1
|
杨 菊, 刘 玥, 曲 春, 孙 健, 李 天, 石 连. [Bisphosphonates-related osteonecrosis of the jaw: A case report]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2025; 57:388-392. [PMID: 40219574 PMCID: PMC11992463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Indexed: 04/14/2025]
Abstract
Osteonecrosis of the mandible is also called avascular necrosis of the jaw, and it is a rare complication of bisphosphonates. It is characterized with pain, swelling, exposure of bone, local infection and pathologic fractures of the jaw. With the widespread usage of bisphosphonates in bone metastasis of malignant tumors and osteoporosis, this rare complication has received more attention in recent years. Here, we reported a case of bisphosphonates-related osteonecrosis of the jaw (BRONJ) caused by intravenous zoledronic acid for osteoporosis. A 62-year-old female patient with 7-year history of Sjögren's syndrome and 3-year history of osteoporosis developed BRONJ after 3-year treatment of zoledronic acid. Two months before she went to the Peking University International Hospital, she visited the dentist for periodontal purulent secretion and extracted one tooth from the right mandible. However, the condition was not improved and she felt persistent pain and swelling in the right mandible. Hence, she received repeated root curettage, but there was no improvement. Finally, she was diagnosed with osteonecrosis of the mandible based on the digital volume tomography scan, which showed right mandibular osteonecrosis bone destruction. She underwent surgical debridement of the necrotic bone and administered intravenous antibio-tics at the Peking University International Hospital. Histopathological analysis of the bone biopsy further confirmed the diagnosis of BRONJ. Her condition was improved successfully during a 3-year follow-up. Osteonecrosis of the mandible become more common with the increased use of bisphosphonates. Recent study has reported that osteonecrosis of the mandible is more likely to occur in patients with Sjögren's syndrome. In addition, age, long-term and irregular administration of glucocorticoids, irregular oral examination and treatment also might be the risk factors in the pathogenesis of osteonecrosis of the mandible. For the elder osteoporosis patients who would receive or had received bisphosphonate-related drugs, oral health status and the disease states associated with necrosis of the mandible such as Sjögren's syndrome should be comprehensively measured and fully evaluated during the whole process. Furthermore, to better understand and prevent or reduce the occurrence of this complication, we reviewed the patho-genesis, diagnosis, treatment, and prevention of BRONJ.
Collapse
Affiliation(s)
- 菊 杨
- 遂宁市中心医院风湿免疫科,四川遂宁 629099Department of Rheumatology and Immunology, Suining Central Hospital, Suining 629099, Sichuan, China
- 北京大学国际医院风湿免疫科,北京 102206Department of Rheumatology and Immunology, Peking University International Hospital, Beijing 102206, China
| | - 玥 刘
- 北京大学国际医院口腔颌面外科,北京 102206Department of Maxillofacial Surgery, Peking University International Hospital, Beijing 102206, China
| | - 春娜 曲
- 北京大学国际医院口腔科,北京 102206Department of Stomatology, Peking University International Hospital, Beijing 102206, China
| | - 健斌 孙
- 北京大学国际医院内分泌科,北京 102206Department of Endocrinology, Peking University International Hospital, Beijing 102206, China
| | - 天英 李
- 北京大学国际医院病理科,北京 102206Department of Pathology, Peking University International Hospital, Beijing 102206, China
| | - 连杰 石
- 北京大学国际医院风湿免疫科,北京 102206Department of Rheumatology and Immunology, Peking University International Hospital, Beijing 102206, China
- 北京大学首钢医院风湿免疫科,北京 100144Department of Rheumatology and Immunology, Peking University Shougang Hospital, Beijing 100144, China
| |
Collapse
|
2
|
Wang Q, Wang X, Chen Y, Pan J. Dual role of Sfrp4 in bone remodelling during orthodontic tooth movement. Orthod Craniofac Res 2025; 28:175-186. [PMID: 39319631 DOI: 10.1111/ocr.12858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
OBJECTIVES The objective of this study was to determine changes in gene expression by establishing an orthodontic tooth movement (OTM) rat model with appropriate and excessive orthodontic force. MATERIALS AND METHODS Using a closed coil nickel-titanium spring, the OTM was carried out to apply a mesial force of 50 or 100 g to the maxillary first molars. Micro-CT, histological and immunohistochemical staining were used to evaluate the bone formation at the tension site and the bone resorption and bone formation at pressure site. Then RNA sequencing and bioinformatic analysis were performed. RESULTS According to the results of the Mirco-CT scan of OTM rat models, both the 50 g group and the 100 g group showed variable degrees of reduction in alveolar bone density on the tension and pressure sides. The results of histological and immunohistochemical staining demonstrated that the periodontal tissue and osteogenic ability of the 50 g group were restored at the 14 days, while the 100 g group caused severe periodontal tissue damage. The GO and KEGG analysis results, as well as the number of differentially expressed genes (DEGs), varied depending on the loading time and value of appliance, according to the results of the RNA sequencing. And the immunohistochemical staining results showed that Sfrp4 functioned by efficiently influencing both bone formation and bone absorption. CONCLUSIONS Appropriate orthodontic force value could cause appropriate movement of teeth in rats without adverse periodontal damage. Simultaneously, distinct gene expression patterns were observed at various force levels and time intervals.
Collapse
Affiliation(s)
- Qing Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuan Wang
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanzheng Chen
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jinsong Pan
- Department of Stomatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Kitazawa S, Haraguchi R, Kitazawa R. Roles of osteoclasts in pathological conditions. Pathol Int 2025; 75:55-68. [PMID: 39704061 PMCID: PMC11849001 DOI: 10.1111/pin.13500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024]
Abstract
Bone is a unique organ crucial for locomotion, mineral metabolism, and hematopoiesis. It maintains homeostasis through a balance between bone formation by osteoblasts and bone resorption by osteoclasts, which is regulated by the basic multicellular unit (BMU). Abnormal bone metabolism arises from an imbalance in the BMU. Osteoclasts, derived from the monocyte-macrophage lineage, are regulated by the RANKL-RANK-OPG system, which is a key factor in osteoclast differentiation. RANKL activates osteoclasts through its receptor RANK, while OPG acts as a decoy receptor that inhibits RANKL. In trabecular bone, high turnover involves rapid bone formation and resorption, influenced by conditions such as malignancy and inflammatory cytokines that increase RANKL expression. Cortical bone remodeling, regulated by aged osteocytes expressing RANKL, is less understood, despite ongoing research into how Rett syndrome, characterized by MeCP2 abnormalities, affects RANKL expression. Balancing trabecular and cortical bone involves mechanisms that preserve cortical bone, despite overall bone mass reduction due to aging or oxidative stress. Research into genes like sFRP4, which modulates bone mass, highlights the complex regulation by BMUs. The roles of the RANKL-RANK-OPG system extend beyond bone, affecting processes such as aortic valve formation and temperature regulation, which highlight the interconnected nature of biological research.
Collapse
Affiliation(s)
- Sohei Kitazawa
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
| | - Ryuma Haraguchi
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
| | - Riko Kitazawa
- Department of Molecular PathologyEhime University Graduate School of Medicine, ShitsukawaToon CityJapan
- Division of Diagnostic PathologyEhime University Hospital, ShitsukawaToon CityJapan
| |
Collapse
|
4
|
Song J, Chang W, Wang Y, Gao P, Zhang J, Xiao Z, An F, Yan C. Inhibitors of the Wnt pathway in osteoporosis: A review of mechanisms of action and potential as therapeutic targets. BIOMOLECULES & BIOMEDICINE 2025; 25:511-524. [PMID: 39606935 PMCID: PMC12010972 DOI: 10.17305/bb.2024.11200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024]
Abstract
The Wnt signaling pathway is one of the most important and critical signaling pathways for maintaining cellular functions, such as cell proliferation and differentiation. Increasing evidence substantiates that the Wnt signaling pathway also plays a significant role in the regulation of bone formation in osteoporosis. Accordingly, inhibitors of this pathway, such as sclerostin, Dickkopf-1 (DKK1), WNT inhibitory factor 1 (WIF1), and secreted frizzled-related proteins (SFRPs), have a negative regulatory role in bone formation and may serve as effective therapeutic targets for osteoporosis. This review examines the mechanisms of action of Wnt signaling pathway inhibitors in osteoporosis, the relationship between the Wnt pathway and its inhibitors, and new molecular targets for osteoporosis treatment. Overall, the regulatory mechanisms of Wnt pathway inhibitors are summarized to provide scientific and theoretical guidance for the treatment and prevention of osteoporosis.
Collapse
Affiliation(s)
- Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Weirong Chang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yujie Wang
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhipan Xiao
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chunlu Yan
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
5
|
Su K, Cui X, Zhou J, Yi Q, Liu O. Construction of an interactome network among circRNA-miRNA-mRNA reveals new biomarkers in hBMSCs osteogenic differentiation. Sci Rep 2024; 14:24507. [PMID: 39424659 PMCID: PMC11489463 DOI: 10.1038/s41598-024-76136-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
Human bone marrow mesenchymal stem cells (hBMSCs) are adult stem cells residing in the bone marrow, characterized by their capacity for multi-directional differentiation, self-renewal, migration, and engraftment. Serving as seed cells, BMSCs play a pivotal role in the regeneration of bone defects. Hence, investigating the transcription factors and signaling pathways involved in the regulation of osteogenic differentiation in BMSCs holds significant importance. Recent research has unveiled that certain circular RNAs (circRNAs) can function as molecular sponges, influencing the osteogenic differentiation process of mesenchymal stem cells. However, many circRNAs remain undiscovered, and their precise mechanisms remain elusive. Therefore, the objective of this study is to construct an osteogenic differentiation-related circRNA-miRNA-mRNA network in hBMSCs. Subsequently, through bioinformatics analysis, we constructed a ceRNA network related to the osteogenic differentiation ability of hBMSCs, comprising 22 circRNAs, 17 miRNAs, and 15 mRNAs. The potential circRNA-miRNA-mRNA axes, including the role of hsa_circ_0001600 in promoting the osteogenic differentiation of hBMSCs through the targeted regulation of hsa-miR-542-3p, were validated through in vitro experiments.
Collapse
Affiliation(s)
- Kaixin Su
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Xiangya Road, Changsha, 410008, Hunan, China
| | - Xinyan Cui
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Xiangya Road, Changsha, 410008, Hunan, China
| | - Jian Zhou
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China
- Department of VIP Dental Service, School of Stomatology, Capital Medical University, Beijing, 100050, China
- Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiao Yi
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Xiangya Road, Changsha, 410008, Hunan, China.
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Academician Workstation for Oral-Maxilofacial and Regenerative Medicine & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
6
|
Weaver SR, Torres HM, Arnold KM, Zars EL, Peralta-Herrera E, Taylor EL, Yu K, Marron Fernandez de Velasco E, Wickman K, McGee-Lawrence ME, Bradley EW, Westendorf JJ. Girk3 deletion increases osteoblast maturation and bone mass accrual in adult male mice. JBMR Plus 2024; 8:ziae108. [PMID: 39228688 PMCID: PMC11370632 DOI: 10.1093/jbmrpl/ziae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 09/05/2024] Open
Abstract
Osteoporosis and other metabolic bone diseases are prevalent in the aging population. While bone has the capacity to regenerate throughout life, bone formation rates decline with age and contribute to reduced bone density and strength. Identifying mechanisms and pathways that increase bone accrual in adults could prevent fractures and accelerate healing. G protein-gated inwardly rectifying K+ (GIRK) channels are key effectors of G protein-coupled receptor signaling. Girk3 was recently shown to regulate endochondral ossification. Here, we demonstrate that deletion of Girk3 increases bone mass after 18 weeks of age. Male 24-week-old Girk3 -/- mice have greater trabecular bone mineral density and bone volume fraction than wildtype (WT) mice. Osteoblast activity is moderately increased in 24-week-old Girk3 -/- mice compared to WT mice. In vitro, Girk3-/- bone marrow stromal cells (BMSCs) are more proliferative than WT BMSCs. Calvarial osteoblasts and BMSCs from Girk3 -/- mice are also more osteogenic than WT cells, with altered expression of genes that regulate the wingless-related integration site (Wnt) family. Wnt inhibition via Dickkopf-1 (Dkk1) or β-catenin inhibition via XAV939 prevents enhanced mineralization, but not proliferation, in Girk3 -/- BMSCs and slows these processes in WT cells. Finally, selective ablation of Girk3 from cells expressing Cre recombinase from the 2.3 kb-Col1a1 promoter, including osteoblasts and osteocytes, is sufficient to increase bone mass and bone strength in male mice at 24 weeks of age. Taken together, these data demonstrate that Girk3 regulates progenitor cell proliferation, osteoblast differentiation, and bone mass accrual in adult male mice.
