151
|
Wang W, Ma W, Li X, Huang Y, Cao X. Platelet-Rich Plasma Suppresses Inflammation Reaction of Rat Articular Chondrocytes via Wingless-Related Integration Site/ β-Catenin Axis. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Our study aims to elucidate the role of platelet-rich plasma (PRP) in rats chondrocytes inflammation and mechanism. PRP was obtained from 8 weeks old rats. Then, the knee joint of bilateral hind limbs was dissected and articular chondrocytes were obtained in super-clean table after
dislocation and identified at the second generation during culture and passage. Chondrocytes were divided into control group 1 (addition of saline), control group 2 (IWP-2, Wnt/β-catenin axis inhibitor) and experimental group (PRP) followed by analysis of mRNA levels of glycogen
synthase kinase-3 (GSK-3β), low-density lipoprotein receptor-associated protein 5 (LRP5), Wnt1 and β-catenin by RT-PCR, IL-1 and TNF-α after 1 week by ELISA. The second generation articular chondrocytes presented polygonal or triangular cell morphology,
positive for collagen II and toluidine blue staining. PRP addition significantly reduced GSK-3β and LRP5 mRNA level, and increased β-catenin and Wnt1 mRNA levels in chondrocytes. Meanwhile, it suppressed IL-1 and TNF-α secretion and Wnt protein production
inhibitor 2. PRP might suppresses inflammatory factors production of rat articular chondrocytes through inhibiting Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Wei Wang
- Department of Orthopedics, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Weiwei Ma
- Department of Orthopedics, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Xu Li
- Department of Orthopedics, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Yihui Huang
- Department of Orthopedics, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Xinyu Cao
- Department of Orthopedics, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| |
Collapse
|
152
|
Helderman RC, Whitney DG, Duta-Mare M, Akhmetshina A, Vujic N, Jayapalan S, Nyman JS, Misra BB, Rosen CJ, Czech MP, Kratky D, Rendina-Ruedy E. Loss of function of lysosomal acid lipase (LAL) profoundly impacts osteoblastogenesis and increases fracture risk in humans. Bone 2021; 148:115946. [PMID: 33838322 PMCID: PMC8108562 DOI: 10.1016/j.bone.2021.115946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/26/2021] [Accepted: 04/01/2021] [Indexed: 11/28/2022]
Abstract
Lysosomal acid lipase (LAL) is essential for cholesteryl ester (CE) and triacylglycerol (TAG) hydrolysis in the lysosome. Clinically, an autosomal recessive LIPA mutation causes LAL deficiency (LALD), previously described as Wolman Disease or Cholesteryl Ester Storage Disease (CESD). LAL-D is associated with ectopic lipid accumulation in the liver, small intestine, spleen, adrenal glands, and blood. Considering the importance of unesterified cholesterol and fatty acids in bone metabolism, we hypothesized that LAL is essential for bone formation, and ultimately, skeletal health. To investigate the role of LAL in skeletal homeostasis, we used LAL-deficient (-/-) mice, in vitro osteoblast cultures, and novel clinical data from LAL-D patients. Both male and female LAL-/- mice demonstarted lower trabecular and cortical bone parameters , which translated to reduced biomechanical properties. Further histological analyses revealed that LAL-/- mice had fewer osteoblasts, with no change in osteoclast or marrow adipocyte numbers. In studying the cell-autonomous role of LAL, we observed impaired differentiation of LAL-/- calvarial osteoblasts and in bone marrow stromal cells treated with the LAL inhibitor lalistat. Consistent with LAL's role in other tissues, lalistat resulted in profound lipid puncta accumulation and an altered intracellular lipid profile. Finally, we analyzed a large de-identified national insurance database (i.e. 2016/2017 Optum Clinformatics®) which revealed that adults (≥18 years) with CESD (n = 3076) had a higher odds ratio (OR = 1.21; 95% CI = 1.03-1.41) of all-cause fracture at any location compared to adults without CESD (n = 13.7 M) after adjusting for demographic variables and osteoporosis. These data demonstrate that alterations in LAL have significant clinical implications related to fracture risk and that LAL's modulation of lipid metabolism is a critical for osteoblast function.
