1
|
Liu Y, Lin Y, Lei Y, Xie W, Wei Y, Zhang H, Zhuang X, Cao H, Wang X. Early developmental anomalies in zebrafish (Danio rerio) embryos induced by the Clematis florida Thunb. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118897. [PMID: 39369922 DOI: 10.1016/j.jep.2024.118897] [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: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The C.florida. is one of the common medicines used by She population in China, with therapeutic effects of promoting blood circulation and anti-inflammatory. According to the acute toxicity grading standard of chemical substances, this herb is a low-toxicity herb. At present, the safety of C.florida., especially its impact on early embryonic development, is still unclear. AIM OF THE STUDY This study investigated the toxic effects of C. florida. on early embryonic development using a zebrafish embryo model. MATERIALS AND METHODS In this study, we used zebrafish embryos exposed to C.florida. at early stage to assess the early developmental toxicity by analyzing the developmental toxicity phenotype, oxidative stress, cell apoptosis, total enzyme activity, behavioral trajectory, and gene expression levels. RESULTS Embryos of the zebrafish exposed to different concentrations of C.florida. exhibited multiple organs and systems developmental disorders, including the heart, vessels, brain, bone, liver, and so on. Especially, with the increase of drug concentration, it is observed that the developmental malformations of the cardiovascular structure and function in larvae are becoming increasingly severe. In addition, results show that the abnormalities in embryonic development may be attributed to oxidative stress induced by apoptosis and activation of immune system resulting from an imbalance in the hematopoietic system. CONCLUSIONS This study provides a comprehensive and detailed summary of the toxic effects of C.florida. on embryonic development, which contributes to a deeper understanding of the potential adverse developmental consequences, and also prompt people to pay considerable attention to its treatment in medicinal practice.
Collapse
Affiliation(s)
- Yingying Liu
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350013, China; Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350011, China
| | - Yuan Lin
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350013, China; Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350011, China
| | - Yuqing Lei
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350013, China
| | - Wenpeng Xie
- Department of Cardiac Surgery, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350011, China
| | - Yalan Wei
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350013, China
| | - Haitao Zhang
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350013, China
| | - Xudong Zhuang
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350013, China
| | - Hua Cao
- Shengli Clinical Medical College of Fujian Medical University Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Xinrui Wang
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350013, China.
| |
Collapse
|
2
|
Masiero C, Aresi C, Forlino A, Tonelli F. Zebrafish Models for Skeletal and Extraskeletal Osteogenesis Imperfecta Features: Unveiling Pathophysiology and Paving the Way for Drug Discovery. Calcif Tissue Int 2024; 115:931-959. [PMID: 39320469 PMCID: PMC11607041 DOI: 10.1007/s00223-024-01282-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024]
Abstract
In the last decades, the easy genetic manipulation, the external fertilization, the high percentage of homology with human genes and the reduced husbandry costs compared to rodents, made zebrafish a valid model for studying human diseases and for developing new therapeutical strategies. Since zebrafish shares with mammals the same bone cells and ossification types, it became widely used to dissect mechanisms and possible new therapeutic approaches in the field of common and rare bone diseases, such as osteoporosis and osteogenesis imperfecta (OI), respectively. OI is a heritable skeletal disorder caused by defects in gene encoding collagen I or proteins/enzymes necessary for collagen I synthesis and secretion. Nevertheless, OI patients can be also characterized by extraskeletal manifestations such as dentinogenesis imperfecta, muscle weakness, cardiac valve and pulmonary abnormalities and skin laxity. In this review, we provide an overview of the available zebrafish models for both dominant and recessive forms of OI. An updated description of all the main similarities and differences between zebrafish and mammal skeleton, muscle, heart and skin, will be also discussed. Finally, a list of high- and low-throughput techniques available to exploit both larvae and adult OI zebrafish models as unique tools for the discovery of new therapeutic approaches will be presented.
Collapse
Affiliation(s)
- Cecilia Masiero
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Carla Aresi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy.
| | - Francesca Tonelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Via Taramelli 3B, 27100, Pavia, Italy
| |
Collapse
|
3
|
Kabir A, B M, A N, Selvaraj V, Sudhakar S. Protein Nano Coop Complexes Promote Fracture Healing and Bone Regeneration in a Zebrafish Osteoporosis Model. Biomacromolecules 2024; 25:7237-7248. [PMID: 39449233 DOI: 10.1021/acs.biomac.4c00931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Nanotherapeutic techniques are becoming increasingly important in the treatment of bone disorders owing to their targeted drug delivery. This study formulates zein nano coop composites containing chimeric antioxidants (ascorbic acid, luteolin, resveratrol, and coenzyme Q) (AZN) and evaluates its application in bone regeneration using osteoblasts and a Zebrafish osteoporosis model. In vitro experiments with human osteoblast-like MG63 cells showed enhancement of bone mineralization and regeneration. It further exhibited high biocompatibility in Zebrafish larvae, with increased calcium/phosphorus deposition and upregulation of osteogenic genes. The study has unequivocally demonstrated the potential of AZN in bone regeneration and fracture healing in both normal and osteoporosis models, underscoring the significance of this research. Further investigations using higher animal models are warranted to expand on these findings. The impact of this research seems far-reaching, with the possible development of new, effective, and safe treatment options for osteoporosis, addressing the limitations of the currently available treatments.
Collapse
Affiliation(s)
- Anisha Kabir
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Mukilarasi B
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Natarajan A
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Vimalraj Selvaraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| |
Collapse
|
4
|
Li Y, Selvaraj V, Saravanan S, Abullais SS, Wankhade V. Exploring the osteogenic potential of chitosan-quercetin bio-conjugate: In vitro and in vivo investigations in osteoporosis models. Int J Biol Macromol 2024; 274:133492. [PMID: 38944072 DOI: 10.1016/j.ijbiomac.2024.133492] [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: 02/09/2024] [Revised: 06/07/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Anti-osteoporotic agents are clinically employed to improve bone health and prevent osteoporotic fractures. In the current study, we investigated the potential of chitosan-quercetin bio-conjugate as an anti-osteoporotic agent. The conjugate was prepared and characterized by FTIR and found notable interactions between chitosan and quercetin. Treating mouse MSCs with the bioconjugate in osteogenic conditions for a week led to elevated expression of differentiation markers Runx2, ALP, and Col-I, as determined by real-time PCR analysis. Evaluation at the cellular level using alizarin red staining demonstrated enhanced calcium deposition in MSCs following treatment with the bioconjugate. Likewise, ELISA analysis showed significantly elevated levels of secretory osteocalcin and osteonectin in groups treated with the conjugate. To broaden our comprehension, we utilized a zebrafish-based in vivo model of dexamethasone-induced osteoporosis to investigate bone regeneration. Toxicity profiling with zebrafish larvae confirmed the bio-conjugate's compatibility at a concentration of 25 μg/ml, underscoring the significance of finding the right dosage. Furthermore, in zebrafish models of osteoporosis, the bio-conjugate demonstrated significant potential for bone regeneration, as indicated by improved bone calcification, callus formation, and overall bone healing in a tail fin fracture model. Additionally, the study revealed that the bio-conjugate inhibited osteoclastic activity, leading to reduced TRAP activity and hydroxyproline release, suggesting its effectiveness in mitigating bone resorption. In conclusion, our research provides compelling evidence for the osteogenic capabilities of the chitosan-quercetin bio-conjugate, highlighting its promising applications in regenerative medicine and the treatment of conditions like osteoporosis.
