the health and life quality of elderly individuals. Reduced osteoblast differentiation and bone formation lead to changes in...
Collapse
|
125
|
Rendina-Ruedy E, Smith BJ. Common Dietary Modifications in Preclinical Models to Study Skeletal Health. Front Endocrinol (Lausanne) 2022; 13:932343. [PMID: 35909523 PMCID: PMC9329513 DOI: 10.3389/fendo.2022.932343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/27/2022] [Indexed: 12/03/2022] Open
Abstract
Bone is a highly dynamic tissue that undergoes continuous remodeling by bone resorbing osteoclasts and bone forming osteoblasts, a process regulated in large part by osteocytes. Dysregulation of these coupled catabolic and anabolic processes as in the case of menopause, type 2 diabetes mellitus, anorexia nervosa, and chronic kidney disease is known to increase fracture risk. Recent advances in the field of bone cell metabolism and bioenergetics have revealed that maintenance of the skeleton places a high energy demand on these cells involved in bone remodeling. These new insights highlight the reason that bone tissue is the beneficiary of a substantial proportion of cardiac output and post-prandial chylomicron remnants and requires a rich supply of nutrients. Studies designed for the specific purpose of investigating the impact of dietary modifications on bone homeostasis or that alter diet composition and food intake to produce the model can be found throughout the literature; however, confounding dietary factors are often overlooked in some of the preclinical models. This review will examine some of the common pre-clinical models used to study skeletal biology and its pathologies and the subsequent impact of various dietary factors on these model systems. Furthermore, the review will include how inadvertent effects of some of these dietary components can influence bone cell function and study outcomes.
Collapse
Affiliation(s)
- Elizabeth Rendina-Ruedy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- *Correspondence: Elizabeth Rendina-Ruedy,
| | - Brenda J. Smith
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
126
|
Kwon SR. The Long, Dynamic Journey to the Elucidation of the Links Between Inflammation, Ectopic Bone Formation, and Wnt Signaling in Ankylosing Spondylitis. JOURNAL OF RHEUMATIC DISEASES 2022; 29:1-3. [PMID: 37476699 PMCID: PMC10324921 DOI: 10.4078/jrd.2022.29.1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/04/2021] [Accepted: 12/05/2021] [Indexed: 07/22/2023]
Affiliation(s)
- Seong-Ryul Kwon
- Rheumatism Center, Department of Internal Medicine, Inha University College of Medicine, Incheon, Korea
| |
Collapse
|
127
|
Kfoury YS, Ji F, Mazzola M, Sykes DB, Scherer AK, Anselmo A, Akiyama Y, Mercier F, Severe N, Kokkaliaris KD, Zhao T, Brouse T, Saez B, Seidl J, Papazian A, Ivanov P, Mansour MK, Sadreyev RI, Scadden DT. tiRNA signaling via stress-regulated vesicle transfer in the hematopoietic niche. Cell Stem Cell 2021; 28:2090-2103.e9. [PMID: 34551362 PMCID: PMC8642285 DOI: 10.1016/j.stem.2021.08.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/23/2021] [Accepted: 08/17/2021] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) transfer complex biologic material between cells. However, the role of this process in vivo is poorly defined. Here, we demonstrate that osteoblastic cells in the bone marrow (BM) niche elaborate extracellular vesicles that are taken up by hematopoietic progenitor cells in vivo. Genotoxic or infectious stress rapidly increased stromal-derived extracellular vesicle transfer to granulocyte-monocyte progenitors. The extracellular vesicles contained processed tRNAs (tiRNAs) known to modulate protein translation. 5'-ti-Pro-CGG-1 was preferentially abundant in osteoblast-derived extracellular vesicles and, when transferred to granulocyte-monocyte progenitors, increased protein translation, cell proliferation, and myeloid differentiation. Upregulating EV transfer improved hematopoietic recovery from genotoxic injury and survival from fungal sepsis. Therefore, EV-mediated tiRNA transfer provides a stress-modulated signaling axis in the BM niche distinct from conventional cytokine-driven stress responses.
Collapse
Affiliation(s)
- Youmna S Kfoury
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Mazzola
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Allison K Scherer
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Anthony Anselmo
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yasutoshi Akiyama
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Francois Mercier
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Nicolas Severe
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Konstantinos D Kokkaliaris
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ting Zhao
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Thomas Brouse
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Borja Saez
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jefferson Seidl
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ani Papazian
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Pavel Ivanov
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Initiative for RNA Medicine, Boston, MA 02115, USA
| | - Michael K Mansour
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
128
|
Chinipardaz Z, Liu M, Graves D, Yang S. Diabetes impairs fracture healing through disruption of cilia formation in osteoblasts. Bone 2021; 153:116176. [PMID: 34508881 PMCID: PMC9160738 DOI: 10.1016/j.bone.2021.116176] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/22/2021] [Accepted: 09/05/2021] [Indexed: 02/06/2023]
Abstract
Diabetes-associated fracture risk and impaired fracture healing represents a serious health threat. It is well known that type 1 diabetes mellitus (T1DM) impairs fracture healing due to its effect on osteoblasts and their progenitor cells. Previous studies have showed that primary cilia and intraflagellar transport protein 80 (IFT80) are critical for bone formation. However, whether TIDM impairs fracture healing due to influencing ciliary gene expression and cilia formation is unknown. Here, we investigated the effect of T1DM on primary cilia in a streptozotocin induced diabetes mouse model and examined the impact of cilia on fracture healing in osteoblasts by deletion of IFT80 in osteoblast linage using osterix (OSX)-cre (OSXcretTAIFT80f/f). The results showed that diabetes inhibited ciliary gene expression and primary cilia formation to an extent that was similar to normoglycemic mice with IFT80 deletion. Moreover, diabetic mice and normoglycemic mice with cilia loss in osteoblasts (OSXcretTAIFT80f/f) both exhibited delayed fracture healing with significantly reduced bone density and mechanical strength as well as with reduced expression of osteoblast markers, decreased angiogenesis and proliferation of bone lining cells at the fracture sites. In vitro studies showed that advanced glycation end products (AGEs) downregulated IFT80 expression in osteoblast progenitors. Moreover, AGEs and IFT80 deletion significantly reduced cilia number and length which inhibited differentiation of primary osteoblast precursors. Thus, this study for the first time report that primary cilia are essential for bone regeneration during fracture healing and loss of cilia caused by diabetes in osteoblasts resulted in defective diabetic fracture healing.