Collapse
Affiliation(s)
- Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | - Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | - Elizabeth L Zars
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | | | | | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, United States
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, United States
| | - Elizabeth W Bradley
- Department of Orthopedics, School of Medicine, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
7
|
Guan Q, Zhang Y, Wang ZK, Liu XH, Zou J, Zhang LL. Skeletal phenotypes and molecular mechanisms in aging mice. Zool Res 2024; 45:724-746. [PMID: 38894518 PMCID: PMC11298674 DOI: 10.24272/j.issn.2095-8137.2023.397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/28/2024] [Indexed: 06/21/2024] Open
Abstract
Aging is an inevitable physiological process, often accompanied by age-related bone loss and subsequent bone-related diseases that pose serious health risks. Research on skeletal diseases caused by aging in humans is challenging due to lengthy study durations, difficulties in sampling, regional variability, and substantial investment. Consequently, mice are preferred for such studies due to their similar motor system structure and function to humans, ease of handling and care, low cost, and short generation time. In this review, we present a comprehensive overview of the characteristics, limitations, applicability, bone phenotypes, and treatment methods in naturally aging mice and prematurely aging mouse models (including SAMP6, POLG mutant, LMNA, SIRT6, ZMPSTE24, TFAM, ERCC1, WERNER, and KL/KL-deficient mice). We also summarize the molecular mechanisms of these aging mouse models, including cellular DNA damage response, senescence-related secretory phenotype, telomere shortening, oxidative stress, bone marrow mesenchymal stem cell (BMSC) abnormalities, and mitochondrial dysfunction. Overall, this review aims to enhance our understanding of the pathogenesis of aging-related bone diseases.
Collapse
Affiliation(s)
- Qiao Guan
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Yuan Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Zhi-Kun Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiao-Hua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Ling-Li Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China. E-mail:
| |
Collapse
|
8
|
Koh NYY, Miszkiewicz JJ, Fac ML, Wee NKY, Sims NA. Preclinical Rodent Models for Human Bone Disease, Including a Focus on Cortical Bone. Endocr Rev 2024; 45:493-520. [PMID: 38315213 PMCID: PMC11244217 DOI: 10.1210/endrev/bnae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/22/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Preclinical models (typically ovariectomized rats and genetically altered mice) have underpinned much of what we know about skeletal biology. They have been pivotal for developing therapies for osteoporosis and monogenic skeletal conditions, including osteogenesis imperfecta, achondroplasia, hypophosphatasia, and craniodysplasias. Further therapeutic advances, particularly to improve cortical strength, require improved understanding and more rigorous use and reporting. We describe here how trabecular and cortical bone structure develop, are maintained, and degenerate with aging in mice, rats, and humans, and how cortical bone structure is changed in some preclinical models of endocrine conditions (eg, postmenopausal osteoporosis, chronic kidney disease, hyperparathyroidism, diabetes). We provide examples of preclinical models used to identify and test current therapies for osteoporosis, and discuss common concerns raised when comparing rodent preclinical models to the human skeleton. We focus especially on cortical bone, because it differs between small and larger mammals in its organizational structure. We discuss mechanisms common to mouse and human controlling cortical bone strength and structure, including recent examples revealing genetic contributors to cortical porosity and osteocyte network configurations during growth, maturity, and aging. We conclude with guidelines for clear reporting on mouse models with a goal for better consistency in the use and interpretation of these models.
Collapse
Affiliation(s)
- Natalie Y Y Koh
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Justyna J Miszkiewicz
- School of Social Science, The University of Queensland, Brisbane, QLD 4072, Australia
- Vertebrate Evolution Development and Ecology, Naturalis Biodiversity Center, 2333 CR Leiden, The Netherlands
| | - Mary Louise Fac
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Natalie K Y Wee
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Natalie A Sims
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
9
|
Van Pee T, Martens DS, Alfano R, Engelen L, Sleurs H, Rasking L, Plusquin M, Nawrot TS. Cord Blood Proteomic Profiles, Birth Weight, and Early Life Growth Trajectories. JAMA Netw Open 2024; 7:e2411246. [PMID: 38743419 PMCID: PMC11094560 DOI: 10.1001/jamanetworkopen.2024.11246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/13/2024] [Indexed: 05/16/2024] Open
Abstract
Importance The cord blood proteome, a repository of proteins derived from both mother and fetus, might offer valuable insights into the physiological and pathological state of the fetus. However, its association with birth weight and growth trajectories early in life remains unexplored. Objective To identify cord blood proteins associated with birth weight and the birth weight ratio (BWR) and to evaluate the associations of these cord blood proteins with early growth trajectories. Design, Setting, and Participants This cohort study included 288 mother-child pairs from the ongoing prospective Environmental Influence on Early Aging birth cohort study. Newborns were recruited from East-Limburg Hospital in Genk, Belgium, between February 2010 and November 2017 and followed up until ages 4 to 6 years. Data were analyzed from February 2022 to September 2023. Main Outcomes and Measures The outcome of interest was the associations of 368 inflammatory-related cord blood proteins with birth weight or BWR and with early life growth trajectories (ie, rapid growth at age 12 months and weight, body mass index [BMI] z score, waist circumference, and overweight at age 4-6 years) using multiple linear regression models. The BWR was calculated by dividing the birth weight by the median birth weight of the population-specific reference growth curve, considering parity, sex, and gestational age. Results are presented as estimates or odds ratios (ORs) for each doubling in proteins. Results The sample included 288 infants (125 [43.4%] male; mean [SD] gestation age, 277.2 [11.6] days). The mean (SD) age of the child at the follow-up examination was 4.6 (0.4) years old. After multiple testing correction, there were significant associations of birth weight and BWR with 7 proteins: 2 positive associations: afamin (birth weight: coefficient, 341.16 [95% CI, 192.76 to 489.50]) and secreted frizzled-related protein 4 (SFRP4; birth weight: coefficient, 242.60 [95% CI, 142.77 to 342.43]; BWR: coefficient, 0.07 [95% CI, 0.04 to 0.10]) and 5 negative associations: cadherin EGF LAG 7-pass G-type receptor 2 (CELSR2; birth weight: coefficient, -237.52 [95% CI, -343.15 to -131.89]), ephrin type-A receptor 4 (EPHA4; birth weight: coefficient, -342.78 [95% CI, -463.10 to -222.47]; BWR: coefficient, -0.11 [95% CI, -0.14 to -0.07]), SLIT and NTRK-like protein 1 (SLITRK1; birth weight: coefficient, -366.32 [95% CI, -476.66 to -255.97]; BWR: coefficient, -0.11 [95% CI, -0.15 to -0.08]), transcobalamin-1 (TCN1; birth weight: coefficient, -208.75 [95% CI, -305.23 to -112.26]), and unc-5 netrin receptor D (UNC5D; birth weight: coefficient, -209.27 [95% CI, -295.14 to -123.40]; BWR: coefficient, -0.07 [95% CI, -0.09 to -0.04]). Further evaluation showed that 2 proteins were still associated with rapid growth at age 12 months (afamin: OR, 0.32 [95% CI, 0.11-0.88]; TCN1: OR, 2.44 [95% CI, 1.26-4.80]). At age 4 to 6 years, CELSR2, EPHA4, SLITRK1, and UNC5D were negatively associated with weight (coefficients, -1.33 to -0.68 kg) and body mass index z score (coefficients, -0.41 to -0.23), and EPHA4, SLITRK1, and UNC5D were negatively associated with waist circumference (coefficients, -1.98 to -0.87 cm). At ages 4 to 6 years, afamin (OR, 0.19 [95% CI, 0.05-0.70]) and SLITRK1 (OR, 0.32 [95% CI, 0.10-0.99]) were associated with lower odds for overweight. Conclusions and Relevance This cohort study found 7 cord blood proteins associated with birth weight and growth trajectories early in life. Overall, these findings suggest that stressors that could affect the cord blood proteome during pregnancy might have long-lasting associations with weight and body anthropometrics.
Collapse
Affiliation(s)
- Thessa Van Pee
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Dries S. Martens
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Rossella Alfano
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Liesa Engelen
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Hanne Sleurs
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Leen Rasking
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Michelle Plusquin
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Tim S. Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Public Health and Primary Care, Leuven University, Leuven, Belgium
| |
Collapse
|
10
|
Kim HY, Charton C, Shim JH, Lim SY, Kim J, Lee S, Ohn JH, Kim BK, Heo CY. Patient-Derived Organoids Recapitulate Pathological Intrinsic and Phenotypic Features of Fibrous Dysplasia. Cells 2024; 13:729. [PMID: 38727265 PMCID: PMC11083396 DOI: 10.3390/cells13090729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Fibrous dysplasia (FD) is a rare bone disorder characterized by the replacement of normal bone with benign fibro-osseous tissue. Developments in our understanding of the pathophysiology and treatment options are impeded by the lack of suitable research models. In this study, we developed an in vitro organotypic model capable of recapitulating key intrinsic and phenotypic properties of FD. Initially, transcriptomic profiling of individual cells isolated from patient lesional tissues unveiled intralesional molecular and cellular heterogeneity. Leveraging these insights, we established patient-derived organoids (PDOs) using primary cells obtained from patient FD lesions. Evaluation of PDOs demonstrated preservation of fibrosis-associated constituent cell types and transcriptional signatures observed in FD lesions. Additionally, PDOs retained distinct constellations of genomic and metabolic alterations characteristic of FD. Histological evaluation further corroborated the fidelity of PDOs in recapitulating important phenotypic features of FD that underscore their pathophysiological relevance. Our findings represent meaningful progress in the field, as they open up the possibility for in vitro modeling of rare bone lesions in a three-dimensional context and may signify the first step towards creating a personalized platform for research and therapeutic studies.
Collapse
Affiliation(s)
- Ha-Young Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Clémentine Charton
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam 13605, Republic of Korea; (C.C.); (J.K.); (S.L.)
| | - Jung Hee Shim
- Department of Research Administration Team, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - So Young Lim
- Department of Plastic and Reconstructive Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Jinho Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam 13605, Republic of Korea; (C.C.); (J.K.); (S.L.)
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Republic of Korea
| | - Sejoon Lee
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam 13605, Republic of Korea; (C.C.); (J.K.); (S.L.)
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Jung Hun Ohn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - Baek Kyu Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - Chan Yeong Heo
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| |
Collapse
|
11
|
Hordyjewska-Kowalczyk E, Wuyts W, Boeckx N, Verdonck A, Hendrickx G, Mortier G. RUNX2-related metaphyseal dysplasia with maxillary hypoplasia: A rare skeletal disorder resembling SFRP4-related Pyle disease. Clin Genet 2024; 105:434-439. [PMID: 38108099 DOI: 10.1111/cge.14474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Metaphyseal dysplasia with maxillary hypoplasia with or without brachydactyly (MDMHB) is an ultra-rare skeletal dysplasia caused by heterozygous intragenic RUNX2 duplications, comprising either exons 3 to 5 or exons 3 to 6 of RUNX2. In this study, we describe a 14-year-old Belgian boy with metaphyseal dysplasia with maxillary hypoplasia but without brachydactyly. Clinical and radiographic examination revealed mild facial dysmorphism, dental anomalies, enlarged clavicles, genua valga and metaphyseal flaring and thin cortices with an osteoporotic skeletal appearance. Exome sequencing led to the identification of a de novo heterozygous tandem duplication within RUNX2, encompassing exons 3 to 7. This duplication is larger than the ones previously reported in MDMHB cases since it extends into the C-terminal activation domain of RUNX2. We review previously reported cases with MDMHB and highlight the resemblance of this disorder with Pyle disease, which may be explained by intersecting molecular pathways between RUNX2 and sFRP4. This study expands our knowledge on the genotypic and phenotypic characteristics of MDMHB and the role of RUNX2 in rare bone disorders.
Collapse
Affiliation(s)
- Ewa Hordyjewska-Kowalczyk
- Laboratory for Skeletal Dysplasia Research, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Wim Wuyts
- Department of Medical Genetics, Antwerp University Hospital and University of Antwerp, Edegem, Belgium
| | - Nele Boeckx
- Department of Medical Genetics, Antwerp University Hospital and University of Antwerp, Edegem, Belgium
| | - An Verdonck
- Department of Oral Health Sciences - Orthodontics, KU Leuven, Leuven, Belgium
- Service of Dentistry, University Hospitals Leuven, Leuven, Belgium
| | - Gretl Hendrickx
- Laboratory for Skeletal Dysplasia Research, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Geert Mortier
- Laboratory for Skeletal Dysplasia Research, Department of Human Genetics, KU Leuven, Leuven, Belgium
- Centre for Human Genetics, University Hospital Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Lillis KV, Grinceviciute R, Diogenes A. Sex-specific nociceptor modulation of the apical periodontitis transcriptome. Front Mol Biosci 2024; 11:1338511. [PMID: 38404963 PMCID: PMC10884291 DOI: 10.3389/fmolb.2024.1338511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction: Apical periodontitis (AP) is a painful disease that develops quickly following dental infections and is primarily characterized by robust inflammation surrounding the tissues of the affected tooth, resulting in disruption of bone homeostasis and periradicular bone loss. Moreover, there are distinct clinical presentations, symptoms, and responses to AP treatment between male and female subjects, creating a desperate need to further understand the sex-specific mechanisms of AP. Methods: With the growing evidence that nociceptors modulate AP development, we utilized RNA sequencing in nociceptor-ablated (Nav1.8 cre+/-, diphtheria toxin Alox+/-) transgenic mice to study the nociceptor regulation of the periapical lesion transcriptome using a rodent model of AP in female mice over 14 days. Results: Overall, we found that female mice exhibit unique patterns of differentially expressed genes throughout AP infection compared to male mice and that the expression of these genes is regulated by nociceptors. Additionally, nociceptor ablation results in a more significant enrichment of biological processes related to immune responses earlier compared to cre-control (Nav1.8 cre+/-) females and greater expression of genes involved in inflammatory processes and osteolytic activity. Discussion: Therefore, while nociceptor ablation augments inflammatory and bone resorption responses in both males and females in a mouse model of AP, transcriptomic analyses demonstrate that the mechanisms through which nociceptors modulate AP are distinct between sexes. These studies will provide the foundation needed to study further mechanisms of sex differences in AP, an area with a desperate need for investigation to treat current AP patients. Understanding these mechanisms can ultimately inform treatment options to alleviate suffering for millions of patients suffering from AP.