Collapse
Affiliation(s)
- Yi Li
- Department of Joint Surgery and Sports Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Vimalraj Selvaraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology-Madras, Chennai - 600 036, Tamil Nadu, India; Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600 077, Tamil Nadu, India.
| | - Sekaran Saravanan
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600 077, Tamil Nadu, India.
| | - Shahabe Saquib Abullais
- Department of Periodontics and Community Dental Science, King Khalid University, College of Dentistry, Abha, Saudi Arabia
| | - Varsha Wankhade
- Department of Zoology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
5
|
Kaliya-Perumal AK, Celik C, Carney TJ, Harris MP, Ingham PW. Genetic regulation of injury-induced heterotopic ossification in adult zebrafish. Dis Model Mech 2024; 17:dmm050724. [PMID: 38736327 DOI: 10.1242/dmm.050724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/05/2024] [Indexed: 05/14/2024] Open
Abstract
Heterotopic ossification is the inappropriate formation of bone in soft tissues of the body. It can manifest spontaneously in rare genetic conditions or as a response to injury, known as acquired heterotopic ossification. There are several experimental models for studying acquired heterotopic ossification from different sources of damage. However, their tenuous mechanistic relevance to the human condition, invasive and laborious nature and/or lack of amenability to chemical and genetic screens, limit their utility. To address these limitations, we developed a simple zebrafish injury model that manifests heterotopic ossification with high penetrance in response to clinically emulating injuries, as observed in human myositis ossificans traumatica. Using this model, we defined the transcriptional response to trauma, identifying differentially regulated genes. Mutant analyses revealed that an increase in the activity of the potassium channel Kcnk5b potentiates injury response, whereas loss of function of the interleukin 11 receptor paralogue (Il11ra) resulted in a drastically reduced ossification response. Based on these findings, we postulate that enhanced ionic signalling, specifically through Kcnk5b, regulates the intensity of the skeletogenic injury response, which, in part, requires immune response regulated by Il11ra.
Collapse
Affiliation(s)
- Arun-Kumar Kaliya-Perumal
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive 636921, Singapore
| | - Cenk Celik
- Department of Genetics, Evolution and Environment, Genetics Institute, University College London, London WC1E 6BT, UK
| | - Tom J Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive 636921, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos 138673, Singapore
| | - Matthew P Harris
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Department of Orthopedic Research, Boston Children's Hospital, Boston, MA 02115, USA
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive 636921, Singapore
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK
| |
Collapse
|
6
|
Sobah ML, Liongue C, Ward AC. Contribution of Signal Transducer and Activator of Transcription 3 (STAT3) to Bone Development and Repair. Int J Mol Sci 2023; 25:389. [PMID: 38203559 PMCID: PMC10778865 DOI: 10.3390/ijms25010389] [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: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcription factor activated canonically by numerous cytokines and other factors, with significant roles in immunity, immune diseases, and cancer. It has also been implicated in several human skeletal disorders, with loss-of-function (LOF) mutations associated with aberrant skeletal development. To gain further insights, two zebrafish STAT3 lines were investigated: a complete LOF knockout (KO) mutant and a partial LOF mutant with the transactivation domain truncated (ΔTAD). Consistent with other studies, the KO mutants were smaller, with reduced length in early embryos exacerbated by a decreased growth rate from 5 days postfertilization (dpf). They displayed skeletal deformities that approached 80% incidence by 30 dpf, with a significant reduction in early bone but not cartilage formation. Further analysis additionally identified considerable abrogation of caudal fin regeneration, concomitant with a paucity of infiltrating macrophages and neutrophils, which may be responsible for this. Most of these phenotypes were also observed in the ΔTAD mutants, indicating that loss of canonical STAT3 signaling was the likely cause. However, the impacts on early bone formation and regeneration were muted in the ΔTAD mutant, suggesting the potential involvement of noncanonical functions in these processes.
Collapse
Affiliation(s)
- Mohamed L. Sobah
- School of Medicine, Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| | - Clifford Liongue
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| | - Alister C. Ward
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia;
| |
Collapse
|
7
|
Ben-Zvi I, Karasik D, Ackert-Bicknell CL. Zebrafish as a Model for Osteoporosis: Functional Validations of Genome-Wide Association Studies. Curr Osteoporos Rep 2023; 21:650-659. [PMID: 37971665 DOI: 10.1007/s11914-023-00831-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW GWAS, as a largely correlational analysis, requires in vitro or in vivo validation. Zebrafish (Danio rerio) have many advantages for studying the genetics of human diseases. Since gene editing in zebrafish has been highly valuable for studying embryonic skeletal developmental processes that are prenatally or perinatally lethal in mammalian models, we are reviewing pros and cons of this model. RECENT FINDINGS The true power for the use of zebrafish is the ease by which the genome can be edited, especially using the CRISPR/Cas9 system. Gene editing, followed by phenotyping, for complex traits such as BMD, is beneficial, but the major physiological differences between the fish and mammals must be considered. Like mammals, zebrafish do have main bone cells; thus, both in vivo stem cell analyses and in vivo imaging are doable. Yet, the "long" bones of fish are peculiar, and their bone cavities do not contain bone marrow. Partial duplication of the zebrafish genome should be taken into account. Overall, small fish toolkit can provide unmatched opportunities for genetic modifications and morphological investigation as a follow-up to human-first discovery.
Collapse
Affiliation(s)
- Inbar Ben-Zvi
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - David Karasik
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| | | |
Collapse
|
8
|
Kague E, Medina-Gomez C, Boyadjiev SA, Rivadeneira F. The genetic overlap between osteoporosis and craniosynostosis. Front Endocrinol (Lausanne) 2022; 13:1020821. [PMID: 36225206 PMCID: PMC9548872 DOI: 10.3389/fendo.2022.1020821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
Osteoporosis is the most prevalent bone condition in the ageing population. This systemic disease is characterized by microarchitectural deterioration of bone, leading to increased fracture risk. In the past 15 years, genome-wide association studies (GWAS), have pinpointed hundreds of loci associated with bone mineral density (BMD), helping elucidate the underlying molecular mechanisms and genetic architecture of fracture risk. However, the challenge remains in pinpointing causative genes driving GWAS signals as a pivotal step to drawing the translational therapeutic roadmap. Recently, a skull BMD-GWAS uncovered an intriguing intersection with craniosynostosis, a congenital anomaly due to premature suture fusion in the skull. Here, we recapitulate the genetic contribution to both osteoporosis and craniosynostosis, describing the biological underpinnings of this overlap and using zebrafish models to leverage the functional investigation of genes associated with skull development and systemic skeletal homeostasis.