Collapse
Affiliation(s)
- Zahra Chinipardaz
- Department of Basic and Translation Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Liu
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dana Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Shuying Yang
- Department of Basic and Translation Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, PA 19104, USA; The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
129
|
Wirth F, Huck K, Lubosch A, Zoeller C, Ghura H, Porubsky S, Nakchbandi IA. Cdc42 in osterix-expressing cells alters osteoblast behavior and myeloid lineage commitment. Bone 2021; 153:116150. [PMID: 34400384 DOI: 10.1016/j.bone.2021.116150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023]
Abstract
Osteoblasts are not only responsible for bone formation. They also support hematopoiesis. This requires responding to cues originating from several signaling pathways, a task performed by Rho GTPases. We therefore examined several transgenic mouse models and used inhibitors of Cdc42 in vitro. Deletion of Cdc42 in vivo using the Osterix promoter suppressed osteoblast function, while its deletion in differentiating osteoblasts using the Collagen-α1(I) promoter decreased osteoblast numbers. In both cases, bone mineral density diminished confirming the importance of Cdc42. Evaluation of hematopoiesis revealed that deletion of Cdc42 using the Osterix, but not the Collagen-α1(I) promoter increased the common myeloid progenitors (CMPs) in the bone marrow as well as the erythrocytes and the thrombocytes/platelets in peripheral blood. Causality between Cdc42 loss in early osteoblasts and increased myelopoiesis was confirmed in vitro. Work in vitro supported the conclusion that interleukin-4 mediated the increase in myelopoiesis. Thus, Cdc42 is required for healthy bone through regulation of bone formation in Osterix-expressing osteoblasts and the number of osteoblasts in differentiating osteoblasts. In addition, its expression in early osteoblasts/stromal cells modulates myelopoiesis. This highlights the importance of osteoblasts in regulating hematopoiesis.
Collapse
Affiliation(s)
- Franziska Wirth
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Katrin Huck
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Alexander Lubosch
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Caren Zoeller
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Hiba Ghura
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Stefan Porubsky
- Institute of Pathology, University of Mainz, 55131 Mainz, Germany
| | - Inaam A Nakchbandi
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany; Max-Planck Institute for Biochemistry, 82152 Martinsried, Germany.
| |
Collapse
|
130
|
Vasiliadis ES, Evangelopoulos DS, Kaspiris A, Vlachos C, Pneumaticos SG. Sclerostin and Its Involvement in the Pathogenesis of Idiopathic Scoliosis. J Clin Med 2021; 10:jcm10225286. [PMID: 34830568 PMCID: PMC8618875 DOI: 10.3390/jcm10225286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 12/28/2022] Open
Abstract
Idiopathic scoliosis is a disorder of unknown etiology. Bone biopsies from idiopathic scoliosis patients revealed changes at cellular and molecular level. Osteocytic sclerostin is downregulated, and serum level of sclerostin is decreased. Osteocytes in idiopathic scoliosis appear to be less active with abnormal canaliculi network. Differentiation of osteoblasts to osteocytes is decelerated, while Wnt/β-catenin signaling pathway is overactivated and affects normal bone mineralization that leads to inferior mechanical properties of the bone, which becomes susceptible to asymmetrical forces and causes deformity of the spinal column. Targeting bone metabolism during growth by stimulating sclerostin secretion from osteocytes and restoring normal function of Wnt/β-catenin signaling pathway could, in theory, increase bone strength and prevent deterioration of the scoliotic deformity.
Collapse
Affiliation(s)
- Elias S. Vasiliadis
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.S.E.); (C.V.); (S.G.P.)
- Correspondence: ; Tel.: +30-2132-086-000
| | - Dimitrios Stergios Evangelopoulos
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.S.E.); (C.V.); (S.G.P.)
| | - Angelos Kaspiris
- Laboratory of Molecular Pharmacology, Division for Orthopaedic Research, School of Health Sciences, University of Patras, 26504 Rion, Greece;
| | - Christos Vlachos
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.S.E.); (C.V.); (S.G.P.)
| | - Spyros G. Pneumaticos
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Hospital, 16541 Athens, Greece; (D.S.E.); (C.V.); (S.G.P.)
| |
Collapse
|
131
|
Castiella T, Iruzubieta P, Monleón E, Cardiel MJ, Gómez-Vallejo J, Monzón M, Junquera MC. Stromal cells of giant cell tumor of bone show primary cilia in giant cell tumor of bone. Microsc Res Tech 2021; 85:1065-1074. [PMID: 34761465 DOI: 10.1002/jemt.23976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/04/2021] [Accepted: 10/26/2021] [Indexed: 01/02/2023]
Abstract
Giant cell tumor of bone (GCTB) is a locally aggressive primary bone neoplasm composed by tumoral stromal cells (SCs) and a reactive component that consists of monocytic/histiocytic cells that give rise by fusion to osteoclast-like multinucleated cells. Recently, specific Histone 3.3 mutations have been demonstrated in SCs of GCTB. Many of the pathways related to bone proliferation and regulation depend on the primary cilium, a microtubule-based organelle that protrudes outside the cell and acts as a sensorial antenna. In the present work, we aimed to study the presence and role of primary cilia in GCTB. Ultrastructural, immunohistochemical, and immunofluorescence studies were performed in order to demonstrate, for the first time, that the primary cilium is located in spindle-shaped SCs of GCTB. Moreover, we showed Hedgehog (Hh) signaling pathway activation in these cells. Hence, primary cilia may play a relevant role in GCTB tumorogenesis through Hh signaling activation in SCs. RESEARCH HIGHLIGHTS: Transmission electron microscopy allows describing and differentiating cellular subpopulations in giant cell tumor of bone (GCTB). The primary cilium is present in some tumoral stromal cells of GCTB. Hedgehog signalling is activated in these cells.
Collapse
Affiliation(s)
- Tomás Castiella
- Department of Pathology, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain.,Institute for Health Research Aragón (IIS), Zaragoza, Spain
| | - Pablo Iruzubieta
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain
| | - Eva Monleón
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain.,Institute for Health Research Aragón (IIS), Zaragoza, Spain
| | - Mª José Cardiel
- Department of Pathology, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Jesús Gómez-Vallejo
- Department of Traumatology and Orthopaedic Surgery, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Marta Monzón
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain.,Institute for Health Research Aragón (IIS), Zaragoza, Spain
| | - Mª Concepción Junquera
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain.,Institute for Health Research Aragón (IIS), Zaragoza, Spain
| |
Collapse
|
132
|
Thomas JR, Sloan K, Cave K, Wallace JM, Roper RJ. Skeletal Deficits in Male and Female down Syndrome Model Mice Arise Independent of Normalized Dyrk1a Expression in Osteoblasts. Genes (Basel) 2021; 12:1729. [PMID: 34828335 PMCID: PMC8624983 DOI: 10.3390/genes12111729] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
Trisomy 21 (Ts21) causes alterations in skeletal development resulting in decreased bone mass, shortened stature and weaker bones in individuals with Down syndrome (DS). There is a sexual dimorphism in bone mineral density (BMD) deficits associated with DS with males displaying earlier deficits than females. The relationships between causative trisomic genes, cellular mechanisms, and influence of sex in DS skeletal abnormalities remain unknown. One hypothesis is that the low bone turnover phenotype observed in DS results from attenuated osteoblast function, contributing to impaired trabecular architecture, altered cortical geometry, and decreased mineralization. DYRK1A, found in three copies in humans with DS, Ts65Dn, and Dp1Tyb DS model mice, has been implicated in the development of postnatal skeletal phenotypes associated with DS. Reduced copy number of Dyrk1a to euploid levels from conception in an otherwise trisomic Ts65Dn mice resulted in a rescue of appendicular bone deficits, suggesting DYRK1A contributes to skeletal development and homeostasis. We hypothesized that reduction of Dyrk1a copy number in trisomic osteoblasts would improve cellular function and resultant skeletal structural anomalies in trisomic mice. Female mice with a floxed Dyrk1a gene (Ts65Dn,Dyrk1afl/wt) were mated with male Osx-Cre+ (expressed in osteoblasts beginning around E13.5) mice, resulting in reduced Dyrk1a copy number in mature osteoblasts in Ts65Dn,Dyrk1a+/+/Osx-Cre P42 male and female trisomic and euploid mice, compared with littermate controls. Male and female Ts65Dn,Dyrk1a+/+/+ (3 copies of DYRK1A in osteoblasts) and Ts65Dn,Dyrk1a+/+/Osx-Cre (2 copies of Dyrk1a in osteoblasts) displayed similar defects in both trabecular architecture and cortical geometry, with no improvements with reduced Dyrk1a in osteoblasts. This suggests that trisomic DYRK1A does not affect osteoblast function in a cell-autonomous manner at or before P42. Although male Dp1Tyb and Ts65Dn mice exhibit similar skeletal deficits at P42 in both trabecular and cortical bone compartments between euploid and trisomic mice, female Ts65Dn mice exhibit significant cortical and trabecular deficits at P42, in contrast to an absence of genotype effect in female Dp1Tyb mice in trabecular bone. Taken together, these data suggest skeletal deficits in DS mouse models and are sex and age dependent, and influenced by strain effects, but are not solely caused by the overexpression of Dyrk1a in osteoblasts. Identifying molecular and cellular mechanisms, disrupted by gene dosage imbalance, that are involved in the development of skeletal phenotypes associated with DS could help to design therapies to rescue skeletal deficiencies seen in DS.