Collapse
Affiliation(s)
| | | | - Anibal Diogenes
- Department of Endodontics, University of Texas Health at San Antonio, San Antonio, TX, United States
| |
Collapse
|
13
|
He X, Hu W, Zhang Y, Chen M, Ding Y, Yang H, He F, Gu Q, Shi Q. Cellular senescence in skeletal disease: mechanisms and treatment. Cell Mol Biol Lett 2023; 28:88. [PMID: 37891477 PMCID: PMC10612178 DOI: 10.1186/s11658-023-00501-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The musculoskeletal system supports the movement of the entire body and provides blood production while acting as an endocrine organ. With aging, the balance of bone homeostasis is disrupted, leading to bone loss and degenerative diseases, such as osteoporosis, osteoarthritis, and intervertebral disc degeneration. Skeletal diseases have a profound impact on the motor and cognitive abilities of the elderly, thus creating a major challenge for both global health and the economy. Cellular senescence is caused by various genotoxic stressors and results in permanent cell cycle arrest, which is considered to be the underlying mechanism of aging. During aging, senescent cells (SnCs) tend to aggregate in the bone and trigger chronic inflammation by releasing senescence-associated secretory phenotypic factors. Multiple signalling pathways are involved in regulating cellular senescence in bone and bone marrow microenvironments. Targeted SnCs alleviate age-related degenerative diseases. However, the association between senescence and age-related diseases remains unclear. This review summarises the fundamental role of senescence in age-related skeletal diseases, highlights the signalling pathways that mediate senescence, and discusses potential therapeutic strategies for targeting SnCs.
Collapse
Affiliation(s)
- Xu He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Wei Hu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Yuanshu Zhang
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214026, People's Republic of China
| | - Mimi Chen
- Department of Orthopedics, Children Hospital of Soochow University, No. 92 Zhongnan Street, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Yicheng Ding
- Xuzhou Medical University, 209 Copper Mountain Road, Xuzhou, 221004, People's Republic of China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Fan He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
| | - Qiaoli Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
| | - Qin Shi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214026, People's Republic of China.
| |
Collapse
|
14
|
Lee SM, Meyer MB, Benkusky NA, Pike JW. Genome-wide analyses of gene expression profile identify key genes and pathways involved in skeletal response to phosphate and 1,25-dihydroxyvitamin D 3 in vivo. J Steroid Biochem Mol Biol 2023; 232:106335. [PMID: 37245694 PMCID: PMC10527973 DOI: 10.1016/j.jsbmb.2023.106335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/11/2023] [Accepted: 05/25/2023] [Indexed: 05/30/2023]
Abstract
Phosphate (P) is an essential element involved in various biological actions, such as bone integrity, energy production, cell signaling and molecular component. P homeostasis is modulated by 4 main tissues; intestine, kidney, bone, and parathyroid gland, where 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), parathyroid hormone and fibroblast growth factor 23 (FGF23) are produced and/or have an influence. In bone, serum P level modulates the production of FGF23 which then controls not only P excretion but also vitamin D metabolism in kidney in an endocrine manner. The hormonally active form of vitamin D, 1,25(OH)2D3, also has a significant effect on skeletal cells via its receptor, the vitamin D receptor, to control gene expression which mediates bone metabolism as well as mineral homeostasis. In this study, we adopted RNA-seq analysis to understand genome-wide skeletal gene expression regulation in response to P and 1,25(OH)2D3. We examined lumbar 5 vertebrae from the mice that were fed P deficient diet for a week followed by an acute high P diet for 3, 6, and 24 h as well as mice treated with 1,25(OH)2D3 intraperitoneally for 6 h. Further identification and exploration of the genes regulated by P and 1,25(OH)2D3 showed that P dynamically modulates the expression of skeletal genes involved in various biological processes while 1,25(OH)2D3 regulates genes highly related to bone metabolism. Our in vivo data were then compared with in vitro data that we previously obtained, which suggests that the gene expression profiles presented in this report mainly represent those of osteocytes. Interestingly, it was found that even though the skeletal response to P is distinguished from that to 1,25(OH)2D3, both factors have an effect on Wnt signaling pathway to modulate bone homeostasis. Taken together, this report presents genome-wide data that provide a foundation to understand molecular mechanisms by which skeletal cells respond to P and 1,25(OH)2D3.
Collapse
Affiliation(s)
- Seong Min Lee
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Mark B Meyer
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nancy A Benkusky
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - J Wesley Pike
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
15
|
Maden M, Polvadore T, Polanco A, Barbazuk WB, Stanley E. Osteoderms in a mammal the spiny mouse Acomys and the independent evolution of dermal armor. iScience 2023; 26:106779. [PMID: 37378333 PMCID: PMC10291248 DOI: 10.1016/j.isci.2023.106779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/06/2023] [Accepted: 04/25/2023] [Indexed: 06/29/2023] Open
Abstract
Osteoderms are bony plates found in the skin of vertebrates, mostly commonly in reptiles where they have evolved independently multiple times, suggesting the presence of a gene regulatory network that is readily activated and inactivated. They are absent in birds and mammals except for the armadillo. However, we have discovered that in one subfamily of rodents, the Deomyinae, there are osteoderms in the skin of their tails. Osteoderm development begins in the proximal tail skin and is complete 6 weeks after birth. RNA sequencing has identified the gene networks involved in their differentiation. There is a widespread down-regulation of keratin genes and an up-regulation of osteoblast genes and a finely balanced expression of signaling pathways as the osteoderms differentiate. Future comparisons with reptilian osteoderms may allow us to understand how these structures have evolved and why they are so rare in mammals.
Collapse
Affiliation(s)
- Malcolm Maden
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Trey Polvadore
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Arod Polanco
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - W. Brad Barbazuk
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Edward Stanley
- Florida Museum of Natural History, University of Florida, Museum Road, Gainesville, FL 32611, USA
| |
Collapse
|
16
|
Bao K, Jiao Y, Xing L, Zhang F, Tian F. The role of wnt signaling in diabetes-induced osteoporosis. Diabetol Metab Syndr 2023; 15:84. [PMID: 37106471 PMCID: PMC10141960 DOI: 10.1186/s13098-023-01067-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023] Open
Abstract
Osteoporosis, a chronic complication of diabetes mellitus, is characterized by a reduction in bone mass, destruction of bone microarchitecture, decreased bone strength, and increased bone fragility. Because of its insidious onset, osteoporosis renders patients highly susceptible to pathological fractures, leading to increased disability and mortality rates. However, the specific pathogenesis of osteoporosis induced by chronic hyperglycemia has not yet been fully elucidated. But it is currently known that the disruption of Wnt signaling triggered by chronic hyperglycemia is involved in the pathogenesis of diabetic osteoporosis. There are two main types of Wnt signaling pathways, the canonical Wnt signaling pathway (β-catenin-dependent) and the non-canonical Wnt signaling pathway (non-β-catenin-dependent), both of which play an important role in regulating the balance between bone formation and bone resorption. Therefore, this review systematically describes the effects of abnormal Wnt pathway signaling on bone homeostasis under hyperglycemia, hoping to reveal the relationship between Wnt signaling and diabetic osteoporosis to further improve understanding of this disease.
Collapse
Affiliation(s)
- Kairan Bao
- Department of Integrated Traditional & Western Medicine, Affiliated hospital of North, China University of Science and Technology, Jianshe South Road 73, Tangshan, 063000, Hebei, People's Republic of China.
| | - Yinghua Jiao
- Department of Integrated Traditional & Western Medicine, Affiliated hospital of North, China University of Science and Technology, Jianshe South Road 73, Tangshan, 063000, Hebei, People's Republic of China
- North China University of Science and Technology, Bohai Road 21, Caofeidian Dis, Tangshan, 063210, Hebei, People's Republic of China
| | - Lei Xing
- Department of Integrated Traditional & Western Medicine, Affiliated hospital of North, China University of Science and Technology, Jianshe South Road 73, Tangshan, 063000, Hebei, People's Republic of China
| | - Fang Zhang
- Department of Integrated Traditional & Western Medicine, Affiliated hospital of North, China University of Science and Technology, Jianshe South Road 73, Tangshan, 063000, Hebei, People's Republic of China
| | - Faming Tian
- Department of Integrated Traditional & Western Medicine, Affiliated hospital of North, China University of Science and Technology, Jianshe South Road 73, Tangshan, 063000, Hebei, People's Republic of China
- North China University of Science and Technology, Bohai Road 21, Caofeidian Dis, Tangshan, 063210, Hebei, People's Republic of China
| |
Collapse
|
17
|
Luan Y, Wu S, Wang M, Pu Y, Zhao Q, Ma Y, Jiang L, He X. Identification of Critical Genes for Ovine Horn Development Based on Transcriptome during the Embryonic Period. BIOLOGY 2023; 12:biology12040591. [PMID: 37106791 PMCID: PMC10136283 DOI: 10.3390/biology12040591] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023]
Abstract
Horns, also known as headgear, are a unique structure of ruminants. As ruminants are globally distributed, the study of horn formation is critical not only for increasing our understanding of natural and sexual selection but also for the breeding of polled sheep breeds to facilitate modern sheep farming. Despite this, a significant number of the underlying genetic pathways in sheep horn remain unclear. In this study, to clarify the gene expression profile of horn buds and investigate the key genes in horn bud formation, RNA-sequencing (RNA-seq) technology was utilized to investigate differential gene expression in the horn buds and adjacent forehead skin of Altay sheep fetuses. There were only 68 differentially expressed genes (DEGs) identified, consisting of 58 up-regulated genes and 10 down-regulated genes. RXFP2 was differentially up-regulated in the horn buds and had the highest significance (p-value = 7.42 × 10-14). In addition, 32 DEGs were horn-related genes identified in previous studies, such as RXFP2, FOXL2, SFRP4, SFRP2, KRT1, KRT10, WNT7B, and WNT3. Further, Gene Ontology (GO) analysis showed that the DEGs were mainly enriched with regard to growth, development, and cell differentiation. Pathway analysis revealed that the Wnt signaling pathway may be responsible for horn development. Further, through combining the protein-protein interaction networks of the DEGs, it was found that the top five hub genes, namely, ACAN, SFRP2, SFRP4, WNT3, and WNT7B, were also associated with horn development. Our results suggest that only a few key genes, including RXFP2, are involved in bud formation. This study not only validates the expression of candidate genes identified at the transcriptome level in previous studies but also provides new possible marker genes for horn development, which may promote our understanding of the genetic mechanisms of horn formation.
Collapse
Affiliation(s)
- Yuanyuan Luan
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Shangjie Wu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Mingkun Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Yabin Pu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Qianjun Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Yuehui Ma
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Lin Jiang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Xiaohong He
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| |
Collapse
|
18
|
Brommage R, Liu J, Powell DR. Skeletal phenotypes in secreted frizzled-related protein 4 gene knockout mice mimic skeletal architectural abnormalities in subjects with Pyle's disease from SFRP4 mutations. Bone Res 2023; 11:9. [PMID: 36808149 PMCID: PMC9941579 DOI: 10.1038/s41413-022-00242-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/26/2022] [Accepted: 11/03/2022] [Indexed: 02/22/2023] Open
Abstract
Mutations in SFRP4 cause Pyle's bone disease with wide metaphyses and increased skeletal fragility. The WNT signaling pathway plays important roles in determining skeletal architecture and SFRP4 is a secreted Frizzled decoy receptor that inhibits WNT signaling. Seven cohorts of male and female Sfrp4 gene knockout mice, examined through 2 years of age, had a normal lifespan but showed cortical and trabecular bone phenotypes. Mimicking human Erlenmeyer flask deformities, bone cross-sectional areas were elevated 2-fold in the distal femur and proximal tibia but only 30% in femur and tibia shafts. Reduced cortical bone thickness was observed in the vertebral body, midshaft femur and distal tibia. Elevated trabecular bone mass and numbers were observed in the vertebral body, distal femur metaphysis and proximal tibia metaphysis. Midshaft femurs retained extensive trabecular bone through 2 years of age. Vertebral bodies had increased compressive strength, but femur shafts had reduced bending strength. Trabecular, but not cortical, bone parameters in heterozygous Sfrp4 mice were modestly affected. Ovariectomy resulted in similar declines in both cortical and trabecular bone mass in wild-type and Sfrp4 KO mice. SFRP4 is critical for metaphyseal bone modeling involved in determining bone width. Sfrp4 KO mice show similar skeletal architecture and bone fragility deficits observed in patients with Pyle's disease with SFRP4 mutations.