Collapse
Affiliation(s)
- Erika Kague
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, United Kingdom
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus Medical Center (MC), University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Simeon A. Boyadjiev
- Department of Pediatrics, University of California, Davis, Sacramento, CA, United States
| | - Fernando Rivadeneira
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center (MC), University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
9
|
Lin WY, Dharini KK, Peng CH, Lin CY, Yeh KT, Lee WC, Lin MD. Zebrafish models for glucocorticoid-induced osteoporosis. Tzu Chi Med J 2022; 34:373-380. [PMID: 36578638 PMCID: PMC9791848 DOI: 10.4103/tcmj.tcmj_80_22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/10/2022] [Accepted: 06/07/2022] [Indexed: 11/30/2022] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is the most common form of secondary osteoporosis due to excessive or long-term glucocorticoid administration, disturbing the homeostasis between bone formation and bone resorption. The bone biology of zebrafish shares a high degree of similarities with mammals. In terms of molecular level, genes and signaling pathways related to skeletogenesis are also highly correlated between zebrafish and humans. Therefore, zebrafish have been utilized to develop multiple GIOP models. Taking advantage of the transparency of zebrafish larvae, their skeletal development and bone mineralization can be readily visualized through in vivo staining without invasive experimental handlings. Moreover, the feasibility of using scales or fin rays to study bone remodeling makes adult zebrafish an ideal model for GIOP research. Here, we reviewed current zebrafish models for GIOP research, focused on the tools and methods established for examining bone homeostasis. As an in vivo, convenient, and robust model, zebrafish have an advantage in performing high-throughput drug screening and could be used to investigate the action mechanisms of therapeutic drugs.
Collapse
Affiliation(s)
- Wen-Ying Lin
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | | | - Cheng-Huan Peng
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,Institute of Medical Science, Tzu Chi University, Hualien, Taiwan,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Kuang-Ting Yeh
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Chih Lee
- Research Center for Global SDGs Challenges, Office of Research and Development, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Wen-Chih Lee, Research Center for Global SDGs Challenges, Office of Research and Development, Tzu Chi University, 701, Zhongyang Road, Section 3, Hualien, Taiwan. E-mail:
Prof. Ming-Der Lin, Department of Molecular Biology and Human Genetics, Tzu Chi University, 701, Zhongyang Road, Section 3, Hualien, Taiwan. E-mail:
| | - Ming-Der Lin
- Institute of Medical Science, Tzu Chi University, Hualien, Taiwan,Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Wen-Chih Lee, Research Center for Global SDGs Challenges, Office of Research and Development, Tzu Chi University, 701, Zhongyang Road, Section 3, Hualien, Taiwan. E-mail:
Prof. Ming-Der Lin, Department of Molecular Biology and Human Genetics, Tzu Chi University, 701, Zhongyang Road, Section 3, Hualien, Taiwan. E-mail:
| |
Collapse
|
10
|
Rosa JT, Tarasco M, Gavaia PJ, Cancela ML, Laizé V. Screening of Mineralogenic and Osteogenic Compounds in Zebrafish—Tools to Improve Assay Throughput and Data Accuracy. Pharmaceuticals (Basel) 2022; 15:ph15080983. [PMID: 36015130 PMCID: PMC9412667 DOI: 10.3390/ph15080983] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/24/2022] [Accepted: 08/03/2022] [Indexed: 12/16/2022] Open
Abstract
Bone disorders affect millions of people worldwide and treatments currently available often produce undesirable secondary effects or have limited efficacy. It is therefore of the utmost interest for patients to develop more efficient drugs with reduced off-target activities. In the long process of drug development, screening and preclinical validation have recently gained momentum with the increased use of zebrafish as a model organism to study pathological processes related to human bone disorders, and the development of zebrafish high-throughput screening assays to identify bone anabolic compounds. In this review, we provided a comprehensive overview of the literature on zebrafish bone-related assays and evaluated their performance towards an integration into screening pipelines for the discovery of mineralogenic/osteogenic compounds. Tools available to standardize fish housing and feeding procedures, synchronize embryo production, and automatize specimen sorting and image acquisition/analysis toward faster and more accurate screening outputs were also presented.
Collapse
Affiliation(s)
- Joana T. Rosa
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
- S2AQUA—Collaborative Laboratory, Association for a Sustainable and Smart Aquaculture, 8700-194 Olhão, Portugal
| | - Marco Tarasco
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
| | - Paulo J. Gavaia
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal
- GreenColab—Associação Oceano Verde, University of Algarve, 8005-139 Faro, Portugal
| | - M. Leonor Cancela
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, 8005-139 Faro, Portugal
| | - Vincent Laizé
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
- S2AQUA—Collaborative Laboratory, Association for a Sustainable and Smart Aquaculture, 8700-194 Olhão, Portugal
- Correspondence:
| |
Collapse
|
11
|
Ethiraj LP, Fong ELS, Liu R, Chan M, Winkler C, Carney TJ. Colorimetric and fluorescent TRAP assays for visualising and quantifying fish osteoclast activity. Eur J Histochem 2022; 66. [PMID: 35330553 PMCID: PMC8992378 DOI: 10.4081/ejh.2022.3369] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/08/2022] [Indexed: 11/22/2022] Open
Abstract
Histochemical detection of tartrate-resistant acid phosphatase (TRAP) activity is a fundamental technique for visualizing osteoclastic bone resorption and assessing osteoclast activity status in tissues. This approach has mostly employed colorimetric detection, which has limited quantification of activity in situ and co-labelling with other skeletal markers. Here, we report simple colorimetric and fluorescent TRAP assays in zebrafish and medaka, two important model organisms for investigating the pathogenesis of bone disorders. We show fluorescent TRAP staining, utilising the ELF97 substrate, is a rapid, robust, and stable system to visualise and quantify osteoclast activity in zebrafish, and is compatible with other fluorescence stains, transgenic lines and antibody approaches. Using this approach, we show that TRAP activity is predominantly found around the base of the zebrafish pharyngeal teeth, where osteoclast activity state appears to be heterogeneous.
Collapse
Affiliation(s)
| | | | - Ranran Liu
- Department of Biological Sciences, National University of Singapore.
| | - Madelynn Chan
- Lee Kong Chian School of Medicine, Nanyang Technological University; Department of Rheumatology, Allergy and Immunology, Tan Tock Seng Hospital.
| | - Christoph Winkler
- Department of Biological Sciences and Centre for Bioimaging Sciences, National University of Singapore.
| | - Tom James Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University; Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research).
| |
Collapse
|
12
|
Houck EL, Cohen EB, Harms CA, Runde BJ, Rudershausen PJ, Buckel JA. Computed Tomographic Assessment of Hooking-Related Injuries in Recreationally Angled Blue Marlin. JOURNAL OF AQUATIC ANIMAL HEALTH 2022; 34:20-27. [PMID: 34738678 DOI: 10.1002/aah.10145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/16/2021] [Accepted: 09/17/2021] [Indexed: 06/13/2023]
Abstract
Acute morbidity and mortality of marlins (family Istiophoridae) in hook-and-line fisheries have been studied; however, there has been little or no investigation of the skeletal injuries incurred from terminal tackles that could lead to decreased rates of postrelease survival. The objective of this study was to evaluate skeletal injuries in recreationally angled Atlantic Blue Marlin Makaira nigricans from the 2019 Big Rock Blue Marlin Tournament in Morehead City, North Carolina. We examined heads of six Blue Marlin that were angled using artificial lures rigged with J-hooks and harvested for weigh-in. The head of each Blue Marlin was scanned using computed tomography (CT) and examined with gross dissection. The CT interpretation revealed that two Blue Marlin had minimally displaced fractures of the maxilla, one of which also had a fracture to the lachrymal bone. These radiographic lesions were associated with penetrating hook injuries. The CT images also revealed degenerative changes within the quadrate-articular joint in four Blue Marlin, which was associated with fish weight; the causes and consequences of these degenerative changes are unknown. Although the hooking-related jaw fractures likely result in acute pain, their impact on postrelease morbidity is unknown and the impact on postrelease mortality is suspected to be small.