Collapse
Affiliation(s)
- Jared R. Thomas
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (J.R.T.); (K.S.); (K.C.)
| | - Kourtney Sloan
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (J.R.T.); (K.S.); (K.C.)
| | - Kelsey Cave
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (J.R.T.); (K.S.); (K.C.)
| | - Joseph M. Wallace
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Randall J. Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; (J.R.T.); (K.S.); (K.C.)
| |
Collapse
|
133
|
Rogers S, Scholpp S. Vertebrate Wnt5a - At the crossroads of cellular signalling. Semin Cell Dev Biol 2021; 125:3-10. [PMID: 34686423 DOI: 10.1016/j.semcdb.2021.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023]
Abstract
Wnt signalling is an essential pathway in embryogenesis, differentiation, cell motility, development, and adult tissue homeostasis in vertebrates. The Wnt signalling network can activate several downstream pathways such as the β-catenin-dependent TCF/LEF transcription, the Wnt/planar cell polarity (PCP) pathway, and the Wnt/Calcium pathway. Wnt5a is a vertebrate Wnt ligand that is most often associated with the Wnt/PCP signalling pathway. Wnt5a/PCP signalling has a well-described role in embryogenesis via binding to a receptor complex of Frizzled and its co-receptors to initiate downstream activation of the c-Jun N-terminal kinase (JNK) signalling cascade and the Rho and Rac GTPases, Rho-Kinase (ROCK). This activation results in the cytoskeletal remodelling required for cell polarity, migration, and subsequently, tissue re-arrangement and organ formation. This review will focus on more recent work that has revealed new roles for Wnt5a ligands and consequently, an emerging broader function. This is partly due to our growing understanding of the crosstalk between the Wnt/PCP pathway with both the Wnt/β-catenin pathway and other signalling pathways, and in part due to the identification of novel atypical receptors for Wnt5a that demonstrate a far broader role for this ligand.
Collapse
Affiliation(s)
- Sally Rogers
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
134
|
Sharma D, Yu Y, Shen L, Zhang GF, Karner CM. SLC1A5 provides glutamine and asparagine necessary for bone development in mice. eLife 2021; 10:71595. [PMID: 34647520 PMCID: PMC8553342 DOI: 10.7554/elife.71595] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/12/2021] [Indexed: 12/03/2022] Open
Abstract
Osteoblast differentiation is sequentially characterized by high rates of proliferation followed by increased protein and matrix synthesis, processes that require substantial amino acid acquisition and production. How osteoblasts obtain or maintain intracellular amino acid production is poorly understood. Here, we identify SLC1A5 as a critical amino acid transporter during bone development. Using a genetic and metabolomic approach, we show SLC1A5 acts cell autonomously to regulate protein synthesis and osteoblast differentiation. SLC1A5 provides both glutamine and asparagine which are essential for osteoblast differentiation. Mechanistically, glutamine and to a lesser extent asparagine support amino acid biosynthesis. Thus, osteoblasts depend on Slc1a5 to provide glutamine and asparagine, which are subsequently used to produce non-essential amino acids and support osteoblast differentiation and bone development.
Collapse
Affiliation(s)
- Deepika Sharma
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, United States
| | - Yilin Yu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Leyao Shen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, Durham, United States.,Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Courtney M Karner
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Charles and Jane Pak Center for Mineral Metabolism and Clinical Research. University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| |
Collapse
|
135
|
Chaugule S, Kim JM, Yang YS, Knobeloch KP, He X, Shim JH. Deubiquitinating Enzyme USP8 Is Essential for Skeletogenesis by Regulating Wnt Signaling. Int J Mol Sci 2021; 22:10289. [PMID: 34638628 PMCID: PMC8508692 DOI: 10.3390/ijms221910289] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 12/20/2022] Open
Abstract
Disturbance in a differentiation program of skeletal stem cells leads to indecorous skeletogenesis. Growing evidence suggests that a fine-tuning of ubiquitin-mediated protein degradation is crucial for skeletal stem cells to maintain their stemness and osteogenic potential. Here, we demonstrate that the deubiquitinating enzyme (DUB) ubiquitin-specific protease 8 (USP8) stabilizes the Wnt receptor frizzled 5 (FZD5) by preventing its lysosomal degradation. This pathway is essential for Wnt/β-catenin signaling and the differentiation of osteoprogenitors to mature osteoblasts. Accordingly, deletion of USP8 in osteoprogenitors (Usp8Osx) resulted in a near-complete blockade in skeletal mineralization, similar to that seen in mice with defective Wnt/β-catenin signaling. Likewise, transplanting USP8-deficient osteoprogenitors under the renal capsule in wild-type secondary hosts did not to induce bone formation. Collectively, this study unveils an essential role for the DUB USP8 in Wnt/β-catenin signaling in osteoprogenitors and osteogenesis during skeletal development.
Collapse
Affiliation(s)
- Sachin Chaugule
- Shim Lab, Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (S.C.); (J.-M.K.); (Y.-S.Y.)
| | - Jung-Min Kim
- Shim Lab, Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (S.C.); (J.-M.K.); (Y.-S.Y.)
| | - Yeon-Suk Yang
- Shim Lab, Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (S.C.); (J.-M.K.); (Y.-S.Y.)
| | - Klaus-Peter Knobeloch
- Institute of Neuropathology, Medical Faculty, University of Freiburg, 79106 Freiburg, Germany;
| | - Xi He
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Department of Neurology, Harvard Medical School, Boston, MA 02115, USA;
| | - Jae-Hyuck Shim
- Shim Lab, Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (S.C.); (J.-M.K.); (Y.-S.Y.)