Collapse
Affiliation(s)
- Robert Brommage
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
- BoneGenomics, The Woodlands, TX, USA.
| | - Jeff Liu
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
- Biogen, Cambridge, MA, USA.
| | - David R Powell
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
| |
Collapse
|
19
|
Park H, Jo S, Jang MA, Choi SH, Kim TH. Dikkopf-1 promotes matrix mineralization of osteoblasts by regulating Ca +-CAMK2A- CREB1 pathway. BMB Rep 2022; 55:627-632. [PMID: 36229414 PMCID: PMC9813425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Indexed: 12/29/2022] Open
Abstract
Dickkopf-1 (DKK1) is a secreted protein that acts as an antagonist of the canonical WNT/β-catenin pathway, which regulates osteoblast differentiation. However, the role of DKK1 on osteoblast differentiation has not yet been fully clarified. Here, we investigate the functional role of DKK1 on osteoblast differentiation. Primary osteoprogenitor cells were isolated from human spinal bone tissues. To examine the role of DKK1 in osteoblast differentiation, we manipulated the expression of DKK1, and the cells were differentiated into mature osteoblasts. DKK1 overexpression in osteoprogenitor cells promoted matrix mineralization of osteoblast differentiation but did not promote matrix maturation. DKK1 increased Ca+ influx and activation of the Ca+/calmodulin-dependent protein kinase II Alpha (CAMK2A)-cAMP response element-binding protein 1 (CREB1) and increased translocation of p-CREB1 into the nucleus. In contrast, stable DKK1 knockdown in human osteosarcoma cell line SaOS2 exhibited reduced nuclear translocation of p-CREB1 and matrix mineralization. Overall, we suggest that manipulating DKK1 regulates the matrix mineralization of osteoblasts by Ca+-CAMK2A-CREB1, and DKK1 is a crucial gene for bone mineralization of osteoblasts. [BMB Reports 2022; 55(12): 627-632].
Collapse
Affiliation(s)
- Hyosun Park
- Hanyang University Institute for Rheumatology Research, Seoul 04763, Korea,Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Sungsin Jo
- Hanyang University Institute for Rheumatology Research, Seoul 04763, Korea
| | - Mi-Ae Jang
- Department of Laboratory Medicine and Genetics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Korea
| | - Sung Hoon Choi
- Department of Orthopedic Surgery, Hanyang University Seoul Hospital, Seoul 04763, Korea
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research, Seoul 04763, Korea,Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea,Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Korea,Corresponding author. Tel: +82-2-2290-9245; Fax: +82-2-2290-9253; E-mail:
| |
Collapse
|
20
|
Durham EL, Grey ZJ, Black L, Howie RN, Barth JL, Lee BS, Cray JJ. Sfrp4 expression in thyroxine treated calvarial cells. Life Sci 2022; 311:121158. [PMID: 36370870 PMCID: PMC9719041 DOI: 10.1016/j.lfs.2022.121158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/24/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
AIMS Evidence suggests alterations of thyroid hormone levels can disrupt normal bone development. Most data suggest the major targets of thyroid hormones to be the Htra1/Igf1 pathway. Recent discovery by our group suggests involvement of targets WNT pathway, specifically overexpression of antagonist Sfrp4 in the presence of exogenous thyroid hormone. MAIN METHODS Here we aimed to model these interactions in vitro using primary and isotype cell lines to determine if thyroid hormone drives increased Sfrp4 expression in cells relevant to craniofacial development. Transcriptional profiling, bioinformatics interrogation, protein and function analyses were used. KEY FINDINGS Affymetrix transcriptional profiling found Sfrp4 overexpression in primary cranial suture derived cells stimulated with thyroxine in vitro. Interrogation of the SFRP4 promoter identified multiple putative binding sites for thyroid hormone receptors. Experimentation with several cell lines demonstrated that thyroxine treatment induced Sfrp4 expression, demonstrating that Sfrp4 mRNA and protein levels are not tightly coupled. Transcriptional and protein analyses demonstrate thyroid hormone receptor binding to the proximal promoter of the target gene Sfrp4 in murine calvarial pre-osteoblasts. Functional analysis after thyroxine hormone stimulation for alkaline phosphatase activity shows that pre-osteoblasts increase alkaline phosphatase activity compared to other cell types, suggesting cell type susceptibility. Finally, we added recombinant SFRP4 to pre-osteoblasts in combination with thyroxine treatment and observed a significant decrease in alkaline phosphatase positivity. SIGNIFICANCE Taken together, these results suggest SFRP4 may be a key regulatory molecule that prevents thyroxine driven osteogenesis. These data corroborate clinical findings indicating a potential for SFRP4 as a diagnostic or therapeutic target for hyperostotic craniofacial disorders.
Collapse
Affiliation(s)
- Emily L Durham
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zachary J Grey
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Laurel Black
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - R Nicole Howie
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Jeremy L Barth
- Department of Regenerative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Beth S Lee
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - James J Cray
- Department of Biomedical Education and Anatomy, College of Medicine, The Ohio State University, Columbus, OH, USA; Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
21
|
Park H, Jo S, Jang MA, Choi SH, Kim TH. Dikkopf-1 promotes matrix mineralization of osteoblasts by regulating Ca +-CAMK2A- CREB1 pathway. BMB Rep 2022; 55:627-632. [PMID: 36229414 PMCID: PMC9813425 DOI: 10.5483/bmbrep.2022.55.12.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/02/2022] [Accepted: 10/07/2022] [Indexed: 11/03/2023] Open
Abstract
Dickkopf-1 (DKK1) is a secreted protein that acts as an antagonist of the canonical WNT/β-catenin pathway, which regulates osteoblast differentiation. However, the role of DKK1 on osteoblast differentiation has not yet been fully clarified. Here, we investigate the functional role of DKK1 on osteoblast differentiation. Primary osteoprogenitor cells were isolated from human spinal bone tissues. To examine the role of DKK1 in osteoblast differentiation, we manipulated the expression of DKK1, and the cells were differentiated into mature osteoblasts. DKK1 overexpression in osteoprogenitor cells promoted matrix mineralization of osteoblast differentiation but did not promote matrix maturation. DKK1 increased Ca+ influx and activation of the Ca+/calmodulin-dependent protein kinase II Alpha (CAMK2A)-cAMP response element-binding protein 1 (CREB1) and increased translocation of p-CREB1 into the nucleus. In contrast, stable DKK1 knockdown in human osteosarcoma cell line SaOS2 exhibited reduced nuclear translocation of p-CREB1 and matrix mineralization. Overall, we suggest that manipulating DKK1 regulates the matrix mineralization of osteoblasts by Ca+-CAMK2A-CREB1, and DKK1 is a crucial gene for bone mineralization of osteoblasts. [BMB Reports 2022; 55(12): 627-632].
Collapse
Affiliation(s)
- Hyosun Park
- Hanyang University Institute for Rheumatology Research, Seoul 04763, Korea
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Sungsin Jo
- Hanyang University Institute for Rheumatology Research, Seoul 04763, Korea
| | - Mi-Ae Jang
- Department of Laboratory Medicine and Genetics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Korea
| | - Sung Hoon Choi
- Department of Orthopedic Surgery, Hanyang University Seoul Hospital, Seoul 04763, Korea
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research, Seoul 04763, Korea
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Korea
| |
Collapse
|
22
|
Takaya K, Asou T, Kishi K. Downregulation of senescence-associated secretory phenotype by knockdown of secreted frizzled-related protein 4 contributes to the prevention of skin aging. Aging (Albany NY) 2022; 14:8167-8178. [PMID: 36084952 DOI: 10.18632/aging.204273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022]
Abstract
There is growing evidence that the appearance and texture of the skin that is altered during the aging process are considerably enhanced by the accumulation of senescent dermal fibroblasts. These senescent cells magnify aging via an inflammatory, histolytic, and senescence-associated secretory phenotype (SASP). Secreted frizzled-related protein 4 (SFRP4) was previously determined to be expressed in dermal fibroblasts of aging skin, and its increased expression has been shown to promote cellular senescence. However, its role in the SASP remains unknown. We found that SFRP4 was significantly expressed in p16ink4a-positive human skin fibroblasts and that treatment with recombinant SFRP4 promoted SASP and senescence, whereas siRNA knockdown of SFRP4 suppressed SASP. Furthermore, we found that knockdown of SFRP4 in mouse skin ameliorates age-related reduction of subcutaneous adipose tissue, panniculus carnosus muscle layer, and thinning and dispersion of collagen fibers. These findings suggest a potential candidate for the development of new skin rejuvenation therapies that suppress SASP.
Collapse
Affiliation(s)
- Kento Takaya
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Toru Asou
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| |
Collapse
|
23
|
Vlashi R, Zhang X, Wu M, Chen G. Wnt signaling: essential roles in osteoblast differentiation, bone metabolism and therapeutic implications for bone and skeletal disorders. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
24
|
Chen Y, Zhang Z, Jin W, Li Z, Bao C, He C, Guo Y, Li C. Integrative Analyses of Antler Cartilage Transcriptome and Proteome of Gansu Red Deer ( Cervus elaphus kansuensis) at Different Growth Stages. Animals (Basel) 2022; 12:934. [PMID: 35405922 PMCID: PMC8997108 DOI: 10.3390/ani12070934] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/22/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
The velvet antler is a unique model for cancer and regeneration research due to its periodic regeneration and rapid growth. Antler growth is mainly triggered by the growth center located in its tip, which consists of velvet skin, mesenchyme and cartilage. Among them, cartilage accounts for most of the growth center. We performed an integrative analysis of the antler cartilage transcriptome and proteome at different antler growth stages. RNA-seq results revealed 24,778 unigenes, 19,243 known protein-coding genes, and 5535 new predicted genes. Of these, 2722 were detected with differential expression patterns among 30 d, 60 d, and 90 d libraries, and 488 differentially expressed genes (DEGs) were screened at 30 d vs. 60 d and 60 d vs. 90 d but not at 30 d vs. 90 d. Proteomic data identified 1361 known proteins and 179 predicted novel proteins. Comparative analyses showed 382 differentially expressed proteins (DEPs), of which 16 had differential expression levels at 30 d vs. 60 d and 60 d vs. 90 d but not at 30 d vs. 90 d. An integrated analysis conducted for DEGs and DEPs showed that gene13546 and its coding protein protein13546 annotated in the Wnt signaling pathway may possess important bio-logical functions in rapid antler growth. This study provides in-depth characterization of candidate genes and proteins, providing further insights into the molecular mechanisms controlling antler development.
Collapse
Affiliation(s)
- Yanxia Chen
- College of Eco–Environment Engineering, Qinghai University, Xining 810016, China; (W.J.); (Z.L.); (C.B.); (C.H.)
| | - Zhenxiang Zhang
- Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China;
| | - Wenjie Jin
- College of Eco–Environment Engineering, Qinghai University, Xining 810016, China; (W.J.); (Z.L.); (C.B.); (C.H.)
| | - Zhaonan Li
- College of Eco–Environment Engineering, Qinghai University, Xining 810016, China; (W.J.); (Z.L.); (C.B.); (C.H.)
| | - Changhong Bao
- College of Eco–Environment Engineering, Qinghai University, Xining 810016, China; (W.J.); (Z.L.); (C.B.); (C.H.)
| | - Caixia He
- College of Eco–Environment Engineering, Qinghai University, Xining 810016, China; (W.J.); (Z.L.); (C.B.); (C.H.)
| | - Yuqin Guo
- Research Monitoring and Evaluation Center of Qinghai National Park, Xining 810016, China;
| | - Changzhong Li
- College of Eco–Environment Engineering, Qinghai University, Xining 810016, China; (W.J.); (Z.L.); (C.B.); (C.H.)