Collapse
Affiliation(s)
- Emma L Houck
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, North Carolina, 27606, USA
| | - Eli B Cohen
- Department of Molecular and Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, North Carolina, 27606, USA
| | - Craig A Harms
- Department of Clinical Sciences, Center for Marine Sciences and Technology, College of Veterinary Medicine, North Carolina State University, 303 College Circle, Morehead City, North Carolina, 28557, USA
| | - Brendan J Runde
- Department of Applied Ecology, Center for Marine Sciences and Technology, North Carolina State University, 303 College Circle, Morehead City, North Carolina, 28557, USA
| | - Paul J Rudershausen
- Department of Applied Ecology, Center for Marine Sciences and Technology, North Carolina State University, 303 College Circle, Morehead City, North Carolina, 28557, USA
| | - Jeffrey A Buckel
- Department of Applied Ecology, Center for Marine Sciences and Technology, North Carolina State University, 303 College Circle, Morehead City, North Carolina, 28557, USA
| |
Collapse
|
13
|
Kague E, Karasik D. Functional Validation of Osteoporosis Genetic Findings Using Small Fish Models. Genes (Basel) 2022; 13:279. [PMID: 35205324 PMCID: PMC8872034 DOI: 10.3390/genes13020279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/11/2022] Open
Abstract
The advancement of human genomics has revolutionized our understanding of the genetic architecture of many skeletal diseases, including osteoporosis. However, interpreting results from human association studies remains a challenge, since index variants often reside in non-coding regions of the genome and do not possess an obvious regulatory function. To bridge the gap between genetic association and causality, a systematic functional investigation is necessary, such as the one offered by animal models. These models enable us to identify causal mechanisms, clarify the underlying biology, and apply interventions. Over the past several decades, small teleost fishes, mostly zebrafish and medaka, have emerged as powerful systems for modeling the genetics of human diseases. Due to their amenability to genetic intervention and the highly conserved genetic and physiological features, fish have become indispensable for skeletal genomic studies. The goal of this review is to summarize the evidence supporting the utility of Zebrafish (Danio rerio) for accelerating our understanding of human skeletal genomics and outlining the remaining gaps in knowledge. We provide an overview of zebrafish skeletal morphophysiology and gene homology, shedding light on the advantages of human skeletal genomic exploration and validation. Knowledge of the biology underlying osteoporosis through animal models will lead to the translation into new, better and more effective therapeutic approaches.
Collapse
Affiliation(s)
- Erika Kague
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK;
| | - David Karasik
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
14
|
Cyphert EL, Zhang N, Learn GD, Hernandez CJ, von Recum HA. Recent Advances in the Evaluation of Antimicrobial Materials for Resolution of Orthopedic Implant-Associated Infections In Vivo. ACS Infect Dis 2021; 7:3125-3160. [PMID: 34761915 DOI: 10.1021/acsinfecdis.1c00465] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
While orthopedic implant-associated infections are rare, revision surgeries resulting from infections incur considerable healthcare costs and represent a substantial research area clinically, in academia, and in industry. In recent years, there have been numerous advances in the development of antimicrobial strategies for the prevention and treatment of orthopedic implant-associated infections which offer promise to improve the limitations of existing delivery systems through local and controlled release of antimicrobial agents. Prior to translation to in vivo orthopedic implant-associated infection models, the properties (e.g., degradation, antimicrobial activity, biocompatibility) of the antimicrobial materials can be evaluated in subcutaneous implant in vivo models. The antimicrobial materials are then incorporated into in vivo implant models to evaluate the efficacy of using the material to prevent or treat implant-associated infections. Recent technological advances such as 3D-printing, bacterial genomic sequencing, and real-time in vivo imaging of infection and inflammation have contributed to the development of preclinical implant-associated infection models that more effectively recapitulate the clinical presentation of infections and improve the evaluation of antimicrobial materials. This Review highlights the advantages and limitations of antimicrobial materials used in conjunction with orthopedic implants for the prevention and treatment of orthopedic implant-associated infections and discusses how these materials are evaluated in preclinical in vivo models. This analysis serves as a resource for biomaterial researchers in the selection of an appropriate orthopedic implant-associated infection preclinical model to evaluate novel antimicrobial materials.
Collapse
Affiliation(s)
- Erika L. Cyphert
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Ningjing Zhang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Greg D. Learn
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Christopher J. Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York 14853, United States
- Hospital for Special Surgery, New York, New York 10021, United States
| | - Horst A. von Recum
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
15
|
Foessl I, Bassett JHD, Bjørnerem Å, Busse B, Calado Â, Chavassieux P, Christou M, Douni E, Fiedler IAK, Fonseca JE, Hassler E, Högler W, Kague E, Karasik D, Khashayar P, Langdahl BL, Leitch VD, Lopes P, Markozannes G, McGuigan FEA, Medina-Gomez C, Ntzani E, Oei L, Ohlsson C, Szulc P, Tobias JH, Trajanoska K, Tuzun Ş, Valjevac A, van Rietbergen B, Williams GR, Zekic T, Rivadeneira F, Obermayer-Pietsch B. Bone Phenotyping Approaches in Human, Mice and Zebrafish - Expert Overview of the EU Cost Action GEMSTONE ("GEnomics of MusculoSkeletal traits TranslatiOnal NEtwork"). Front Endocrinol (Lausanne) 2021; 12:720728. [PMID: 34925226 PMCID: PMC8672201 DOI: 10.3389/fendo.2021.720728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
A synoptic overview of scientific methods applied in bone and associated research fields across species has yet to be published. Experts from the EU Cost Action GEMSTONE ("GEnomics of MusculoSkeletal Traits translational Network") Working Group 2 present an overview of the routine techniques as well as clinical and research approaches employed to characterize bone phenotypes in humans and selected animal models (mice and zebrafish) of health and disease. The goal is consolidation of knowledge and a map for future research. This expert paper provides a comprehensive overview of state-of-the-art technologies to investigate bone properties in humans and animals - including their strengths and weaknesses. New research methodologies are outlined and future strategies are discussed to combine phenotypic with rapidly developing -omics data in order to advance musculoskeletal research and move towards "personalised medicine".