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
136
|
Rauner M, Murray M, Thiele S, Watts D, Neumann D, Gabet Y, Hofbauer LC, Wielockx B. Epo/EpoR signaling in osteoprogenitor cells is essential for bone homeostasis and Epo-induced bone loss. Bone Res 2021; 9:42. [PMID: 34518518 PMCID: PMC8437981 DOI: 10.1038/s41413-021-00157-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/05/2021] [Accepted: 04/20/2021] [Indexed: 11/09/2022] Open
Abstract
High erythropoietin (Epo) levels are detrimental to bone health in adult organisms. Adult mice receiving high doses of Epo lose bone mass due to suppressed bone formation and increased bone resorption. In humans, high serum Epo levels are linked to fractures in elderly men. Our earlier studies indicated that Epo modulates osteoblast activity; however, direct evidence that Epo acts via its receptor (EpoR) on osteoblasts in vivo is still missing. Here, we created mice lacking EpoR in osteoprogenitor cells to specifically address this gap. Deletion of EpoR in osteoprogenitors (EpoR:Osx-cre, cKO) starting at 5 weeks of age did not alter red blood cell parameters but increased vertebral bone volume by 25% in 12-week-old female mice. This was associated with low bone turnover. Histological (osteoblast number, bone formation rate) and serum (P1NP, osteocalcin) bone formation parameters were all reduced, as were the number of osteoclasts and TRAP serum level. Differentiation of osteoblast precursors isolated from cKO versus control mice resulted in lower expression of osteoblast marker genes including Runx2, Alp, and Col1a1 on day 21, whereas the mineralization capacity was similar. Moreover, the RANKL/OPG ratio, which determines the osteoclast-supporting potential of osteoblasts, was substantially decreased by 50%. Similarly, coculturing cKO osteoblasts with control or cKO osteoclast precursors produced significantly fewer osteoclasts than coculture with control osteoblasts. Finally, exposing female mice to Epo pumps (10 U·d−1) for 4 weeks resulted in trabecular bone loss (−25%) and increased osteoclast numbers (1.7-fold) in control mice only, not in cKO mice. Our data show that EpoR in osteoprogenitors is essential in regulating osteoblast function and osteoblast-mediated osteoclastogenesis via the RANKL/OPG axis. Thus, osteogenic Epo/EpoR signaling controls bone mass maintenance and contributes to Epo-induced bone loss.
Collapse
Affiliation(s)
- Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.
| | - Marta Murray
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Deepika Watts
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yankel Gabet
- Department of Anatomy & Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lorenz C Hofbauer
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
137
|
Snider TN, Louie KW, Zuzo G, Ruellas ACDO, Solem RC, Cevidanes LHS, Zhang H, Mishina Y. Quantification of three-dimensional morphology of craniofacial mineralized tissue defects in Tgfbr2/Osx-Cre mice. ORAL SCIENCE INTERNATIONAL 2021; 18:193-202. [PMID: 34720652 PMCID: PMC8552916 DOI: 10.1002/osi2.1099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Craniofacial morphology is affected by the growth, development, and three-dimensional (3D) relationship of mineralized structures including the skull, jaws, and teeth. Despite fulfilling different purposes within this region, cranial bones and tooth dentin are derived from mesenchymal cells that are affected by perturbations within the TGF-β signaling pathway. TGFBR2 encodes a transmembrane receptor that is part of the canonical, SMAD-dependent TGF-β signaling pathway and mutations within this gene are associated with Loeys-Dietz syndrome, a condition which often presents with craniofacial signs including craniosynostosis and cleft palate. To investigate the role of Tgfbr2 in immature, but committed, mineralized tissue forming cells, we analyzed postnatal craniofacial morphology in mice with conditional Tgfbr2 deletion in Osx-expressing cells. Novel application of a 3D shape-based comparative technique revealed that Tgfbr2 in Osx-expressing cells results in impaired postnatal molar root and anterior cranial growth. These findings support those from studies using similar Tgfbr2 conditional knockout models, highlight the anomalous facial and dental regions/structures using tomographic imaging-based techniques, and provide insight into the role of Tgfbr2 during postnatal craniofacial development.
Collapse
Affiliation(s)
- Taylor Nicholas Snider
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Ke’ale W. Louie
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Gabrielle Zuzo
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | | | - Richard Christian Solem
- Department of Pediatric and Orthodontic Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Lucia H. S. Cevidanes
- Department of Pediatric and Orthodontic Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Honghao Zhang
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
138
|
Couasnay G, Madel MB, Lim J, Lee B, Elefteriou F. Sites of Cre-recombinase activity in mouse lines targeting skeletal cells. J Bone Miner Res 2021; 36:1661-1679. [PMID: 34278610 DOI: 10.1002/jbmr.4415] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022]
Abstract
The Cre/Lox system is a powerful tool in the biologist's toolbox, allowing loss-of-function and gain-of-function studies, as well as lineage tracing, through gene recombination in a tissue-specific and inducible manner. Evidence indicates, however, that Cre transgenic lines have a far more nuanced and broader pattern of Cre activity than initially thought, exhibiting "off-target" activity in tissues/cells other than the ones they were originally designed to target. With the goal of facilitating the comparison and selection of optimal Cre lines to be used for the study of gene function, we have summarized in a single manuscript the major sites and timing of Cre activity of the main Cre lines available to target bone mesenchymal stem cells, chondrocytes, osteoblasts, osteocytes, tenocytes, and osteoclasts, along with their reported sites of "off-target" Cre activity. We also discuss characteristics, advantages, and limitations of these Cre lines for users to avoid common risks related to overinterpretation or misinterpretation based on the assumption of strict cell-type specificity or unaccounted effect of the Cre transgene or Cre inducers. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Greig Couasnay
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | | | - Joohyun Lim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Florent Elefteriou
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
139
|
Keikhosravani P, Maleki-Ghaleh H, Kahaie Khosrowshahi A, Bodaghi M, Dargahi Z, Kavanlouei M, Khademi-Azandehi P, Fallah A, Beygi-Khosrowshahi Y, Siadati MH. Bioactivity and Antibacterial Behaviors of Nanostructured Lithium-Doped Hydroxyapatite for Bone Scaffold Application. Int J Mol Sci 2021; 22:ijms22179214. [PMID: 34502124 PMCID: PMC8430817 DOI: 10.3390/ijms22179214] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 12/11/2022] Open
Abstract
The material for bone scaffold replacement should be biocompatible and antibacterial to prevent scaffold-associated infection. We biofunctionalized the hydroxyapatite (HA) properties by doping it with lithium (Li). The HA and 4 Li-doped HA (0.5, 1.0, 2.0, 4.0 wt.%) samples were investigated to find the most suitable Li content for both aspects. The synthesized nanoparticles, by the mechanical alloying method, were cold-pressed uniaxially and then sintered for 2 h at 1250 °C. Characterization using field-emission scanning electron microscopy (FE-SEM) revealed particle sizes in the range of 60 to 120 nm. The XRD analysis proved the formation of HA and Li-doped HA nanoparticles with crystal sizes ranging from 59 to 89 nm. The bioactivity of samples was investigated in simulated body fluid (SBF), and the growth of apatite formed on surfaces was evaluated using SEM and EDS. Cellular behavior was estimated by MG63 osteoblast-like cells. The results of apatite growth and cell analysis showed that 1.0 wt.% Li doping was optimal to maximize the bioactivity of HA. Antibacterial characteristics against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) were performed by colony-forming unit (CFU) tests. The results showed that Li in the structure of HA increases its antibacterial properties. HA biofunctionalized by Li doping can be considered a suitable option for the fabrication of bone scaffolds due to its antibacterial and unique bioactivity properties.