| |
Collapse
|
25
|
Kaya S, Schurman CA, Dole NS, Evans DS, Alliston T. Prioritization of Genes Relevant to Bone Fragility Through the Unbiased Integration of Aging Mouse Bone Transcriptomics and Human GWAS Analyses. J Bone Miner Res 2022; 37:804-817. [PMID: 35094432 PMCID: PMC9018503 DOI: 10.1002/jbmr.4516] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 12/20/2021] [Accepted: 01/17/2022] [Indexed: 11/10/2022]
Abstract
Identifying new genetic determinants of bone mineral density (BMD) and fracture promises to yield improved diagnostics and therapies for bone fragility. However, prioritizing candidate genes from genome-wide screens can be challenging. To overcome this challenge, we prioritized mouse genes that are differentially expressed in aging mouse bone based on whether their human homolog is associated with human BMD and/or fracture. Unbiased RNA-seq analysis of young and old male C57BL/6 mouse cortical bone identified 1499, 1685, and 5525 differentially expressed genes (DEGs) in 1, 2, and 2.5-year-old bone, relative to 2-month-old bone, respectively. Gene-based scores for heel ultrasound bone mineral density (eBMD) and fracture were estimated using published genome-wide association studies (GWAS) results of these traits in the UK Biobank. Enrichment analysis showed that mouse bone DEG sets for all three age groups, relative to young bone, are significantly enriched for eBMD, but only the oldest two DEG sets are enriched for fracture. Using gene-based scores, this approach prioritizes among thousands of DEGs by a factor of 5- to 100-fold, yielding 10 and 21 genes significantly associated with fracture in the two oldest groups of mouse DEGs. Though these genes were not the most differentially expressed, they included Sost, Lrp5, and others with well-established functions in bone. Several others have, as yet, unknown roles in the skeleton. Therefore, this study accelerates identification of new genetic determinants of bone fragility by prioritizing a clinically relevant and experimentally tractable number of candidate genes for functional analysis. Finally, we provide a website (www.mouse2human.org) to enable other researchers to easily apply our strategy. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
| | - Charles A. Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA
| | - Neha S. Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Periosteal apposition and endosteal remodeling regulate cortical bone expansion and thickness, both critical determinants of bone strength. Yet, the cellular characteristics and local or paracrine factors that regulate the periosteum and endosteum remain largely elusive. Here we discuss novel insights in cortical bone growth, expansion, and homeostasis, provided by the study of Secreted Frizzled Receptor Protein 4 (Sfrp4), a decoy receptor for Wnt ligands. RECENT FINDINGS SFRP4 loss-of function mutations cause Pyle disease, a rare skeletal disorder characterized by cortical bone thinning and increased fragility fractures despite increased trabecular bone density. On the endosteal surface, Sfrp4-mediated repression of non-canonical Wnt signaling regulates endosteal resorption. On the periosteum, Sfrp4 identifies as a critical functional mediator of periosteal stem cell/progenitor expansion and differentiation. Analysis of signaling pathways regulating skeletal stem cells/progenitors provides an opportunity to advance our understanding of the mechanisms involved in cortical bone biology.
Collapse
Affiliation(s)
- Ruiying Chen
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Roland Baron
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
- Endocrine Unit, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Francesca Gori
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
27
|
Yoon HS, Kim HY, Cho KA, Kim YH, Woo SY, Kim HS, Kang JL, Ryu KH, Park JW. Procollagen C-Endopeptidase Enhancer 2 Secreted by Tonsil-Derived Mesenchymal Stem Cells Increases the Oxidative Burst of Promyelocytic HL-60 Cells. BIOLOGY 2022; 11:biology11020255. [PMID: 35205121 PMCID: PMC8869569 DOI: 10.3390/biology11020255] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 12/19/2022]
Abstract
Simple Summary Tonsil-derived mesenchymal stem cells (TMSCs) improved the reactive oxygen species (ROS) production in human promyelocytic leukaemia cells (HL-60) differentiated into neutrophil-like cells (dHL-60). TMSC-induced enhancement of ROS generation in dHL-60 cells was different depending on the TMSC donor. Comparison of RNA-sequencing data between high and low potentiating TMSC groups for ROS generation in dHL-60 cells showed elevated expressions of four genes: secreted frizzled-related protein 4, mesenteric estrogen-dependent adipogenesis, microfibrillar associated protein 5, and procollagen C-endopeptidase enhancer 2 (PCOLCE2). Real-time PCR and Western blotting confirmed high levels of PCOLCE2 in the high potentiating TMSC group for ROS generation in dHL-60 cells. In addition, knockdown of PCOLCE2 in TMSCs reduced the enhancing efficacy of TMSCs regarding ROS generation in dHL-60 cells. Finally, treatment of recombinant PCOLCE2 protein augmented ROS production in dHL-60 cells with concomitant increases of NADPH oxidase (NOX) 3, NOX4, NOX5, and dual oxidase 2. Taken together, this study showed that PCOLCE2 levels in TMSCs could be used to select TMSCs with the high potentiating ability for ROS generation in neutrophils, and both TMSCs and PCOLCE2 may have the potential to enhance a frontline defence by increasing the efficiency of ROS generation in neutrophils. Abstract Reactive oxygen species (ROS) generated by neutrophils provide a frontline defence against invading pathogens. We investigated the supportive effect of tonsil-derived mesenchymal stem cells (TMSCs) on ROS generation from neutrophils using promyelocytic HL-60 cells. Methods: Differentiated HL-60 (dHL-60) cells were cocultured with TMSCs isolated from 25 independent donors, and ROS generation in dHL-60 cells was measured using luminescence. RNA sequencing and real-time PCR were performed to identify the candidate genes of TMSCs involved in augmenting the oxidative burst of dHL-60 cells. Transcriptome analysis of TMSCs derived from 25 independent donors revealed high levels of procollagen C-endopeptidase enhancer 2 (PCOLCE2) in TMSCs, which were highly effective in potentiating ROS generation in dHL-60 cells. In addition, PCOLCE2 knockdown in TMSCs abrogated TMSC-induced enhancement of ROS production in dHL-60 cells, indicating that TMSCs increased the oxidative burst in dHL-60 cells via PCOLCE2. Furthermore, the direct addition of recombinant PCOLCE2 protein increased ROS production in dHL-60 cells. These results suggest that PCOLCE2 secreted by TMSCs may be used as a therapeutic candidate to enhance host defences by increasing neutrophil oxidative bursts. PCOLCE2 levels in TMSCs could be used as a marker to select TMSCs exhibiting high efficacy for enhancing neutrophil oxidative bursts.
Collapse
Affiliation(s)
- Hee-Soo Yoon
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07084, Korea; (H.-S.Y.); (H.-Y.K.)
| | - Hee-Yeon Kim
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07084, Korea; (H.-S.Y.); (H.-Y.K.)
| | - Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07084, Korea; (K.-A.C.); (Y.-H.K.); (S.-Y.W.)
| | - Yu-Hee Kim
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07084, Korea; (K.-A.C.); (Y.-H.K.); (S.-Y.W.)
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07084, Korea; (K.-A.C.); (Y.-H.K.); (S.-Y.W.)
| | - Han-Su Kim
- Department of Otorhinolaryngology, College of Medicine, Ewha Womans University, Seoul 07985, Korea;
| | - Jihee-Lee Kang
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07084, Korea;
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07084, Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul 07084, Korea
- Correspondence: (K.-H.R.); (J.-W.P.); Tel.: +82-2-2653-3718 (K.-H.R.); +82-2-6986-6201 (J.-W.P.)
| | - Joo-Won Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07084, Korea; (H.-S.Y.); (H.-Y.K.)
- Correspondence: (K.-H.R.); (J.-W.P.); Tel.: +82-2-2653-3718 (K.-H.R.); +82-2-6986-6201 (J.-W.P.)
| |
Collapse
|
28
|
Li C, Liu K, Dai J, Li X, Liu X, Ni W, Li H, Wang D, Qiao J, Wang Y, Cui Y, Xia X, Hu S. Whole-genome resequencing to investigate the determinants of the multi-lumbar vertebrae trait in sheep. Gene 2022; 809:146020. [PMID: 34656743 DOI: 10.1016/j.gene.2021.146020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 10/03/2021] [Accepted: 10/11/2021] [Indexed: 12/25/2022]
Abstract
Multi-lumbar vertebrae trait is a beneficial mutation that can significantly improve livestock meat production. However, the genetic basis of the multi-lumbar vertebrae in sheep is still unclear. Here, we analysed the number of lumbar vertebrae of Duolang sheep and found three different traits of lumbar vertebrae number. Compared with the normal sheep, the length and weight of animal carcass from the multi-lumbar vertebrae sheep increased by 2.21 cm and 0.78 kg, respectively. We performed high-throughput genome resequencing on multi-lumbar vertebrae (n = 18) and normal (n = 11) Duolang sheep and obtained a total of more than 528.87 GB data. We found that the most significantly selective region were located in the 49.68-49.74 MB of chromosome 4 by selective-sweep analysis. We annotated this region and found that it contains SFRP4 which is known to regulate bone development. We further used the PCR-SSCP technology to detect the single nucleotide polymorphism (SNP) of the putative candidate SFRP4 and found that the two SNPs (rs600370085:C > T and rs415133338: A > G) of this gene were significantly associated with the multi-lumbar vertebrae of Duolang sheep. Our study indicates that the SFRP4 may be a potential major gene that affects the number of lumbar vertebrae in Duolang sheep, and has the potential to be utilized for sheep breeding in the future.
Collapse
Affiliation(s)
- Cunyuan Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China; College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Kaiping Liu
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Jihong Dai
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Xiaoyue Li
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Xia Liu
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Wei Ni
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Hui Li
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Dawei Wang
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Jun Qiao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Yue Wang
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yuying Cui
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Xianzhu Xia
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Shengwei Hu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China; College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, China.
| |
Collapse
|
29
|
Novel animal model of soft tissue tumor due to aberrant hedgehog signaling activation in pericyte lineage. Cell Tissue Res 2022; 388:63-73. [DOI: 10.1007/s00441-022-03578-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/10/2022] [Indexed: 11/02/2022]
|
30
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
31
|
Estrogen receptor alpha and NFATc1 bind to a bone mineral density-associated SNP to repress WNT5B in osteoblasts. Am J Hum Genet 2022; 109:97-115. [PMID: 34906330 DOI: 10.1016/j.ajhg.2021.11.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/17/2021] [Indexed: 12/27/2022] Open
Abstract
Genetic factors and estrogen deficiency contribute to the development of osteoporosis. The single-nucleotide polymorphism (SNP) rs2887571 is predicted from genome-wide association studies (GWASs) to associate with osteoporosis but has had an unknown mechanism. Analysis of osteoblasts from 110 different individuals who underwent joint replacement revealed that the genotype of rs2887571 correlates with WNT5B expression. Analysis of our ChIP-sequencing data revealed that SNP rs2887571 overlaps with an estrogen receptor alpha (ERα) binding site. Here we show that 17β-estradiol (E2) suppresses WNT5B expression and further demonstrate the mechanism of ERα binding at the enhancer containing rs2887571 to suppress WNT5B expression differentially in each genotype. ERα interacts with NFATc1, which is predicted to bind directly at rs2887571. CRISPR-Cas9 and ChIP-qPCR experiments confirm differential regulation of WNT5B between each allele. Homozygous GG has a higher binding affinity for ERα than homozygous AA and results in greater suppression of WNT5B expression. Functionally, WNT5B represses alkaline phosphatase expression and activity, decreasing osteoblast differentiation and mineralization. Furthermore, WNT5B increases interleukin-6 expression and suppresses E2-induced expression of alkaline phosphatase during osteoblast differentiation. We show that WNT5B suppresses the differentiation of osteoblasts via receptor tyrosine kinase-like orphan receptor 1/2 (ROR1/2), which activates DVL2/3/RAC1/CDC42/JNK/SIN3A signaling and inhibits β-catenin activity. Together, our data provide mechanistic insight into how ERα and NFATc1 regulate the non-coding SNP rs2887571, as well as the function of WNT5B on osteoblasts, which could provide alternative therapeutic targets for osteoporosis.
Collapse
|
32
|
Isojima T, Sims NA. Cortical bone development, maintenance and porosity: genetic alterations in humans and mice influencing chondrocytes, osteoclasts, osteoblasts and osteocytes. Cell Mol Life Sci 2021; 78:5755-5773. [PMID: 34196732 PMCID: PMC11073036 DOI: 10.1007/s00018-021-03884-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/06/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
Cortical bone structure is a crucial determinant of bone strength, yet for many years studies of novel genes and cell signalling pathways regulating bone strength have focused on the control of trabecular bone mass. Here we focus on mechanisms responsible for cortical bone development, growth, and degeneration, and describe some recently described genetic-driven modifications in humans and mice that reveal how these processes may be controlled. We start with embryonic osteogenesis of preliminary bone structures preceding the cortex and describe how this structure consolidates then matures to a dense, vascularised cortex containing an increasing proportion of lamellar bone. These processes include modelling-induced, and load-dependent, asymmetric cortical expansion, which enables the cortex's transition from a highly porous woven structure to a consolidated and thickened highly mineralised lamellar bone structure, infiltrated by vascular channels. Sex-specific differences emerge during this process. With aging, the process of consolidation reverses: cortical pores enlarge, leading to greater cortical porosity, trabecularisation and loss of bone strength. Each process requires co-ordination between bone formation, bone mineralisation, vascularisation, and bone resorption, with a need for locational-, spatial- and cell-specific signalling pathways to mediate this co-ordination. We will discuss these processes, and a number of cell-signalling pathways identified in both murine and human genetic studies to regulate cortical bone mass, including signalling through gp130, STAT3, PTHR1, WNT16, NOTCH, NOTUM and sFRP4.
Collapse
Affiliation(s)
- Tsuyoshi Isojima
- St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC, 3122, Australia
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC, 3122, Australia.