Collapse
Affiliation(s)
- Ines Foessl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| | - J. H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Åshild Bjørnerem
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
- Norwegian Research Centre for Women’s Health, Oslo University Hospital, Oslo, Norway
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Ângelo Calado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | | | - Maria Christou
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Eleni Douni
- Institute for Bioinnovation, Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece
- Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Imke A. K. Fiedler
- Department of Osteology and Biomechanics, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Eva Hassler
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University Graz, Graz, Austria
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Erika Kague
- The School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, United Kingdom
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Patricia Khashayar
- Center for Microsystems Technology, Imec and Ghent University, Ghent, Belgium
| | - Bente L. Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Victoria D. Leitch
- Innovative Manufacturing Cooperative Research Centre, Royal Melbourne Institute of Technology, School of Engineering, Carlton, VIC, Australia
| | - Philippe Lopes
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Georgios Markozannes
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
| | | | | | - Evangelia Ntzani
- Department of Hygiene and Epidemiology, Medical School, University of Ioannina, Ioannina, Greece
- Department of Health Services, Policy and Practice, Center for Research Synthesis in Health, School of Public Health, Brown University, Providence, RI, United States
| | - Ling Oei
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pawel Szulc
- INSERM UMR 1033, University of Lyon, Lyon, France
| | - Jonathan H. Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School, Bristol, University of Bristol, Bristol, United Kingdom
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Şansın Tuzun
- Physical Medicine & Rehabilitation Department, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Amina Valjevac
- Department of Human Physiology, School of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Bert van Rietbergen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Tatjana Zekic
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | | | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| |
Collapse
|
16
|
Rauner M, Foessl I, Formosa MM, Kague E, Prijatelj V, Lopez NA, Banerjee B, Bergen D, Busse B, Calado Â, Douni E, Gabet Y, Giralt NG, Grinberg D, Lovsin NM, Solan XN, Ostanek B, Pavlos NJ, Rivadeneira F, Soldatovic I, van de Peppel J, van der Eerden B, van Hul W, Balcells S, Marc J, Reppe S, Søe K, Karasik D. Perspective of the GEMSTONE Consortium on Current and Future Approaches to Functional Validation for Skeletal Genetic Disease Using Cellular, Molecular and Animal-Modeling Techniques. Front Endocrinol (Lausanne) 2021; 12:731217. [PMID: 34938269 PMCID: PMC8686830 DOI: 10.3389/fendo.2021.731217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/30/2021] [Indexed: 12/26/2022] Open
Abstract
The availability of large human datasets for genome-wide association studies (GWAS) and the advancement of sequencing technologies have boosted the identification of genetic variants in complex and rare diseases in the skeletal field. Yet, interpreting results from human association studies remains a challenge. To bridge the gap between genetic association and causality, a systematic functional investigation is necessary. Multiple unknowns exist for putative causal genes, including cellular localization of the molecular function. Intermediate traits ("endophenotypes"), e.g. molecular quantitative trait loci (molQTLs), are needed to identify mechanisms of underlying associations. Furthermore, index variants often reside in non-coding regions of the genome, therefore challenging for interpretation. Knowledge of non-coding variance (e.g. ncRNAs), repetitive sequences, and regulatory interactions between enhancers and their target genes is central for understanding causal genes in skeletal conditions. Animal models with deep skeletal phenotyping and cell culture models have already facilitated fine mapping of some association signals, elucidated gene mechanisms, and revealed disease-relevant biology. However, to accelerate research towards bridging the current gap between association and causality in skeletal diseases, alternative in vivo platforms need to be used and developed in parallel with the current -omics and traditional in vivo resources. Therefore, we argue that as a field we need to establish resource-sharing standards to collectively address complex research questions. These standards will promote data integration from various -omics technologies and functional dissection of human complex traits. In this mission statement, we review the current available resources and as a group propose a consensus to facilitate resource sharing using existing and future resources. Such coordination efforts will maximize the acquisition of knowledge from different approaches and thus reduce redundancy and duplication of resources. These measures will help to understand the pathogenesis of osteoporosis and other skeletal diseases towards defining new and more efficient therapeutic targets.
Collapse
Affiliation(s)
- Martina Rauner
- Department of Medicine III, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- University Hospital Carl Gustav Carus, Dresden, Germany
| | - Ines Foessl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| | - Melissa M. Formosa
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Erika Kague
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Vid Prijatelj
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- The Generation R Study, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nerea Alonso Lopez
- Rheumatology and Bone Disease Unit, CGEM, Institute of Genetics and Cancer (IGC), Edinburgh, United Kingdom
| | - Bodhisattwa Banerjee
- Musculoskeletal Genetics Laboratory, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Dylan Bergen
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ângelo Calado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Eleni Douni
- Department of Biotechnology, Agricultural University of Athens, Athens, Greece
- Institute for Bioinnovation, B.S.R.C. “Alexander Fleming”, Vari, Greece
| | - Yankel Gabet
- Department of Anatomy & Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Natalia García Giralt
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - Nika M. Lovsin
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Xavier Nogues Solan
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - Barbara Ostanek
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Nathan J. Pavlos
- Bone Biology & Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | | | - Ivan Soldatovic
- Institute of Medical Statistics and Informatic, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Bram van der Eerden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Wim van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Susanna Balcells
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - Janja Marc
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Sjur Reppe
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
- Marcus Research Institute, Hebrew SeniorLife, Boston, MA, United States
| |
Collapse
|
17
|
Marí-Beffa M, Mesa-Román AB, Duran I. Zebrafish Models for Human Skeletal Disorders. Front Genet 2021; 12:675331. [PMID: 34490030 PMCID: PMC8418114 DOI: 10.3389/fgene.2021.675331] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/08/2021] [Indexed: 12/17/2022] Open
Abstract
In 2019, the Nosology Committee of the International Skeletal Dysplasia Society provided an updated version of the Nosology and Classification of Genetic Skeletal Disorders. This is a reference list of recognized diseases in humans and their causal genes published to help clinician diagnosis and scientific research advances. Complementary to mammalian models, zebrafish has emerged as an interesting species to evaluate chemical treatments against these human skeletal disorders. Due to its versatility and the low cost of experiments, more than 80 models are currently available. In this article, we review the state-of-art of this “aquarium to bedside” approach describing the models according to the list provided by the Nosology Committee. With this, we intend to stimulate research in the appropriate direction to efficiently meet the actual needs of clinicians under the scope of the Nosology Committee.
Collapse
Affiliation(s)
- Manuel Marí-Beffa
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, IBIMA, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, Málaga, Spain
| | - Ana B Mesa-Román
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, IBIMA, Málaga, Spain
| | - Ivan Duran
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, IBIMA, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, Málaga, Spain
| |
Collapse
|
18
|
Formosa MM, Bergen DJM, Gregson CL, Maurizi A, Kämpe A, Garcia-Giralt N, Zhou W, Grinberg D, Ovejero Crespo D, Zillikens MC, Williams GR, Bassett JHD, Brandi ML, Sangiorgi L, Balcells S, Högler W, Van Hul W, Mäkitie O. A Roadmap to Gene Discoveries and Novel Therapies in Monogenic Low and High Bone Mass Disorders. Front Endocrinol (Lausanne) 2021; 12:709711. [PMID: 34539568 PMCID: PMC8444146 DOI: 10.3389/fendo.2021.709711] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022] Open
Abstract
Genetic disorders of the skeleton encompass a diverse group of bone diseases differing in clinical characteristics, severity, incidence and molecular etiology. Of particular interest are the monogenic rare bone mass disorders, with the underlying genetic defect contributing to either low or high bone mass phenotype. Extensive, deep phenotyping coupled with high-throughput, cost-effective genotyping is crucial in the characterization and diagnosis of affected individuals. Massive parallel sequencing efforts have been instrumental in the discovery of novel causal genes that merit functional validation using in vitro and ex vivo cell-based techniques, and in vivo models, mainly mice and zebrafish. These translational models also serve as an excellent platform for therapeutic discovery, bridging the gap between basic science research and the clinic. Altogether, genetic studies of monogenic rare bone mass disorders have broadened our knowledge on molecular signaling pathways coordinating bone development and metabolism, disease inheritance patterns, development of new and improved bone biomarkers, and identification of novel drug targets. In this comprehensive review we describe approaches to further enhance the innovative processes taking discoveries from clinic to bench, and then back to clinic in rare bone mass disorders. We highlight the importance of cross laboratory collaboration to perform functional validation in multiple model systems after identification of a novel disease gene. We describe the monogenic forms of rare low and high rare bone mass disorders known to date, provide a roadmap to unravel the genetic determinants of monogenic rare bone mass disorders using proper phenotyping and genotyping methods, and describe different genetic validation approaches paving the way for future treatments.