Collapse
Affiliation(s)
- Pardis Keikhosravani
- Department of Materials Science and Engineering, K. N. Toosi University of Technology, Tehran P.O. Box 19919-43344, Iran; (P.K.); (M.H.S.)
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Hossein Maleki-Ghaleh
- Department of Materials Science and Engineering, K. N. Toosi University of Technology, Tehran P.O. Box 19919-43344, Iran; (P.K.); (M.H.S.)
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 51368, Iran
- Correspondence: (H.M.-G.); (Y.B.-K.); Tel.: +98-919-110-5425 (H.M.-G.)
| | - Amir Kahaie Khosrowshahi
- Department of Chemical Engineering, Sahand University of Technology, Tabriz P.O. Box 51335-1996, Iran;
- Tissue Engineering and Stem Cells Research Center, Sahand University of Technology, Tabriz P.O. Box 51335-1996, Iran
| | - Mahdi Bodaghi
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK;
| | - Ziba Dargahi
- Department of Materials Engineering, University of Tabriz, Tabriz 51368, Iran;
| | - Majid Kavanlouei
- Materials Engineering Department, Faculty of Engineering, Urmia University, Urmia P.O. Box 57561-51818, Iran;
| | - Pooriya Khademi-Azandehi
- Research Center for Advanced Materials, Faculty of Materials Engineering, Sahand University of Technology, Tabriz P.O. Box 51335-1996, Iran;
| | - Ali Fallah
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey;
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
| | - Younes Beygi-Khosrowshahi
- Chemical Engineering Group, Faculty of Engineering, Azarbaijan Shahid Madani University, Tabriz P.O. Box 53751-71379, Iran
- Correspondence: (H.M.-G.); (Y.B.-K.); Tel.: +98-919-110-5425 (H.M.-G.)
| | - M. Hossein Siadati
- Department of Materials Science and Engineering, K. N. Toosi University of Technology, Tehran P.O. Box 19919-43344, Iran; (P.K.); (M.H.S.)
| |
Collapse
|
140
|
Mesnieres M, Böhm AM, Peredo N, Trompet D, Valle-Tenney R, Bajaj M, Corthout N, Nefyodova E, Cardoen R, Baatsen P, Munck S, Nagy A, Haigh JJ, Khurana S, Verfaillie CM, Maes C. Fetal hematopoietic stem cell homing is controlled by VEGF regulating the integrity and oxidative status of the stromal-vascular bone marrow niches. Cell Rep 2021; 36:109618. [PMID: 34433017 PMCID: PMC8411121 DOI: 10.1016/j.celrep.2021.109618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/28/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem and progenitor cell (HSPC) engraftment after transplantation during anticancer treatment depends on support from the recipient bone marrow (BM) microenvironment. Here, by studying physiological homing of fetal HSPCs, we show the critical requirement of balanced local crosstalk within the skeletal niche for successful HSPC settlement in BM. Transgene-induced overproduction of vascular endothelial growth factor (VEGF) by osteoprogenitor cells elicits stromal and endothelial hyperactivation, profoundly impacting the stromal-vessel interface and vascular architecture. Concomitantly, HSPC homing and survival are drastically impaired. Transcriptome profiling, flow cytometry, and high-resolution imaging indicate alterations in perivascular and endothelial cell characteristics, vascular function and cellular metabolism, associated with increased oxidative stress within the VEGF-enriched BM environment. Thus, developmental HSPC homing to bone is controlled by local stromal-vascular integrity and the oxidative-metabolic status of the recipient milieu. Interestingly, irradiation of adult mice also induces stromal VEGF expression and similar osteo-angiogenic niche changes, underscoring that our findings may contribute targets for improving stem cell therapies. Establishment of BM hematopoiesis is coupled to development of the skeletal niches Primary HSPC seeding of bone depends on balanced molecular crosstalk in the niche Stromal VEGF triggers EC activation and controls stromal-vascular niche integrity Excessive skeletal VEGF deranges cell metabolism and induces oxidative stress in BM
Collapse
Affiliation(s)
- Marion Mesnieres
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Anna-Marei Böhm
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Nicolas Peredo
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Roger Valle-Tenney
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Manmohan Bajaj
- Stem Cell and Developmental Biology Unit, Stem Cell Institute Leuven, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Nikky Corthout
- VIB-KU Leuven Center for Brain & Disease Research, VIB BioImaging Center, KU Leuven, 3000 Leuven, Belgium; Research Group Molecular Neurobiology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
| | - Elena Nefyodova
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Ruben Cardoen
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Pieter Baatsen
- VIB-KU Leuven Center for Brain & Disease Research, VIB BioImaging Center, KU Leuven, 3000 Leuven, Belgium; Research Group Molecular Neurobiology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
| | - Sebastian Munck
- VIB-KU Leuven Center for Brain & Disease Research, VIB BioImaging Center, KU Leuven, 3000 Leuven, Belgium; Research Group Molecular Neurobiology, Department of Neurosciences, KU Leuven, 3000 Leuven, Belgium
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada; Department of Obstetrics and Gynecology, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jody J Haigh
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Research Institute in Oncology and Hematology, Cancer Care Manitoba, Winnipeg, MB, Canada
| | - Satish Khurana
- Stem Cell and Developmental Biology Unit, Stem Cell Institute Leuven, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; School of Biology, Indian Institute of Science Education and Research (IISER), Thiruvananthapuram, 695551 Kerala, India
| | - Catherine M Verfaillie
- Stem Cell and Developmental Biology Unit, Stem Cell Institute Leuven, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
141
|
Xu JQ, Tang N, Zhang LF, Tan C, Su Y, George DM, He GX, Huang TL. A bibliometric analysis of Wnt signaling pathway: from the top-100 cited articles to emerging trends. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1065. [PMID: 34422977 PMCID: PMC8339812 DOI: 10.21037/atm-21-174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/02/2021] [Indexed: 11/07/2022]
Abstract
Background Wnt signaling pathway plays a vital role in the regulation of development. An increasing number of articles about Wnt pathway components have been published. By analyzing these studies’ characteristics and qualities, we aim to reveal the current research focus and emerging trends in Wnt signaling. Methods The databases of Web of Science Core Collection, BIOSIS Citation Index, MEDLINE, etc. were utilized to identify articles on May 23rd, 2020. Wnt signaling pathway-related articles were identified, the 100 most cited articles and articles in the last decade were selected and calculated for citations without self-citation. The subsequent analysis included citation density (citations/article age), time-related flux, authorship, institution, journal, geographic distribution, and theme. Results These articles were published mainly from 2000 to 2009 (62%). Citations per article ranged from 599 to 3,780 with a median number of 880 times. Most studies (66%) came from the United States. Nusse Roel and Clevers Hans (15 and 13 papers) have contributed significantly to the field. The most highlighted study themes were cancer (15%), embryo development (14%), and cytoplasm signal transduction (11%). From 2011 to 2020, interest in emerging subtopics, including osteogenesis, immune, apoptosis, autophagy, microRNA, and cancer stem cell, are rising. Conclusions Cancer, embryo development, stem cell, and signal transduction process still play a major role in the field. With multiple emerging subtopics and investigation on an integrated view of the Wnt signal network, the association of Wnt with diseases was further revealed.