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia.
| |
Collapse
|
33
|
Choi JH, Wang Z, Ross FP, van der Meulen MCH, Bostrom MPG. Systemic osteoprotegerin does not improve peri-implant bone volume or osseointegration in rabbits. J Orthop Res 2021; 39:1611-1621. [PMID: 33049072 DOI: 10.1002/jor.24884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/05/2020] [Accepted: 09/29/2020] [Indexed: 02/04/2023]
Abstract
Anti-RANKL (receptor activator of nuclear factor kappa-B ligand) agents function by blocking the differentiation of osteoclasts, thereby proving useful in the clinical management of postmenopausal osteoporosis. The effects of such agents on osseointegration is less well understood. The purpose of the current study was to investigate whether osteoprotegerin (OPG), an osteoclast inhibitor, enhances the known anabolic effects of mechanical loading (VEH) and intermittent PTH (iPTH) using a well-established rabbit model of osseointegration. In the first set of experiments, OPG was administered either alone or combined with iPTH to study its effects on measured bone mass. The second set of experiments was conducted using a higher dosage of OPG (10 mg/kg) to explore its early impact at the cellular and molecular levels. All subjects had mechanical load applied to the implant on one extremity, and no load applied on the contralateral side. In the first set of experiments, OPG alone decreased peri-implant bone mass compared to the mechanical loading group, whereas OPG + iPTH increased peri-implant bone mass compared to the OPG group. In the second set of experiments, high-dose OPG significantly decreased osteoclast number (-74.3%) at 1 week. However, this effect was not sustained as osteoclast number returned to baseline by 2 weeks. These results suggest that systemic administration of OPG does not enhance osseointegration, but rather has a detrimental effect.
Collapse
Affiliation(s)
- Joseph H Choi
- Hospital for Special Surgery, New York, New York, USA
| | - Zhiwei Wang
- Hospital for Special Surgery, New York, New York, USA.,Orthopaedic Department, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | | | | | | |
Collapse
|
34
|
Pecorelli A, Cordone V, Schiavone ML, Caffarelli C, Cervellati C, Cerbone G, Gonnelli S, Hayek J, Valacchi G. Altered Bone Status in Rett Syndrome. Life (Basel) 2021; 11:life11060521. [PMID: 34205017 PMCID: PMC8230033 DOI: 10.3390/life11060521] [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: 05/10/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 11/24/2022] Open
Abstract
Rett syndrome (RTT) is a monogenic neurodevelopmental disorder primarily caused by mutations in X-linked MECP2 gene, encoding for methyl-CpG binding protein 2 (MeCP2), a multifaceted modulator of gene expression and chromatin organization. Based on the type of mutation, RTT patients exhibit a broad spectrum of clinical phenotypes with various degrees of severity. In addition, as a complex multisystem disease, RTT shows several clinical manifestations ranging from neurological to non-neurological symptoms. The most common non-neurological comorbidities include, among others, orthopedic complications, mainly scoliosis but also early osteopenia/osteoporosis and a high frequency of fractures. A characteristic low bone mineral density dependent on a slow rate of bone formation due to dysfunctional osteoblast activity rather than an increase in bone resorption is at the root of these complications. Evidence from human and animal studies supports the idea that MECP2 mutation could be associated with altered epigenetic regulation of bone-related factors and signaling pathways, including SFRP4/WNT/β-catenin axis and RANKL/RANK/OPG system. More research is needed to better understand the role of MeCP2 in bone homeostasis. Indeed, uncovering the molecular mechanisms underlying RTT bone problems could reveal new potential pharmacological targets for the treatment of these complications that adversely affect the quality of life of RTT patients for whom the only therapeutic approaches currently available include bisphosphonates, dietary supplements, and physical activity.
Collapse
Affiliation(s)
- Alessandra Pecorelli
- Animal Science Department, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC 28081, USA;
- Correspondence: (A.P.); (G.V.)
| | - Valeria Cordone
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Maria Lucia Schiavone
- Animal Science Department, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC 28081, USA;
| | - Carla Caffarelli
- Department of Medicine, Surgery and Neuroscience, University of Siena, Policlinico Le Scotte, 53100 Siena, Italy; (C.C.); (S.G.)
| | - Carlo Cervellati
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy;
| | - Gaetana Cerbone
- Division of Medical Genetics, “S.G. Moscati” Hospital, 74100 Avellino, Italy;
| | - Stefano Gonnelli
- Department of Medicine, Surgery and Neuroscience, University of Siena, Policlinico Le Scotte, 53100 Siena, Italy; (C.C.); (S.G.)
| | - Joussef Hayek
- Toscana Life Sciences Foundation, 53100 Siena, Italy;
| | - Giuseppe Valacchi
- Animal Science Department, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC 28081, USA;
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (A.P.); (G.V.)
| |
Collapse
|
35
|
Brommage R, Jeter-Jones S, Xiong W, Liu J. MicroCT analyses of mouse femoral neck architecture. Bone 2021; 145:115040. [PMID: 31437568 DOI: 10.1016/j.bone.2019.115040] [Citation(s) in RCA: 5] [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: 12/07/2018] [Revised: 07/24/2019] [Accepted: 08/17/2019] [Indexed: 10/26/2022]
Abstract
Hip fractures at the femoral neck are a major cause of morbidity and mortality, but aside from biomechanical strength testing, little is known about femoral neck architecture in mice. Procedures were optimized to analyze high-resolution (6 μm voxel size) microCT scans of the mouse femoral neck to provide bone mass and architectural information. Similar to histomorphometric observations in rats, the boundary between cortical and trabecular bone is difficult to identify in the mouse femoral mid-neck and these compartments were not analyzed separately. Analyses included total area, mineralized bone area, and bone volume fraction (BV/TV). Femoral neck architecture varies in C57BL/6J, 129/SvEv and BALB/c mouse strains. Bone cross sectional area and BV/TV were low in Lrp5 but elevated in Sost gene knockout mice. Sfrp4 gene knockout resulted in high total area, normal bone area, low BV/TV and, as indicated by BS/BV values, greater trabecularization. Femoral neck BV/TV declined with age and ovariectomy, but increased with teriparatide treatment. These findings demonstrate that the architecture of the mouse femoral neck mimics phenotypes and treatment effects observed at other skeletal sites and is a relevant bone site for translational studies examining osteoporosis therapies.
Collapse
Affiliation(s)
- Robert Brommage
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA.
| | | | - Wendy Xiong
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA
| | - Jeff Liu
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA
| |
Collapse
|
36
|
Cheng J, Gu W, Lan T, Deng J, Ni Z, Zhang Z, Hu Y, Sun X, Yang Y, Xu Q. Single-cell RNA sequencing reveals cell type- and artery type-specific vascular remodelling in male spontaneously hypertensive rats. Cardiovasc Res 2021; 117:1202-1216. [PMID: 32589721 PMCID: PMC7983007 DOI: 10.1093/cvr/cvaa164] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/08/2020] [Accepted: 06/18/2020] [Indexed: 01/09/2023] Open
Abstract
AIMS Hypertension is a major risk factor for cardiovascular diseases. However, vascular remodelling, a hallmark of hypertension, has not been systematically characterized yet. We described systematic vascular remodelling, especially the artery type- and cell type-specific changes, in hypertension using spontaneously hypertensive rats (SHRs). METHODS AND RESULTS Single-cell RNA sequencing was used to depict the cell atlas of mesenteric artery (MA) and aortic artery (AA) from SHRs. More than 20 000 cells were included in the analysis. The number of immune cells more than doubled in aortic aorta in SHRs compared to Wistar Kyoto controls, whereas an expansion of MA mesenchymal stromal cells (MSCs) was observed in SHRs. Comparison of corresponding artery types and cell types identified in integrated datasets unravels dysregulated genes specific for artery types and cell types. Intersection of dysregulated genes with curated gene sets including cytokines, growth factors, extracellular matrix (ECM), receptors, etc. revealed vascular remodelling events involving cell-cell interaction and ECM re-organization. Particularly, AA remodelling encompasses upregulated cytokine genes in smooth muscle cells, endothelial cells, and especially MSCs, whereas in MA, change of genes involving the contractile machinery and downregulation of ECM-related genes were more prominent. Macrophages and T cells within the aorta demonstrated significant dysregulation of cellular interaction with vascular cells. CONCLUSION Our findings provide the first cell landscape of resistant and conductive arteries in hypertensive animal models. Moreover, it also offers a systematic characterization of the dysregulated gene profiles with unbiased, artery type-specific and cell type-specific manners during hypertensive vascular remodelling.
Collapse
Affiliation(s)
- Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Wenduo Gu
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Ting Lan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Jiacheng Deng
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Zhichao Ni
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Zhongyi Zhang
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Yanhua Hu
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Xiaolei Sun
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
- Vascular Surgery Department, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou 646000, Sichuan, China
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Qingbo Xu
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China
| |
Collapse
|
37
|
Jiang H, Liu J, Fan C, Wang J, Li W. lncRNAS56464.1 as a ceRNA promotes the proliferation of fibroblast‑like synoviocytes in experimental arthritis via the Wnt signaling pathway and sponges miR‑152‑3p. Int J Mol Med 2021; 47:17. [PMID: 33448322 PMCID: PMC7834957 DOI: 10.3892/ijmm.2021.4850] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/04/2020] [Indexed: 12/23/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that occurs in approximately 1.0% of the general population. In RA patients, physical disability and joint damage are the major prognostic factors, which are associated with a reduction in the quality of life and early mortality. At present, the exact molecular mechanism of RA remains elusive. Long noncoding RNAs (lncRNAs) have been revealed to play a regulatory role in the pathogenesis of RA. To reveal the function of lncRNAs in rheumatoid arthritis, lncRNAS56464.1 was screened to verify its targeting of the microRNA (miR)-152-3p/Wnt pathway and its effect on the proliferation of fibroblast-like synoviocytes (FLS). In the present study, based on the competing endogenous RNA (ceRNA) theory, siRNA was designed for transfection into FLS to calculate the lncRNAS56464.1 interference efficiency and then the effect of lncRNAS56464.1 interference on FLS proliferation was detected by MTT assay. Then, lncRNAS56464.1 targeting of the miR-152-3p/Wnt pathway was detected by a dual-luciferase reporter assay. In addition, RT-qPCR, immunofluorescence and western blotting techniques were employed to detect the expression of lncRNAS56464.1, miR-152-3p and some key genes of the Wnt signaling pathway in FLS after lncRNAS56464.1 interference. The results revealed that lncRNAS56464.1 could combine with miR-152-3p and promoted the proliferation of FLS. In addition, lncRNAS56464.1 interference could not only decrease the proliferation of FLS and the expression of Wnt1, β-catenin, c-Myc, cyclin D1, and p-GSK-3β/GSK-3β, but it also increased the expression of SFRP4. The present data indicated that lncRNAS56464.1 could target the miR-152-3p/Wnt pathway to induce synovial cell proliferation and then participate in the pathogenesis of RA.
Collapse
Affiliation(s)
- Hui Jiang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Jian Liu
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Chang Fan
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Jing Wang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Weiping Li
- School of Basic Medical Science, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
38
|
Sowińska-Seidler A, Sztromwasser P, Zawadzka K, Sielski D, Bukowska-Olech E, Zawadzki P, Kozłowski K, Jamsheer A. The First Report of Biallelic Missense Mutations in the SFRP4 Gene Causing Pyle Disease in Two Siblings. Front Genet 2020; 11:593407. [PMID: 33193738 PMCID: PMC7646522 DOI: 10.3389/fgene.2020.593407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/30/2020] [Indexed: 12/28/2022] Open
Abstract
Background Pyle disease is a rare autosomal recessive bone dysplasia characterized by the broadening of metaphyses with generalized cortical thinning. Homozygous truncating mutations in secreted frizzled-related protein 4 (SFRP4) were, to date, the only known variants causative for this type of skeletal disorder. SFRP4 controls cortical and trabecular bone remodeling by differential regulation of the canonical and non-canonical WNT signaling in both bone compartments. Loss-of-function mutations in the SFRP4 gene lead to the protein deficiency causing skeletal phenotype typical for Pyle disease. Results Herein, we report on the first SFRP4 missense mutations that occurred in compound heterozygosity in two siblings affected by Pyle disease, and which we have identified using a whole-genome sequencing approach followed by a comprehensive in silico pathogenicity assessment. The variants we have found were extremely rare and evaluated to be disease-causing by several online available tools and software. Conclusion With this paper, we have shown that Pyle disease may be related not only to SFRP4 truncating mutations but also to other loss-of-function alterations that possibly impair the protein capacity to bind WNT ligands. As we have expanded here, the range of deleterious variants underlying Pyle disease, we contribute to the knowledge on the pathogenesis of this rare skeletal disorder.