Collapse
Affiliation(s)
- Melissa M. Formosa
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Dylan J. M. Bergen
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
- The Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Celia L. Gregson
- The Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Antonio Maurizi
- Department of Applied Clinical Sciences and Biotechnological, University of L’Aquila, L’Aquila, Italy
| | - Anders Kämpe
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Natalia Garcia-Giralt
- IMIM (Hospital del Mar Research Institute), Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Barcelona, Spain
| | - Wei Zhou
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Diana Ovejero Crespo
- IMIM (Hospital del Mar Research Institute), Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Barcelona, Spain
| | - M. Carola Zillikens
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - J. H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Maria Luisa Brandi
- Department of Surgery and Translational Medicine (M.L.B.), University of Florence, Florence, Italy
| | - Luca Sangiorgi
- Department of Medical Genetics and Skeletal Rare Diseases, IRCCS Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Susanna Balcells
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Outi Mäkitie
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Centre, Folkhälsan Institute of Genetics, Helsinki, Finland
| |
Collapse
|
19
|
Stevenson NL, Bergen DJM, Lu Y, Prada-Sanchez ME, Kadler KE, Hammond CL, Stephens DJ. Giantin is required for intracellular N-terminal processing of type I procollagen. J Cell Biol 2021; 220:212045. [PMID: 33944912 PMCID: PMC8103548 DOI: 10.1083/jcb.202005166] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 01/22/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022] Open
Abstract
Knockout of the golgin giantin leads to skeletal and craniofacial defects driven by poorly studied changes in glycosylation and extracellular matrix deposition. Here, we sought to determine how giantin impacts the production of healthy bone tissue by focusing on the main protein component of the osteoid, type I collagen. Giantin mutant zebrafish accumulate multiple spontaneous fractures in their caudal fin, suggesting their bones may be more brittle. Inducing new experimental fractures revealed defects in the mineralization of newly deposited collagen as well as diminished procollagen reporter expression in mutant fish. Analysis of a human giantin knockout cell line expressing a GFP-tagged procollagen showed that procollagen trafficking is independent of giantin. However, our data show that intracellular N-propeptide processing of pro-α1(I) is defective in the absence of giantin. These data demonstrate a conserved role for giantin in collagen biosynthesis and extracellular matrix assembly. Our work also provides evidence of a giantin-dependent pathway for intracellular procollagen processing.
Collapse
Affiliation(s)
- Nicola L Stevenson
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Dylan J M Bergen
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK.,Musculoskeletal Research Unit, Translational Health Sciences, University of Bristol, Bristol, UK.,Bristol Medical School, Faculty of Health Sciences, University of Bristol, Southmead Hospital, Bristol, UK
| | - Yinhui Lu
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - M Esther Prada-Sanchez
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, UK.,School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Karl E Kadler
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - David J Stephens
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
20
|
Rao S, Pham TH, Poudyal S, Cheng LW, Nazareth SC, Wang PC, Chen SC. First report on genetic characterization, cell-surface properties and pathogenicity of Lactococcus garvieae, emerging pathogen isolated from cage-cultured cobia (Rachycentron canadum). Transbound Emerg Dis 2021; 69:1197-1211. [PMID: 33759359 DOI: 10.1111/tbed.14083] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/24/2021] [Accepted: 03/19/2021] [Indexed: 11/29/2022]
Abstract
The diseased cage-cultured cobia (Rachycentron canadum) displayed clinical signs, haemorrhagic eyes, dorsal darkness and gross pathological lesions, enlargement of spleen and liver. Haemorrhages were found in brain, heart and liver with cumulative mortality rates ranging from 20% to 50%. Extensive congestion in the heart, liver, spleen, kidney and brain was observed histopathologically. Epicarditis and meningitis were also revealed in diseased cobia. All isolates recovered from the organs (liver, spleen, head kidney, posterior kidney, brain and muscle) of cobia were found to be gram-positive, non-motile, ovoid cocci, short-chain-forming (diplococci) and α-haemolytic. The API 32 strep system together with the polymerase chain reaction assay for species-specific primers (pLG1 and pLG2) and the internal transcribed spacer (ITS) region (G1 and L1 primers) confirmed all four selected isolates as Lactococcus garvieae. Partial 16S rDNA nucleotide sequence (~1,100 bp) of one representative L. garvieae isolate AOD109191 (GenBank accession number, MW328528.1) shared 99.9% identities with the 16S rDNA nucleotide sequence of L. garvieae (GenBank accession numbers: MT604790.1). Transmission electron microscopy (TEM) evaluation of one representative L. garvieae isolate (AOD109191) and the results of multiplex PCR did not reveal the presence of the capsular gene cluster (CGC), thus categorizing the isolate as the KG+ phenotype. Capsule staining and TEM observations confirmed the presence of a hyaluronic acid-like capsule, a possible virulence factor in KG+ phenotype L. garvieae isolates. The pathogenic potential of the representative isolate (AOD109191) was assessed through intraperitoneal injection challenges in cobia. The gross lesions and histopathological changes found in experimentally infected cobia were similar to those seen in naturally infected fish. This is the first report that confirms L. garvieae-induced 'warm water lactococcsis' can cause outbreaks of diseases in cage-cultured cobia.