Collapse
Affiliation(s)
- Jia-Qi Xu
- Orthopaedic Department, The Second Xiangya Hospital of Central South University, Changsha, China.,Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Ning Tang
- Orthopaedic Department, The Second Xiangya Hospital of Central South University, Changsha, China
| | | | - Chen Tan
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Yang Su
- Orthopaedic Department, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Daniel M George
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Guang-Xu He
- Orthopaedic Department, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tian-Long Huang
- Orthopaedic Department, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
142
|
Du Y, Zhang M, Liu X, Li Z, Hu M, Tian Y, Lv L, Zhang X, Liu Y, Zhang P, Zhou Y. CDC20 promotes bone formation via APC/C dependent ubiquitination and degradation of p65. EMBO Rep 2021; 22:e52576. [PMID: 34382737 PMCID: PMC8419691 DOI: 10.15252/embr.202152576] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/23/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
The E3 ubiquitin ligase complex CDC20‐activated anaphase‐promoting complex/Cyclosome (APC/CCDC20) plays a critical role in governing mitotic progression by targeting key cell cycle regulators for degradation. Cell division cycle protein 20 homolog (CDC20), the co‐activator of APC/C, is required for full ubiquitin ligase activity. In addition to its well‐known cell cycle‐related functions, we demonstrate that CDC20 plays an essential role in osteogenic commitment of bone marrow mesenchymal stromal/stem cells (BMSCs). Cdc20 conditional knockout mice exhibit decreased bone formation and impaired bone regeneration after injury. Mechanistically, we discovered a functional interaction between the WD40 domain of CDC20 and the DNA‐binding domain of p65. Moreover, CDC20 promotes the ubiquitination and degradation of p65 in an APC11‐dependent manner. More importantly, knockdown of p65 rescues the bone loss in Cdc20 conditional knockout mice. Our current work reveals a cell cycle‐independent function of CDC20, establishes APC11CDC20 as a pivotal regulator for bone formation by governing the ubiquitination and degradation of p65, and may pave the way for treatment of bone‐related diseases.
Collapse
Affiliation(s)
- Yangge Du
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Min Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xuejiao Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Menglong Hu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yueming Tian
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
143
|
Shin TH, Theodorou E, Holland C, Yamin R, Raggio CL, Giampietro PF, Sweetser DA. TLE4 Is a Critical Mediator of Osteoblast and Runx2-Dependent Bone Development. Front Cell Dev Biol 2021; 9:671029. [PMID: 34422801 PMCID: PMC8377417 DOI: 10.3389/fcell.2021.671029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
Healthy bone homeostasis hinges upon a delicate balance and regulation of multiple processes that contribute to bone development and metabolism. While examining hematopoietic regulation by Tle4, we have uncovered a previously unappreciated role of Tle4 on bone calcification using a novel Tle4 null mouse model. Given the significance of osteoblasts in both hematopoiesis and bone development, this study investigated how loss of Tle4 affects osteoblast function. We used dynamic bone formation parameters and microCT to characterize the adverse effects of Tle4 loss on bone development. We further demonstrated loss of Tle4 impacts expression of several key osteoblastogenic genes, including Runx2, Oc, and Ap, pointing toward a potential novel mechanism for Tle4-dependent regulation of mammalian bone development in collaboration with the RUNX family members.
Collapse
Affiliation(s)
- Thomas H. Shin
- Department of Pediatrics, Center of Genomic Medicine, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Molecular and Translational Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Evangelos Theodorou
- Department of Pediatrics, Center of Genomic Medicine, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Carl Holland
- Department of Pediatrics, Center of Genomic Medicine, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Rae’e Yamin
- Department of Pediatrics, Center of Genomic Medicine, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Cathleen L. Raggio
- Department of Pediatric Orthopedics, Hospital for Special Surgery, New York, NY, United States
| | | | - David A. Sweetser
- Department of Pediatrics, Center of Genomic Medicine, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
144
|
Ko FC, Kobelski MM, Zhang W, Grenga GM, Martins JS, Demay MB. Phosphate restriction impairs mTORC1 signaling leading to increased bone marrow adipose tissue and decreased bone in growing mice. J Bone Miner Res 2021; 36:1510-1520. [PMID: 33900666 DOI: 10.1002/jbmr.4312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023]
Abstract
Bone marrow stromal cells (BMSCs) are multipotent cells that differentiate into cells of the osteogenic and adipogenic lineage. A striking inverse relationship between bone marrow adipose tissue (BMAT) and bone volume is seen in several conditions, suggesting that differentiation of BMSCs into bone marrow adipocytes diverts cells from the osteogenic lineage, thereby compromising the structural and mechanical properties of bone. Phosphate restriction of growing mice acutely decreases bone formation, blocks osteoblast differentiation and increases BMAT. Studies performed to evaluate the cellular and molecular basis for the effects of acute phosphate restriction demonstrate that it acutely increases 5' adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and inhibits mammalian target of rapamycin complex 1 (mTORC1) signaling in osteoblasts. This is accompanied by decreased expression of Wnt10b in BMSCs. Phosphate restriction also promotes expression of the key adipogenic transcription factors, peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT-enhancer binding protein α (CEBPα), in CXCL12 abundant reticular (CAR) cells, which represent undifferentiated BMSCs and are the main source of BMAT and osteoblasts in the adult murine skeleton. Consistent with this, lineage tracing studies reveal that the BMAT observed in phosphate-restricted mice is of CAR cell origin. To determine whether circumventing the decrease in mTORC1 signaling in maturing osteoblasts attenuates the osteoblast and BMAT phenotype, phosphate-restricted mice with OSX-CreERT2 -mediated haploinsufficiency of the mTORC1 inhibitor, TSC2, were generated. TSC2 haploinsufficiency in preosteoblasts/osteoblasts normalized bone volume and osteoblast number in phosphate-restricted mice and attenuated the increase in BMAT observed. Thus, acute phosphate restriction leads to decreased bone and increases BMAT by impairing mTORC1 signaling in osterix-expressing cells. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Frank C Ko
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | | | - Wanlin Zhang
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gina M Grenga
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Janaina S Martins
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
145
|
Liu Z, Hussien AA, Wang Y, Heckmann T, Gonzalez R, Karner CM, Snedeker JG, Gray RS. An adhesion G protein-coupled receptor is required in cartilaginous and dense connective tissues to maintain spine alignment. eLife 2021; 10:67781. [PMID: 34318745 PMCID: PMC8328515 DOI: 10.7554/elife.67781] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is the most common spine disorder affecting children worldwide, yet little is known about the pathogenesis of this disorder. Here, we demonstrate that genetic regulation of structural components of the axial skeleton, the intervertebral discs, and dense connective tissues (i.e., ligaments and tendons) is essential for the maintenance of spinal alignment. We show that the adhesion G protein-coupled receptor ADGRG6, previously implicated in human AIS association studies, is required in these tissues to maintain typical spine alignment in mice. Furthermore, we show that ADGRG6 regulates biomechanical properties of tendon and stimulates CREB signaling governing gene expression in cartilaginous tissues of the spine. Treatment with a cAMP agonist could mirror aspects of receptor function in culture, thus defining core pathways for regulating these axial cartilaginous and connective tissues. As ADGRG6 is a key gene involved in human AIS, these findings open up novel therapeutic opportunities for human scoliosis.