Collapse
Affiliation(s)
- Anna Sowińska-Seidler
- Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Paweł Sztromwasser
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Łódź, Poland.,MNM Diagnostics, Poznan, Poland
| | | | | | | | - Paweł Zawadzki
- MNM Diagnostics, Poznan, Poland.,Molecular Biophysics Division, Faculty of Physics, A. Mickiewicz University, Poznan, Poland
| | - Kazimierz Kozłowski
- Department of Medical Imaging, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Poland.,Centers for Medical Genetics GENESIS, Poznan, Poland
| |
Collapse
|
39
|
Lee SJ, Lehar A, Meir JU, Koch C, Morgan A, Warren LE, Rydzik R, Youngstrom DW, Chandok H, George J, Gogain J, Michaud M, Stoklasek TA, Liu Y, Germain-Lee EL. Targeting myostatin/activin A protects against skeletal muscle and bone loss during spaceflight. Proc Natl Acad Sci U S A 2020; 117:23942-23951. [PMID: 32900939 PMCID: PMC7519220 DOI: 10.1073/pnas.2014716117] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Among the physiological consequences of extended spaceflight are loss of skeletal muscle and bone mass. One signaling pathway that plays an important role in maintaining muscle and bone homeostasis is that regulated by the secreted signaling proteins, myostatin (MSTN) and activin A. Here, we used both genetic and pharmacological approaches to investigate the effect of targeting MSTN/activin A signaling in mice that were sent to the International Space Station. Wild type mice lost significant muscle and bone mass during the 33 d spent in microgravity. Muscle weights of Mstn-/- mice, which are about twice those of wild type mice, were largely maintained during spaceflight. Systemic inhibition of MSTN/activin A signaling using a soluble form of the activin type IIB receptor (ACVR2B), which can bind each of these ligands, led to dramatic increases in both muscle and bone mass, with effects being comparable in ground and flight mice. Exposure to microgravity and treatment with the soluble receptor each led to alterations in numerous signaling pathways, which were reflected in changes in levels of key signaling components in the blood as well as their RNA expression levels in muscle and bone. These findings have implications for therapeutic strategies to combat the concomitant muscle and bone loss occurring in people afflicted with disuse atrophy on Earth as well as in astronauts in space, especially during prolonged missions.
Collapse
Affiliation(s)
- Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032;
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam Lehar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | - Jessica U Meir
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Christina Koch
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Andrew Morgan
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Lara E Warren
- Center for the Advancement of Science in Space, Houston, TX 77058
| | - Renata Rydzik
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, CT 06030
| | | | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | | | - Michael Michaud
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | | | - Yewei Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | - Emily L Germain-Lee
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030
- Connecticut Children's Center for Rare Bone Disorders, Farmington, CT 06032
| |
Collapse
|
40
|
Mashhadikhan M, Kheiri H, Dehghanifard A. DNA methylation and gene expression of sFRP2, sFRP4, Dkk 1, and Wif1 during osteoblastic differentiation of bone marrow derived mesenchymal stem cells. J Oral Biosci 2020; 62:349-356. [PMID: 32835781 DOI: 10.1016/j.job.2020.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Bone marrow derived mesenchymal stem cells (BMSCs) are an irresistible choice for use in stem cell therapy and regenerative medicine. BMSCs osteoblastic differentiation is also important in bone development, diseases, malignancies, and cancers studies. Wnt signaling pathway antagonists, Dickkopf-1 (Dkk 1), Secreted Frizzled-Related Proteins (sFRPs), and Wnt Inhibitory Factor 1 (Wif1) play important roles in inducing osteoblastic differentiation. This study is the first to investigate the association between DNA methylation and gene expression of Dkk1, sFRP2, sFRP4, and Wif1 during BMSCs osteoblastic differentiation. METHODS Human BMSCs were isolated and characterized using flow cytometry. Then, cells were treated with osteo-differentiation medium for three weeks. Alizarin red S staining and polymerase chain reaction (PCR) (alkaline phosphatase/osteocalcin) were performed for confirmation. The expression of Dkk 1, sFRP2, sFRP4, and Wif1 genes was evaluated at days 7, 14, and 21 using real-time PCR. Methylation-specific PCR (MSP) was performed to detect the methylation status of the promoters of the genes. RESULTS Data showed significant decreases (P < 0.05) during various days of BMSCs differentiation, while the promoters of the genes remained mostly un-methylated. CONCLUSIONS The down-regulation of Dkk 1, sFRP2, sFRP4, and Wif1 regulates various stages of human BMSCs during osteoblastic differentiation. DNA methylation does not interfere in the down-regulation of these genes, except for Wif1. We propose that the Wnt antagonist gene promoters should remain un-methylated during osteoblastic differentiation of BMSCs and that the down-regulation of these genes may contribute to other epigenetic mechanisms, other than DNA methylation, which implicitly indicates the role of DNA methylation in osteogenic cancers.
Collapse
Affiliation(s)
- Maedeh Mashhadikhan
- Department of Biology, Faculty of Sciences, Tehran Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Hamidreza Kheiri
- Cancer Gene Therapy Research Center, Zanjan University of Medical Science, Zanjan, Iran.
| | - Ali Dehghanifard
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
41
|
Kamizaki K, Endo M, Minami Y, Kobayashi Y. Role of noncanonical Wnt ligands and Ror-family receptor tyrosine kinases in the development, regeneration, and diseases of the musculoskeletal system. Dev Dyn 2020; 250:27-38. [PMID: 31925877 DOI: 10.1002/dvdy.151] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/26/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023] Open
Abstract
The Ror-family receptor tyrosine kinases (RTKs), consisting of Ror1 and Ror2, play crucial roles in morphogenesis and formation of various tissues/organs, including the bones and skeletal muscles, the so-called musculoskeletal system, during embryonic development, by acting as receptors or coreceptors for a noncanonical Wnt protein Wnt5a. Furthermore, several lines of evidence have indicated that Ror1 and/or Ror2 play critical roles in the regeneration and maintenance of the musculoskeletal system in adults. Considering the anatomical and functional relationship between the skeleton and skeletal muscles, their structural and functional association might be tightly regulated during their embryonic development, development after birth, and their regeneration after injury in adults. Importantly, in addition to their congenital anomalies, much attention has been paid onto the age-related disorders of the musculoskeletal system, including osteopenia and sarcopenia, which affect severely the quality of life. In this article, we overview recent advances in our understanding of the roles of Ror1- and/or Ror2-mediated signaling in the embryonic development, regeneration in adults, and congenital and age-related disorders of the musculoskeletal system and discuss possible therapeutic approaches to locomotive syndromes by modulating Ror1- and/or Ror2-mediated signaling.
Collapse
Affiliation(s)
- Koki Kamizaki
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Mitsuharu Endo
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | | |
Collapse
|
42
|
Yorgan TA, Sari H, Rolvien T, Windhorst S, Failla AV, Kornak U, Oheim R, Amling M, Schinke T. Mice lacking plastin-3 display a specific defect of cortical bone acquisition. Bone 2020; 130:115062. [PMID: 31678489 DOI: 10.1016/j.bone.2019.115062] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/20/2022]
Abstract
Although inactivating mutations of PLS3, encoding the actin-bundling protein plastin-3, have been identified to cause X-linked osteoporosis, the cellular and molecular influence of PLS3 on bone remodeling is poorly defined. Moreover, although a previous study has demonstrated moderate osteopenia in 12 week-old Pls3-deficient mice based on μCT scanning, there is no reported analysis of such a model on the basis of undecalcified histology and bone-specific histomorphometry. To fill this knowledge gap we applied a deep phenotyping approach and studied Pls3-deficient mice at different ages. Surprisingly, we did not detect significant differences between wildtype and Pls3-deficient littermates with respect to trabecular bone mass, and the same was the case for all histomorphometric parameters determined at 12 weeks of age. Remarkably however, the cortical thickness in both, tibia and femur, was significantly reduced in Pls3-deficient mice in all age groups. We additionally studied the ex vivo behavior of Pls3-deficient primary osteoblasts, which displayed moderately impaired mineralization capacity. Of note, while most osteoblastogenesis markers were not differentially expressed between wildtype and Pls3-deficient cultures, the expression of Sfrp4 was significantly reduced in the latter, a potentially relevant finding, since Sfrp4 inactivation, in mice and humans, specifically causes cortical thinning. We finally addressed the question, if Pls3-deficiency would impair the osteoanabolic influence of parathyroid hormone (PTH). For this purpose we applied daily injection of PTH into wildtype and Pls3-deficient mice and found a similar response regardless of the genotype. Taken together, our data reveal that Pls3-deficiency in mice only recapitulates the cortical bone phenotype of individuals with X-linked osteoporosis by negatively affecting the early stage of cortical bone acquisition.
Collapse
Affiliation(s)
- Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hatice Sari
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Antonio Virgilio Failla
- Microscopy Core Facility, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
43
|
Huybrechts Y, Mortier G, Boudin E, Van Hul W. WNT Signaling and Bone: Lessons From Skeletal Dysplasias and Disorders. Front Endocrinol (Lausanne) 2020; 11:165. [PMID: 32328030 PMCID: PMC7160326 DOI: 10.3389/fendo.2020.00165] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Skeletal dysplasias are a diverse group of heritable diseases affecting bone and cartilage growth. Throughout the years, the molecular defect underlying many of the diseases has been identified. These identifications led to novel insights in the mechanisms regulating bone and cartilage growth and homeostasis. One of the pathways that is clearly important during skeletal development and bone homeostasis is the Wingless and int-1 (WNT) signaling pathway. So far, three different WNT signaling pathways have been described, which are all activated by binding of the WNT ligands to the Frizzled (FZD) receptors. In this review, we discuss the skeletal disorders that are included in the latest nosology of skeletal disorders and that are caused by genetic defects involving the WNT signaling pathway. The number of skeletal disorders caused by defects in WNT signaling genes and the clinical phenotype associated with these disorders illustrate the importance of the WNT signaling pathway during skeletal development as well as later on in life to maintain bone mass. The knowledge gained through the identification of the genes underlying these monogenic conditions is used for the identification of novel therapeutic targets. For example, the genes underlying disorders with altered bone mass are all involved in the canonical WNT signaling pathway. Consequently, targeting this pathway is one of the major strategies to increase bone mass in patients with osteoporosis. In addition to increasing the insights in the pathways regulating skeletal development and bone homeostasis, knowledge of rare skeletal dysplasias can also be used to predict possible adverse effects of these novel drug targets. Therefore, this review gives an overview of the skeletal and extra-skeletal phenotype of the different skeletal disorders linked to the WNT signaling pathway.
Collapse
|
44
|
The Regulation of Bone Metabolism and Disorders by Wnt Signaling. Int J Mol Sci 2019; 20:ijms20225525. [PMID: 31698687 PMCID: PMC6888566 DOI: 10.3390/ijms20225525] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
Wnt, a secreted glycoprotein, has an approximate molecular weight of 40 kDa, and it is a cytokine involved in various biological phenomena including ontogeny, morphogenesis, carcinogenesis, and maintenance of stem cells. The Wnt signaling pathway can be classified into two main pathways: canonical and non-canonical. Of these, the canonical Wnt signaling pathway promotes osteogenesis. Sclerostin produced by osteocytes is an inhibitor of this pathway, thereby inhibiting osteogenesis. Recently, osteoporosis treatment using an anti-sclerostin therapy has been introduced. In this review, the basics of Wnt signaling, its role in bone metabolism and its involvement in skeletal disorders have been covered. Furthermore, the clinical significance and future scopes of Wnt signaling in osteoporosis, osteoarthritis, rheumatoid arthritis and neoplasia are discussed.
Collapse
|
45
|
Sfrp4 repression of the Ror2/Jnk cascade in osteoclasts protects cortical bone from excessive endosteal resorption. Proc Natl Acad Sci U S A 2019; 116:14138-14143. [PMID: 31239337 DOI: 10.1073/pnas.1900881116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Loss-of-function mutations in the Wnt inhibitor secreted frizzled receptor protein 4 (SFRP4) cause Pyle's disease (OMIM 265900), a rare skeletal disorder characterized by wide metaphyses, significant thinning of cortical bone, and fragility fractures. In mice, we have shown that the cortical thinning seen in the absence of Sfrp4 is associated with decreased periosteal and endosteal bone formation and increased endocortical resorption. While the increase in Rankl/Opg in cortical bone of mice lacking Sfrp4 suggests an osteoblast-dependent effect on endocortical osteoclast (OC) activity, whether Sfrp4 can cell-autonomously affect OCs is not known. We found that Sfrp4 is expressed during bone marrow macrophage OC differentiation and that Sfrp4 significantly suppresses the ability of early and late OC precursors to respond to Rankl-induced OC differentiation. Sfrp4 deletion in OCs resulted in activation of canonical Wnt/β-catenin and noncanonical Wnt/Ror2/Jnk signaling cascades. However, while inhibition of canonical Wnt/β-catenin signaling did not alter the effect of Sfrp4 on OCgenesis, blocking the noncanonical Wnt/Ror2/Jnk cascade markedly suppressed its regulation of OC differentiation in vitro. Importantly, we report that deletion of Ror2 exclusively in OCs (CtskCreRor2 fl/fl ) in Sfrp4 null mice significantly reversed the increased number of endosteal OCs seen in these mice and reduced their cortical thinning. Altogether, these data show autocrine and paracrine effects of Sfrp4 in regulating OCgenesis and demonstrate that the increase in endosteal OCs seen in Sfrp4 -/- mice is a consequence of noncanonical Wnt/Ror2/Jnk signaling activation in OCs overriding the negative effect that activation of canonical Wnt/β-catenin signaling has on OCgenesis.