Collapse
Affiliation(s)
- Shreesha Rao
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Trung Hieu Pham
- International Degree Program of Ornamental Fish Technology and Aquatic Animal Health, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Sayuj Poudyal
- International Degree Program of Ornamental Fish Technology and Aquatic Animal Health, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Li-Wu Cheng
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan.,Southern Taiwan Fish Diseases Research Centre, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Sandra Celenia Nazareth
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Pei-Chi Wang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan.,International Degree Program of Ornamental Fish Technology and Aquatic Animal Health, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan.,Southern Taiwan Fish Diseases Research Centre, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan.,Research Centre for Fish Vaccine and Diseases, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Shih-Chu Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan.,International Degree Program of Ornamental Fish Technology and Aquatic Animal Health, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan.,Southern Taiwan Fish Diseases Research Centre, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan.,Research Centre for Fish Vaccine and Diseases, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan.,Research Centre for Animal Biologics, National Pingtung University of Science and Technology, Pingtung, Taiwan
| |
Collapse
|
21
|
Dietrich K, Fiedler IA, Kurzyukova A, López-Delgado AC, McGowan LM, Geurtzen K, Hammond CL, Busse B, Knopf F. Skeletal Biology and Disease Modeling in Zebrafish. J Bone Miner Res 2021; 36:436-458. [PMID: 33484578 DOI: 10.1002/jbmr.4256] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022]
Abstract
Zebrafish are teleosts (bony fish) that share with mammals a common ancestor belonging to the phylum Osteichthyes, from which their endoskeletal systems have been inherited. Indeed, teleosts and mammals have numerous genetically conserved features in terms of skeletal elements, ossification mechanisms, and bone matrix components in common. Yet differences related to bone morphology and function need to be considered when investigating zebrafish in skeletal research. In this review, we focus on zebrafish skeletal architecture with emphasis on the morphology of the vertebral column and associated anatomical structures. We provide an overview of the different ossification types and osseous cells in zebrafish and describe bone matrix composition at the microscopic tissue level with a focus on assessing mineralization. Processes of bone formation also strongly depend on loading in zebrafish, as we elaborate here. Furthermore, we illustrate the high regenerative capacity of zebrafish bones and present some of the technological advantages of using zebrafish as a model. We highlight zebrafish axial and fin skeleton patterning mechanisms, metabolic bone disease such as after immunosuppressive glucocorticoid treatment, as well as osteogenesis imperfecta (OI) and osteopetrosis research in zebrafish. We conclude with a view of why larval zebrafish xenografts are a powerful tool to study bone metastasis. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Kristin Dietrich
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Imke Ak Fiedler
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasia Kurzyukova
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Alejandra C López-Delgado
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Karina Geurtzen
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Interdisciplinary Competence Center for Interface Research (ICCIR), Hamburg, Germany
| | - Franziska Knopf
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| |
Collapse
|
22
|
Tobias JH, Duncan EL, Kague E, Hammond CL, Gregson CL, Bassett D, Williams GR, Min JL, Gaunt TR, Karasik D, Ohlsson C, Rivadeneira F, Edwards JR, Hannan FM, Kemp JP, Gilbert SJ, Alonso N, Hassan N, Compston JE, Ralston SH. Opportunities and Challenges in Functional Genomics Research in Osteoporosis: Report From a Workshop Held by the Causes Working Group of the Osteoporosis and Bone Research Academy of the Royal Osteoporosis Society on October 5th 2020. Front Endocrinol (Lausanne) 2021; 11:630875. [PMID: 33658983 PMCID: PMC7917291 DOI: 10.3389/fendo.2020.630875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
The discovery that sclerostin is the defective protein underlying the rare heritable bone mass disorder, sclerosteosis, ultimately led to development of anti-sclerostin antibodies as a new treatment for osteoporosis. In the era of large scale GWAS, many additional genetic signals associated with bone mass and related traits have since been reported. However, how best to interrogate these signals in order to identify the underlying gene responsible for these genetic associations, a prerequisite for identifying drug targets for further treatments, remains a challenge. The resources available for supporting functional genomics research continues to expand, exemplified by "multi-omics" database resources, with improved availability of datasets derived from bone tissues. These databases provide information about potential molecular mediators such as mRNA expression, protein expression, and DNA methylation levels, which can be interrogated to map genetic signals to specific genes based on identification of causal pathways between the genetic signal and the phenotype being studied. Functional evaluation of potential causative genes has been facilitated by characterization of the "osteocyte signature", by broad phenotyping of knockout mice with deletions of over 7,000 genes, in which more detailed skeletal phenotyping is currently being undertaken, and by development of zebrafish as a highly efficient additional in vivo model for functional studies of the skeleton. Looking to the future, this expanding repertoire of tools offers the hope of accurately defining the major genetic signals which contribute to osteoporosis. This may in turn lead to the identification of additional therapeutic targets, and ultimately new treatments for osteoporosis.
Collapse
Affiliation(s)
- Jonathan H. Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Emma L. Duncan
- Department of Twin Research & Genetic Epidemiology, School of Life Course Sciences, King’s College London, London, United Kingdom
| | - Erika Kague
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Chrissy L. Hammond
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Celia L. Gregson
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion & Reproduction, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion & Reproduction, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Josine L. Min
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Tom R. Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - James R. Edwards
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Fadil M. Hannan
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - John P. Kemp
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- University of Queensland Diamantina Institute, University of Queensland, Woolloongabba, Queensland, QLD, Australia
| | - Sophie J. Gilbert
- Biomechanics and Bioengineering Centre Versus Arthritis, Cardiff School of Biosciences, Cardiff, United Kingdom
| | - Nerea Alonso
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Neelam Hassan
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Juliet E. Compston
- Department of Medicine, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Stuart H. Ralston
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
23
|
McGowan LM, Kague E, Vorster A, Newham E, Cross S, Hammond CL. Wnt16 Elicits a Protective Effect Against Fractures and Supports Bone Repair in Zebrafish. JBMR Plus 2021; 5:e10461. [PMID: 33778326 PMCID: PMC7990157 DOI: 10.1002/jbm4.10461] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/09/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022] Open
Abstract
Bone homeostasis is a dynamic, multicellular process that is required throughout life to maintain bone integrity, prevent fracture, and respond to skeletal damage. WNT16 has been linked to bone fragility and osteoporosis in human genome wide‐association studies, as well as the functional hematopoiesis of leukocytes in vivo. However, the mechanisms by which WNT16 promotes bone health and repair are not fully understood. In this study, CRISPR‐Cas9 was used to generate mutant zebrafish lacking Wnt16 (wnt16−/−) to study its effect on bone dynamically. The wnt16 mutants displayed variable tissue mineral density (TMD) and were susceptible to spontaneous fractures and the accumulation of bone calluses at an early age. Fractures were induced in the lepidotrichia of the caudal fins of wnt16−/− and WT zebrafish; this model was used to probe the mechanisms by which Wnt16 regulates skeletal and immune cell dynamics in vivo. In WT fins, wnt16 expression increased significantly during the early stages for bone repair. Mineralization of bone during fracture repair was significantly delayed in wnt16 mutants compared with WT zebrafish. Surprisingly, there was no evidence that the recruitment of innate immune cells to fractures or soft callus formation was altered in wnt16 mutants. However, osteoblast recruitment was significantly delayed in wnt16 mutants postfracture, coinciding with precocious activation of the canonical Wnt signaling pathway. In situ hybridization suggests that canonical Wnt‐responsive cells within fractures are osteoblast progenitors, and that osteoblast differentiation during bone repair is coordinated by the dynamic expression of runx2a and wnt16. This study highlights zebrafish as an emerging model for functionally validating osteoporosis–associated genes and investigating fracture repair dynamically in vivo. Using this model, it was found that Wnt16 protects against fracture and supports bone repair, likely by modulating canonical Wnt activity via runx2a to facilitate osteoblast differentiation and bone matrix deposition. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Erika Kague
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Alistair Vorster
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Elis Newham
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| | - Stephen Cross
- Wolfson Bioimaging Facility University of Bristol Bristol UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience University of Bristol Bristol UK
| |
Collapse
|
24
|
Carnovali M, Banfi G, Mariotti M. Age-dependent modulation of bone metabolism in zebrafish scales as new model of male osteoporosis in lower vertebrates. GeroScience 2020; 43:927-940. [PMID: 32997256 PMCID: PMC8110640 DOI: 10.1007/s11357-020-00267-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
After middle age, in human bone, the resorption usually exceeds formation resulting in bone loss and increased risk of fractures in the aged population. Only few in vivo models in higher vertebrates are available for pathogenic and therapeutic studies about bone aging. Among these, male Danio rerio (zebrafish) can be successfully used as low vertebrate model to study degenerative alterations that affect the skeleton during aging, reducing the role of sex hormones. In this paper, we investigated the early bone aging mechanisms in male zebrafish (3, 6, 9 months old) scales evaluating the physiological changes and the effects of prednisolone, a pro-osteoporotic drug. The results evidentiated an age-dependent reduction of the mineralization rate in the fish scales, as highlighted by growing circle measurements. Indeed, the osteoblastic ALP activity at the matrix deposition site was found progressively downregulated. The higher TRAP activity was found in 63% of 9-month-old fish scales associated with resorption lacunae along the scale border. Gene expression analysis evidentiated that an increase of the tnfrsf1b (homolog of human rank) in aging scales may be responsible for resorption stimulation. Interestingly, prednisolone inhibited the physiological growth of the scale and induced in aged scales a more significant bone resorption compared with untreated fish (3.8% vs 1.02%). Bone markers analysis shown a significant reduction of ALP/TRAP ratio due to a prednisolone-dependent stimulation of tnfsf11 (homolog of human rankl) in scales of older fish. The results evidentiated for the first time the presence of a senile male osteoporosis in lower vertebrate. This new model could be helpful to identify the early mechanisms of bone aging and new therapeutic strategies to prevent age-related bone alterations in humans.