Collapse
Affiliation(s)
- Zhaoyang Liu
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, United States.,Department of Nutritional Sciences, The University of Texas at Austin, Austin, United States
| | - Amro A Hussien
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland.,Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Yunjia Wang
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, United States.,Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Terry Heckmann
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, United States
| | - Roberto Gonzalez
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, United States
| | - Courtney M Karner
- Department of Internal Medicine, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jess G Snedeker
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland.,Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ryan S Gray
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, United States.,Department of Nutritional Sciences, The University of Texas at Austin, Austin, United States
| |
Collapse
|
146
|
Davis JL, Thaler R, Cox L, Ricci B, Zannit HM, Wan F, Faccio R, Dudakovic A, van Wijnen AJ, Veis DJ. Constitutive activation of NF-κB inducing kinase (NIK) in the mesenchymal lineage using Osterix (Sp7)- or Fibroblast-specific protein 1 (S100a4)-Cre drives spontaneous soft tissue sarcoma. PLoS One 2021; 16:e0254426. [PMID: 34292968 PMCID: PMC8297882 DOI: 10.1371/journal.pone.0254426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 06/27/2021] [Indexed: 01/02/2023] Open
Abstract
Aberrant NF-κB signaling fuels tumor growth in multiple human cancer types including both hematologic and solid malignancies. Chronic elevated alternative NF-κB signaling can be modeled in transgenic mice upon activation of a conditional NF-κB-inducing kinase (NIK) allele lacking the regulatory TRAF3 binding domain (NT3). Here, we report that expression of NT3 in the mesenchymal lineage with Osterix (Osx/Sp7)-Cre or Fibroblast-Specific Protein 1 (FSP1)-Cre caused subcutaneous, soft tissue tumors. These tumors displayed significantly shorter latency and a greater multiple incidence rate in Fsp1-Cre;NT3 compared to Osx-Cre;NT3 mice, regardless of sex. Histological assessment revealed poorly differentiated solid tumors with some spindled patterns, as well as robust RelB immunostaining, confirming activation of alternative NF-κB. Even though NT3 expression also occurs in the osteolineage in Osx-Cre;NT3 mice, we observed no bony lesions. The staining profiles and pattern of Cre expression in the two lines pointed to a mesenchymal tumor origin. Immunohistochemistry revealed that these tumors stain strongly for alpha-smooth muscle actin (αSMA), although vimentin staining was uniform only in Osx-Cre;NT3 tumors. Negative CD45 and S100 immunostains precluded hematopoietic and melanocytic origins, respectively, while positive staining for cytokeratin 19 (CK19), typically associated with epithelia, was found in subpopulations of both tumors. Principal component, differential expression, and gene ontology analyses revealed that NT3 tumors are distinct from normal mesenchymal tissues and are enriched for NF-κB related biological processes. We conclude that constitutive activation of the alternative NF-κB pathway in the mesenchymal lineage drives spontaneous sarcoma and provides a novel mouse model for NF-κB related sarcomas.
Collapse
Affiliation(s)
- Jennifer L. Davis
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States of America
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States of America
| | - Linda Cox
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States of America
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Biancamaria Ricci
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States of America
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO, United States of America
| | - Heather M. Zannit
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States of America
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO, United States of America
| | - Fei Wan
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Roberta Faccio
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States of America
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO, United States of America
- Shriners Hospitals for Children–St. Louis, St. Louis, MO, United States of America
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States of America
| | - Andre J. van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States of America
| | - Deborah J. Veis
- Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States of America
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, United States of America
- Shriners Hospitals for Children–St. Louis, St. Louis, MO, United States of America
| |
Collapse
|
147
|
Cong Q, Liu Y, Zhou T, Zhou Y, Xu R, Cheng C, Chung HS, Yan M, Zhou H, Liao Z, Gao B, Bocobo GA, Covington TA, Song HJ, Su P, Yu PB, Yang Y. A self-amplifying loop of YAP and SHH drives formation and expansion of heterotopic ossification. Sci Transl Med 2021; 13:13/599/eabb2233. [PMID: 34162750 PMCID: PMC8638088 DOI: 10.1126/scitranslmed.abb2233] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 12/03/2020] [Accepted: 05/30/2021] [Indexed: 12/15/2022]
Abstract
Heterotopic ossification (HO) occurs as a common complication after injury or in genetic disorders. The mechanisms underlying HO remain incompletely understood, and there are no approved prophylactic or secondary treatments available. Here, we identify a self-amplifying, self-propagating loop of Yes-associated protein (YAP)-Sonic hedgehog (SHH) as a core molecular mechanism underlying diverse forms of HO. In mouse models of progressive osseous heteroplasia (POH), a disease caused by null mutations in GNAS, we found that Gnas-/- mesenchymal cells secreted SHH, which induced osteoblast differentiation of the surrounding wild-type cells. We further showed that loss of Gnas led to activation of YAP transcription activity, which directly drove Shh expression. Secreted SHH further induced YAP activation, Shh expression, and osteoblast differentiation in surrounding wild-type cells. This self-propagating positive feedback loop was both necessary and sufficient for HO expansion and could act independently of Gnas in fibrodysplasia ossificans progressiva (FOP), another genetic HO, and nonhereditary HO mouse models. Genetic or pharmacological inhibition of YAP or SHH abolished HO in POH and FOP and acquired HO mouse models without affecting normal bone homeostasis, providing a previously unrecognized therapeutic rationale to prevent, reduce, and shrink HO.