Collapse
Affiliation(s)
- Ron C Helderman
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA; Center for Bone Biology, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Daniel G Whitney
- Department of Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, MI 48108, USA; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, 48107, USA
| | - Madalina Duta-Mare
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - Alena Akhmetshina
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - Nemanja Vujic
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - Shobana Jayapalan
- Center for Bone Biology, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffry S Nyman
- Center for Bone Biology, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Biswapriya B Misra
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27104, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria
| | - Elizabeth Rendina-Ruedy
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA; Center for Bone Biology, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
153
|
Abstract
Osteogenesis imperfecta (OI) is a disease characterised by altered bone tissue material properties together with abnormal micro and macro-architecture and thus bone fragility, increased bone turnover and hyperosteocytosis. Increasingly appreciated are the soft tissue changes, sarcopenia in particular. Approaches to treatment are now multidisciplinary, with bisphosphonates having been the primary pharmacological intervention over the last 20 years. Whilst meta-analyses suggest that anti-fracture efficacy across the life course is equivocal, there is good evidence that for children bisphosphonates reduce fracture risk, increase vertebral size and improve vertebral shape, as well as improving motor function and mobility. The genetics of OI continues to provide insights into the molecular pathogenesis of the disease, although the pathophysiology is less clear. The complexity of the multi-scale interactions of bone tissue with cellular function are gradually being disentangled, but the fundamental question of why increased tissue brittleness should be associated with so many other changes is unclear; ER stress, pro-inflammatory cytokines, accelerated senesence and altered matrix component release might all contribute, but a unifying hypothesis remains elusive. New approaches to therapy are focussed on increasing bone mass, following the paradigm established by the treatment of postmenopausal osteoporosis. For adults, this brings the prospect of restoring previously lost bone - for children, particularly at the severe end of the spectrum, the possibility of further reducing fracture frequency and possibly altering growth and long term function are attractive. The alternatives that might affect tissue brittleness are autophagy enhancement (through the removal of abnormal type I collagen aggregates) and stem cell transplantation - both still at the preclinical stage of assessment. Preclinical assessment is not supportive of targeting inflammatory pathways, although understanding why TGFb signalling is increased, and whether that presents a treatment target in OI, remains to be established.
Collapse
Affiliation(s)
- Fawaz Arshad
- Academic Unit of Child Health, Sheffield Children's Hospital, Department of Oncology and Metabolism, University of Sheffield, S10 2TH, UK
| | - Nick Bishop
- Academic Unit of Child Health, Sheffield Children's Hospital, Department of Oncology and Metabolism, University of Sheffield, S10 2TH, UK.
| |
Collapse
|
154
|
Ponzetti M, Rucci N. Osteoblast Differentiation and Signaling: Established Concepts and Emerging Topics. Int J Mol Sci 2021; 22:ijms22136651. [PMID: 34206294 PMCID: PMC8268587 DOI: 10.3390/ijms22136651] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoblasts, the cells that build up our skeleton, are remarkably versatile and important cells that need tight regulation in all the phases of their differentiation to guarantee proper skeletal development and homeostasis. Although we know many of the key pathways involved in osteoblast differentiation and signaling, it is becoming clearer and clearer that this is just the tip of the iceberg, and we are constantly discovering novel concepts in osteoblast physiology. In this review, we discuss well-established pathways of osteoblastic differentiation, i.e., the classical ones committing mesenchymal stromal cells to osteoblast, and then osteocytes as well as recently emerged players. In particular, we discuss micro (mi)RNAs, long non-coding (lnc)RNAs, circular (circ)RNAs, and extracellular vesicles, focusing on the mechanisms through which osteoblasts are regulated by these factors, and conversely, how they use extracellular vesicles to communicate with the surrounding microenvironment.