Collapse
Affiliation(s)
| | - Giuseppe Banfi
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Mariotti
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy. .,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
25
|
Moss JJ, Hammond CL, Lane JD. Zebrafish as a model to study autophagy and its role in skeletal development and disease. Histochem Cell Biol 2020; 154:549-564. [PMID: 32915267 PMCID: PMC7609422 DOI: 10.1007/s00418-020-01917-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2020] [Indexed: 12/13/2022]
Abstract
In the last twenty years, research using zebrafish as a model organism has increased immensely. With the many advantages that zebrafish offer such as high fecundity, optical transparency, ex vivo development, and genetic tractability, they are well suited to studying developmental processes and the effect of genetic mutations. More recently, zebrafish models have been used to study autophagy. This important protein degradation pathway is needed for cell and tissue homeostasis in a variety of contexts. Correspondingly, its dysregulation has been implicated in multiple diseases including skeletal disorders. In this review, we explore how zebrafish are being used to study autophagy in the context of skeletal development and disease, and the ways these areas are intersecting to help identify potential therapeutic targets for skeletal disorders.
Collapse
Affiliation(s)
- Joanna J Moss
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, UK.,School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, UK.
| | - Jon D Lane
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, UK.
| |
Collapse
|
26
|
Busse B, Galloway JL, Gray RS, Harris MP, Kwon RY. Zebrafish: An Emerging Model for Orthopedic Research. J Orthop Res 2020; 38:925-936. [PMID: 31773769 PMCID: PMC7162720 DOI: 10.1002/jor.24539] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/16/2019] [Indexed: 02/04/2023]
Abstract
Advances in next-generation sequencing have transformed our ability to identify genetic variants associated with clinical disorders of the musculoskeletal system. However, the means to functionally validate and analyze the physiological repercussions of genetic variation have lagged behind the rate of genetic discovery. The zebrafish provides an efficient model to leverage genetic analysis in an in vivo context. Its utility for orthopedic research is becoming evident in regard to both candidate gene validation as well as therapeutic discovery in tissues such as bone, tendon, muscle, and cartilage. With the development of new genetic and analytical tools to better assay aspects of skeletal tissue morphology, mineralization, composition, and biomechanics, researchers are emboldened to systematically approach how the skeleton develops and to identify the root causes, and potential treatments, of skeletal disease. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:925-936, 2020.
Collapse
Affiliation(s)
- Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529, Hamburg, Germany
- all authors contributed equally to this work and are listed in alphabetical order
| | - Jenna L. Galloway
- Center for Regenerative Medicine, Harvard Stem Cell Institute, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street Boston, MA 02114, United States of America
- all authors contributed equally to this work and are listed in alphabetical order
| | - Ryan S. Gray
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, Texas, United States of America
- all authors contributed equally to this work and are listed in alphabetical order
| | - Matthew P. Harris
- Department of Genetics, Harvard Medical School; Department of Orthopedic Research, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA, 02115, United States of America
- all authors contributed equally to this work and are listed in alphabetical order
| | - Ronald Y. Kwon
- Department of Orthopaedics and Sports Medicine; Department of Mechanical Engineering; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
- all authors contributed equally to this work and are listed in alphabetical order
| |
Collapse
|
27
|
Zebrafish Models of Human Skeletal Disorders: Embryo and Adult Swimming Together. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1253710. [PMID: 31828085 PMCID: PMC6886339 DOI: 10.1155/2019/1253710] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/11/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023]
Abstract
Danio rerio (zebrafish) is an elective model organism for the study of vertebrate development because of its high degree of homology with human genes and organs, including bone. Zebrafish embryos, because of the optical clarity, small size, and fast development, can be easily used in large-scale mutagenesis experiments to isolate mutants with developmental skeletal defects and in high-throughput screenings to find new chemical compounds for the ability to revert the pathological phenotype. On the other hand, the adult zebrafish represents another powerful resource for pathogenic and therapeutic studies about adult human bone diseases. In fish, some characteristics such as bone turnover, reparation, and remodeling of the adult bone tissue cannot be found at the embryonic stage. Several pathological models have been established in adult zebrafish such as bone injury models, osteoporosis, and genetic diseases such as osteogenesis imperfecta. Given the growing interest for metabolic diseases and their complications, adult zebrafish models of type 2 diabetes and obesity have been recently generated and analyzed for bone complications using scales as model system. Interestingly, an osteoporosis-like phenotype has been found to be associated with metabolic alterations suggesting that bone complications share the same mechanisms in humans and fish. Embryo and adult represent powerful resources in rapid development to study bone physiology and pathology from different points of view.
Collapse
|
28
|
First person – Monika J. Tomecka. Dis Model Mech 2019. [PMCID: PMC6765197 DOI: 10.1242/dmm.041848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
First Person is a series of interviews with the first authors of a selection of papers published in Disease Models & Mechanisms (DMM), helping early-career researchers promote themselves alongside their papers. Monika Tomecka is first author on ‘Clinical pathologies of bone fracture modelled in zebrafish’, published in DMM. Monika conducted the research described in this article while a PhD student in Tom Carney's lab at the Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore and Henry Roehl's lab, Department of Biomedical Science, The University of Sheffield, UK. She is now a CEO at uFraction8, an engineering company that develops biotech instruments for industrial cell culture harvesting.
Collapse
|