Collapse
Affiliation(s)
- Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Taifeng Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Yaxing Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Ruoshi Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Caiqi Cheng
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Hye Soo Chung
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Meijun Yan
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Hang Zhou
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Zhiheng Liao
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Bo Gao
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Geoffrey A Bocobo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Taylor A Covington
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Hyeon Ju Song
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Peiqiang Su
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA.
| |
Collapse
|
148
|
Yadav PS, Feng S, Cong Q, Kim H, Liu Y, Yang Y. Stat3 loss in mesenchymal progenitors causes Job syndrome-like skeletal defects by reducing Wnt/β-catenin signaling. Proc Natl Acad Sci U S A 2021; 118:e2020100118. [PMID: 34172578 PMCID: PMC8256036 DOI: 10.1073/pnas.2020100118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Job syndrome is a rare genetic disorder caused by STAT3 mutations and primarily characterized by immune dysfunction along with comorbid skeleton developmental abnormalities including osteopenia, recurrent fracture of long bones, and scoliosis. So far, there is no definitive cure for the skeletal defects in Job syndrome, and treatments are limited to management of clinical symptoms only. Here, we have investigated the molecular mechanism whereby Stat3 regulates skeletal development and osteoblast differentiation. We showed that removing Stat3 function in the developing limb mesenchyme or osteoprogenitor cells in mice resulted in shortened and bow limbs with multiple fractures in long bones that resembled the skeleton symptoms in the Job Syndrome. However, Stat3 loss did not alter chondrocyte differentiation and hypertrophy in embryonic development, while osteoblast differentiation was severely reduced. Genome-wide transcriptome analyses as well as biochemical and histological studies showed that Stat3 loss resulted in down-regulation of Wnt/β-catenin signaling. Restoration of Wnt/β-catenin signaling by injecting BIO, a small molecule inhibitor of GSK3, or crossing with a Lrp5 gain of function (GOF) allele, rescued the bone reduction phenotypes due to Stat3 loss to a great extent. These studies uncover the essential functions of Stat3 in maintaining Wnt/β-catenin signaling in early mesenchymal or osteoprogenitor cells and provide evidence that bone defects in the Job Syndrome are likely caused by Wnt/β-catenin signaling reduction due to reduced STAT3 activities in bone development. Enhancing Wnt/β-catenin signaling could be a therapeutic approach to reduce bone symptoms of Job syndrome patients.
Collapse
Affiliation(s)
- Prem Swaroop Yadav
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Shuhao Feng
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Hanjun Kim
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115;
- Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
149
|
Skeletal Deformities in Osterix-Cre;Tgfbr2 f/f Mice May Cause Postnatal Death. Genes (Basel) 2021; 12:genes12070975. [PMID: 34202311 PMCID: PMC8307487 DOI: 10.3390/genes12070975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/25/2022] Open
Abstract
Transforming growth factor β (TGFβ) signaling plays an important role in skeletal development. We previously demonstrated that the loss of TGFβ receptor II (Tgfbr2) in Osterix-Cre-expressing mesenchyme results in defects in bones and teeth due to reduced proliferation and differentiation in pre-osteoblasts and pre-odontoblasts. These Osterix-Cre;Tgfbr2f/f mice typically die within approximately four weeks for unknown reasons. To investigate the cause of death, we performed extensive pathological analysis on Osterix-Cre- (Cre-), Osterix-Cre+;Tgfbr2f/wt (HET), and Osterix-Cre+;Tgfbr2f/f (CKO) mice. We also crossed Osterix-Cre mice with the ROSA26mTmG reporter line to identify potential off-target Cre expression. The findings recapitulated published skeletal and tooth abnormalities and revealed previously unreported osteochondral dysplasia throughout both the appendicular and axial skeletons in the CKO mice, including the calvaria. Alterations to the nasal area and teeth suggest a potentially reduced capacity to sense and process food, while off-target Cre expression in the gastrointestinal tract may indicate an inability to absorb nutrients. Additionally, altered nasal passages and unexplained changes in diaphragmatic muscle support the possibility of hypoxia. We conclude that these mice likely died due to a combination of breathing difficulties, malnutrition, and starvation resulting primarily from skeletal deformities that decreased their ability to sense, gather, and process food.
Collapse
|
150
|
Romero-Márquez JM, Varela-López A, Navarro-Hortal MD, Badillo-Carrasco A, Forbes-Hernández TY, Giampieri F, Domínguez I, Madrigal L, Battino M, Quiles JL. Molecular Interactions between Dietary Lipids and Bone Tissue during Aging. Int J Mol Sci 2021; 22:ijms22126473. [PMID: 34204176 PMCID: PMC8233828 DOI: 10.3390/ijms22126473] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 01/06/2023] Open
Abstract
Age-related bone disorders such as osteoporosis or osteoarthritis are a major public health problem due to the functional disability for millions of people worldwide. Furthermore, fractures are associated with a higher degree of morbidity and mortality in the long term, which generates greater financial and health costs. As the world population becomes older, the incidence of this type of disease increases and this effect seems notably greater in those countries that present a more westernized lifestyle. Thus, increased efforts are directed toward reducing risks that need to focus not only on the prevention of bone diseases, but also on the treatment of persons already afflicted. Evidence is accumulating that dietary lipids play an important role in bone health which results relevant to develop effective interventions for prevent bone diseases or alterations, especially in the elderly segment of the population. This review focuses on evidence about the effects of dietary lipids on bone health and describes possible mechanisms to explain how lipids act on bone metabolism during aging. Little work, however, has been accomplished in humans, so this is a challenge for future research.
Collapse
Affiliation(s)
- Jose M. Romero-Márquez
- Department of Physiology, Institute of Nutrition and Food Technology ‘‘José Mataix”, Biomedical Research Centre, University of Granada, Armilla, Avda. del Conocimiento s.n., 18100 Armilla, Spain; (J.M.R.-M.); (A.V.-L.); (M.D.N.-H.); (A.B.-C.)
| | - Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology ‘‘José Mataix”, Biomedical Research Centre, University of Granada, Armilla, Avda. del Conocimiento s.n., 18100 Armilla, Spain; (J.M.R.-M.); (A.V.-L.); (M.D.N.-H.); (A.B.-C.)
| | - María D. Navarro-Hortal
- Department of Physiology, Institute of Nutrition and Food Technology ‘‘José Mataix”, Biomedical Research Centre, University of Granada, Armilla, Avda. del Conocimiento s.n., 18100 Armilla, Spain; (J.M.R.-M.); (A.V.-L.); (M.D.N.-H.); (A.B.-C.)
| | - Alberto Badillo-Carrasco
- Department of Physiology, Institute of Nutrition and Food Technology ‘‘José Mataix”, Biomedical Research Centre, University of Granada, Armilla, Avda. del Conocimiento s.n., 18100 Armilla, Spain; (J.M.R.-M.); (A.V.-L.); (M.D.N.-H.); (A.B.-C.)
| | - Tamara Y. Forbes-Hernández
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
| | - Francesca Giampieri
- Department of Clinical Sicences, Università Politecnica delle Marche, 60131 Ancona, Italy; (F.G.); (M.B.)
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Irma Domínguez
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain;
- Universidad Internacional Iberoamericana, Calle 15 Num. 36, Entre 10 y 12 IMI III, Campeche 24560, Mexico;
| | - Lorena Madrigal
- Universidad Internacional Iberoamericana, Calle 15 Num. 36, Entre 10 y 12 IMI III, Campeche 24560, Mexico;
| | - Maurizio Battino
- Department of Clinical Sicences, Università Politecnica delle Marche, 60131 Ancona, Italy; (F.G.); (M.B.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - José L. Quiles
- Department of Physiology, Institute of Nutrition and Food Technology ‘‘José Mataix”, Biomedical Research Centre, University of Granada, Armilla, Avda. del Conocimiento s.n., 18100 Armilla, Spain; (J.M.R.-M.); (A.V.-L.); (M.D.N.-H.); (A.B.-C.)
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21, 39011 Santander, Spain;
- Correspondence:
| |
Collapse
|