Collapse
|
46
|
Ueland T, Stilgren L, Bollerslev J. Bone Matrix Levels of Dickkopf and Sclerostin are Positively Correlated with Bone Mass and Strength in Postmenopausal Osteoporosis. Int J Mol Sci 2019; 20:ijms20122896. [PMID: 31197079 PMCID: PMC6627473 DOI: 10.3390/ijms20122896] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 02/07/2023] Open
Abstract
Wnt signaling plays a pivotal role in maintaining bone mass. Secreted pathway modulators such as sclerostin (SOST) and Dickkopfs (DKKs) may influence bone mass inhibiting the canonical Wnt pathway. We evaluated whether bone protein content of secreted Wnt antagonists is related to age, bone mass, and strength in postmenopausal osteoporosis. We measured cortical and trabecular bone contents of SOST and Dickkopf-1 (DKK1) in combined extracts obtained after ethylenediaminetetraacetic acid and guanidine hydrochloride extraction in 56 postmenopausal women aged 47–74 (mean, 63) yr with a previous distal forearm fracture and a hip or spine Z-score less than 0. Our findings were (i) SOST and DKK1 protein levels were higher in trabecular bone, (ii) cortical and trabecular DKK1 and trabecular SOST correlated positively with bone matrix levels of osteocalcin (r between 0.28 and 0.45, p < 0.05), (iii) cortical DKK1 correlated with lumbar spine bone mineral density (BMD) (r = 0.32, p < 0.05) and femoral neck BMD (r = 0.41, p < 0.01), and (iv) cortical DKK1 and SOST correlated with apparent bone volumetric density and compressive strength (r between 0.34 and 0.51, p < 0.01). In conclusion, cortical bone matrix levels of DKK1 and SOST were positively correlated with bone mass and bone strength in postmenopausal osteoporotic women.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute for of Internal Medicine, Faculty of Medicine, University of Oslo, 0027 Oslo, Norway.
- KG Jebsen TREC, University of Tromsø, 9010 Tromsø, Norway.
| | - Lis Stilgren
- Department of Endocrinology, Svendborg Hospital, 5700 Svendborg, Denmark.
| | - Jens Bollerslev
- Section of Specialized Endocrinology, Oslo University Hospital; Faculty of Medicine, University of Oslo, 0027 Oslo, Norway.
| |
Collapse
|
47
|
Shen J, Zhao J, Ye QY, Gu XD. Interference of miR-943-3p with secreted frizzled-related proteins4 (SFRP4) in an asthma mouse model. Cell Tissue Res 2019; 378:67-80. [PMID: 31101982 DOI: 10.1007/s00441-019-03026-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 04/01/2019] [Indexed: 01/08/2023]
Abstract
The aim of this study is to investigate the potential roles of miR-943-3p and its target gene secreted frizzled-related proteins4 (SFRP4) in allergic asthma and elucidate its underlying mechanism, which may prompt a new clue about developing novel treatments of this disease. An allergic asthma mouse model was generated by challenging with ovalbumin (OVA); lung pathological features of mice were viewed using H&E staining; thickness of subepithelial fibrosis and smooth muscle was measured using Masson's trichrome staining. Inflammatory cells from bronchoalveolar lavage fluid (BALF) were counted based on Diff-Quik staining and morphometric analysis. Expressions of miR-943-3p, SFRP4 and Wnt signal pathway-associated proteins were detected using RT-PCR or immunoblotting, respectively. SFRP4 was downregulated in the bronchial biopsies of allergic asthma patients and represented a unique intersection between differentially expressed genes (DEGs) and genes in the Wnt signal pathway. Both miR-943-3p upregulation and SFRP4 downregulation were detected in allergic asthma patients and OVA-induced mice. Besides, OVA-induced mice possessed more inflammatory cells in BALF including macrophage (mac), eosinophil (eos), lymphocyte (lym) and neutrophil (neu), higher expression of collagen, β-catenin and c-Myc as well as thicker subepithelial fibrosis and smooth muscle in lung than control mice. In vivo delivery of miR-943-3p agomir worsened these symptoms, while both miR-943-3p antagomir and Ad-SFRP4 administration effectively alleviated this disease. Taken together, miR-943-3p accelerated the progression of airway inflammation and remodeling in allergic asthma via suppressing the activity of SFRP4 through Wnt signaling pathway in asthma patients and OVA-induced mice.
Collapse
Affiliation(s)
- Jian Shen
- Department of Pediatrics, Shuguang Hospital Affiliated to Shanghai Traditional Chinese Medical University, No. 528 Zhangheng Road, Pudong New Area, Shanghai, 201203, China.
| | - Jun Zhao
- Department of Pediatrics, Shuguang Hospital Affiliated to Shanghai Traditional Chinese Medical University, No. 528 Zhangheng Road, Pudong New Area, Shanghai, 201203, China
| | - Qing-Yan Ye
- Department of Pediatrics, Shuguang Hospital Affiliated to Shanghai Traditional Chinese Medical University, No. 528 Zhangheng Road, Pudong New Area, Shanghai, 201203, China
| | - Xi-Dong Gu
- Department of Clinical Laboratory, Shuguang Hospital Affiliated to Shanghai Traditional Chinese Medical University, Shanghai, 201203, China
| |
Collapse
|
48
|
Guo L, Bertola DR, Takanohashi A, Saito A, Segawa Y, Yokota T, Ishibashi S, Nishida Y, Yamamoto GL, Franco JFDS, Honjo RS, Kim CA, Musso CM, Timmons M, Pizzino A, Taft RJ, Lajoie B, Knight MA, Fischbeck KH, Singleton AB, Ferreira CR, Wang Z, Yan L, Garbern JY, Simsek-Kiper PO, Ohashi H, Robey PG, Boyde A, Matsumoto N, Miyake N, Spranger J, Schiffmann R, Vanderver A, Nishimura G, Passos-Bueno MRDS, Simons C, Ishikawa K, Ikegawa S. Bi-allelic CSF1R Mutations Cause Skeletal Dysplasia of Dysosteosclerosis-Pyle Disease Spectrum and Degenerative Encephalopathy with Brain Malformation. Am J Hum Genet 2019; 104:925-935. [PMID: 30982609 PMCID: PMC6507048 DOI: 10.1016/j.ajhg.2019.03.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/04/2019] [Indexed: 11/18/2022] Open
Abstract
Colony stimulating factor 1 receptor (CSF1R) plays key roles in regulating development and function of the monocyte/macrophage lineage, including microglia and osteoclasts. Mono-allelic mutations of CSF1R are known to cause hereditary diffuse leukoencephalopathy with spheroids (HDLS), an adult-onset progressive neurodegenerative disorder. Here, we report seven affected individuals from three unrelated families who had bi-allelic CSF1R mutations. In addition to early-onset HDLS-like neurological disorders, they had brain malformations and skeletal dysplasia compatible to dysosteosclerosis (DOS) or Pyle disease. We identified five CSF1R mutations that were homozygous or compound heterozygous in these affected individuals. Two of them were deep intronic mutations resulting in abnormal inclusion of intron sequences in the mRNA. Compared with Csf1r-null mice, the skeletal and neural phenotypes of the affected individuals appeared milder and variable, suggesting that at least one of the mutations in each affected individual is hypomorphic. Our results characterized a unique human skeletal phenotype caused by CSF1R deficiency and implied that bi-allelic CSF1R mutations cause a spectrum of neurological and skeletal disorders, probably depending on the residual CSF1R function.
Collapse
Affiliation(s)
- Long Guo
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan
| | - Débora Romeo Bertola
- Unidade de Genética Clínica, Instituto da Criança do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil; Instituto de Biociências da Universidade de São Paulo, São Paulo 05508-090, Brazil.
| | - Asako Takanohashi
- Division of Neurology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Asuka Saito
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Yuko Segawa
- Department of Orthopedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Satoru Ishibashi
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Yoichiro Nishida
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Guilherme Lopes Yamamoto
- Unidade de Genética Clínica, Instituto da Criança do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil; Instituto de Biociências da Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - José Francisco da Silva Franco
- Unidade de Genética Clínica, Instituto da Criança do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Rachel Sayuri Honjo
- Unidade de Genética Clínica, Instituto da Criança do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Chong Ae Kim
- Unidade de Genética Clínica, Instituto da Criança do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Camila Manso Musso
- Instituto de Biociências da Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Margaret Timmons
- Developmental and Metabolic Neurology Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Amy Pizzino
- Division of Neurology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan J Taft
- Illumina, Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | - Bryan Lajoie
- Illumina, Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | - Melanie A Knight
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Kenneth H Fischbeck
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute of Aging, NIH, Bethesda, MD 20892, USA
| | - Carlos R Ferreira
- Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA, and Division of Genetics and Metabolism, Children's National Health System, Washington, DC 20010, USA
| | - Zheng Wang
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan; Department of Medical Genetics, Institute of Basic Medical Sciences, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100005, People's Republic of China
| | - Li Yan
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - James Y Garbern
- Center of Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Pelin O Simsek-Kiper
- Department of Pediatrics, Hacettepe University Medical Faculty, Ankara 06100, Turkey
| | - Hirofumi Ohashi
- Division of Medical Genetics, Saitama Children's Medical Center, Saitama 330-8777, Japan
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD 20892, USA
| | - Alan Boyde
- Biophysics, Oral Growth and Development, Dental Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Jürgen Spranger
- Central German Competence Center for Rare Diseases (MKSE), Magdeburg 39120, Germany; Greenwood Genetic Center, Greenwood, SC 29646, USA
| | | | - Adeline Vanderver
- Division of Neurology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gen Nishimura
- Intractable Disease Center, Saitama University Hospital, Moro 350-0495, Japan
| | | | - Cas Simons
- Translational Bioinformatics Group, Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kinya Ishikawa
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan.
| |
Collapse
|
49
|
Chesi A, Wagley Y, Johnson ME, Manduchi E, Su C, Lu S, Leonard ME, Hodge KM, Pippin JA, Hankenson KD, Wells AD, Grant SFA. Genome-scale Capture C promoter interactions implicate effector genes at GWAS loci for bone mineral density. Nat Commun 2019; 10:1260. [PMID: 30890710 PMCID: PMC6425012 DOI: 10.1038/s41467-019-09302-x] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 03/05/2019] [Indexed: 12/21/2022] Open
Abstract
Osteoporosis is a devastating disease with an essential genetic component. GWAS have discovered genetic signals robustly associated with bone mineral density (BMD), but not the precise localization of effector genes. Here, we carry out physical and direct variant to gene mapping in human mesenchymal progenitor cell-derived osteoblasts employing a massively parallel, high resolution Capture C based method in order to simultaneously characterize the genome-wide interactions of all human promoters. By intersecting our Capture C and ATAC-seq data, we observe consistent contacts between candidate causal variants and putative target gene promoters in open chromatin for ~ 17% of the 273 BMD loci investigated. Knockdown of two novel implicated genes, ING3 at ‘CPED1-WNT16’ and EPDR1 at ‘STARD3NL’, inhibits osteoblastogenesis, while promoting adipogenesis. This approach therefore aids target discovery in osteoporosis, here on the example of two relevant genes involved in the fate determination of mesenchymal progenitors, and can be applied to other common genetic diseases. GWAS have identified numerous genetic loci for bone mineral density (BMD) and fracture risk. Here, the authors map these variants to putative target genes using ATAC-seq and Capture C of human osteoblasts and confirm ING3 and EPDR1 as BMD genes in in vitro osteoblast differentiation experiments.
Collapse
Affiliation(s)
- Alessandra Chesi
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA
| | - Yadav Wagley
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, 48109, MI, USA
| | - Matthew E Johnson
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA
| | - Elisabetta Manduchi
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA.,Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | - Chun Su
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA
| | - Sumei Lu
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA
| | - Michelle E Leonard
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA
| | - Kenyaita M Hodge
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA
| | - James A Pippin
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, 48109, MI, USA
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA. .,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA. .,Divisions of Genetics and Endocrinology, Children's Hospital of Philadelphia, Philadelphia, 19104, PA, USA.
| |
Collapse
|
50
|
Abstract
The group of sclerosing bone dysplasia's is a clinically and genetically heterogeneous group of rare bone disorders which, according to the latest Nosology and classification of genetic skeletal disorders (2015), can be subdivided in three subgroups; the neonatal osteosclerotic dysplasias, the osteopetroses and related disorders and the other sclerosing bone disorders. Here, we give an overview of the most important radiographic and clinical symptoms, the underlying genetic defect and potential treatment options of the different sclerosing dysplasias included in these subgroups.
Collapse
Affiliation(s)
- Eveline Boudin
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Wim Van Hul
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.
| |
Collapse
|