Collapse
|
155
|
Mukkamalla SKR, Malipeddi D. Myeloma Bone Disease: A Comprehensive Review. Int J Mol Sci 2021; 22:6208. [PMID: 34201396 PMCID: PMC8227693 DOI: 10.3390/ijms22126208] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/17/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a neoplastic clonal proliferation of plasma cells in the bone marrow microenvironment, characterized by overproduction of heavy- and light-chain monoclonal proteins (M-protein). These proteins are mainly found in the serum and/or urine. Reduction in normal gammaglobulins (immunoparesis) leads to an increased risk of infection. The primary site of origin is the bone marrow for nearly all patients affected by MM with disseminated marrow involvement in most cases. MM is known to involve bones and result in myeloma bone disease. Osteolytic lesions are seen in 80% of patients with MM which are complicated frequently by skeletal-related events (SRE) such as hypercalcemia, bone pain, pathological fractures, vertebral collapse, and spinal cord compression. These deteriorate the patient's quality of life and affect the overall survival of the patient. The underlying pathogenesis of myeloma bone disease involves uncoupling of the bone remodeling processes. Interaction of myeloma cells with the bone marrow microenvironment promotes the release of many biochemical markers including osteoclast activating factors and osteoblast inhibitory factors. Elevated levels of osteoclast activating factors such as RANK/RANKL/OPG, MIP-1-α., TNF-α, IL-3, IL-6, and IL-11 increase bone resorption by osteoclast stimulation, differentiation, and maturation, whereas osteoblast inhibitory factors such as the Wnt/DKK1 pathway, secreted frizzle related protein-2, and runt-related transcription factor 2 inhibit osteoblast differentiation and formation leading to decreased bone formation. These biochemical factors also help in development and utilization of appropriate anti-myeloma treatments in myeloma patients. This review article summarizes the pathophysiology and the recent developments of abnormal bone remodeling in MM, while reviewing various approved and potential treatments for myeloma bone disease.
Collapse
Affiliation(s)
| | - Dhatri Malipeddi
- Internal Medicine, Canton Medical Education Foundation/NEOMED, Canton, OH 44710, USA;
| |
Collapse
|
156
|
Systems genetics in diversity outbred mice inform BMD GWAS and identify determinants of bone strength. Nat Commun 2021; 12:3408. [PMID: 34099702 PMCID: PMC8184749 DOI: 10.1038/s41467-021-23649-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
Genome-wide association studies (GWASs) for osteoporotic traits have identified over 1000 associations; however, their impact has been limited by the difficulties of causal gene identification and a strict focus on bone mineral density (BMD). Here, we use Diversity Outbred (DO) mice to directly address these limitations by performing a systems genetics analysis of 55 complex skeletal phenotypes. We apply a network approach to cortical bone RNA-seq data to discover 66 genes likely to be causal for human BMD GWAS associations, including the genes SERTAD4 and GLT8D2. We also perform GWAS in the DO for a wide-range of bone traits and identify Qsox1 as a gene influencing cortical bone accrual and bone strength. In this work, we advance our understanding of the genetics of osteoporosis and highlight the ability of the mouse to inform human genetics. Osteoporosis GWAS faces two challenges, causal gene discovery and a lack of phenotypic diversity. Here, the authors use the Diversity Outbred mouse population to inform human GWAS using networks and map genetic loci for 55 bone traits, identifying new potential bone strength genes.
Collapse
|
157
|
Stürznickel J, Jähn-Rickert K, Zustin J, Hennig F, Delsmann MM, Schoner K, Rehder H, Kreczy A, Schinke T, Amling M, Kornak U, Oheim R. Compound Heterozygous Frameshift Mutations in MESD Cause a Lethal Syndrome Suggestive of Osteogenesis Imperfecta Type XX. J Bone Miner Res 2021; 36:1077-1087. [PMID: 33596325 DOI: 10.1002/jbmr.4277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/03/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022]
Abstract
Multiple genes are known to be associated with osteogenesis imperfecta (OI), a phenotypically and genetically heterogenous bone disorder, marked predominantly by low bone mineral density and increased risk of fractures. Recently, mutations affecting MESD, which encodes for a chaperone required for trafficking of the low-density lipoprotein receptors LRP5 and LRP6 in the endoplasmic reticulum, were described to cause autosomal-recessive OI XX in homozygous children. In the present study, whole-exome sequencing of three stillbirths in one family was performed to evaluate the presence of a hereditary disorder. To further characterize the skeletal phenotype, fetal autopsy, bone histology, and quantitative backscattered electron imaging (qBEI) were performed, and the results were compared with those from an age-matched control with regular skeletal phenotype. In each of the affected individuals, compound heterozygous mutations in MESD exon 2 and exon 3 were detected. Based on the skeletal phenotype, which was characterized by multiple intrauterine fractures and severe skeletal deformity, OI XX was diagnosed in these individuals. Histological evaluation of MESD specimens revealed an impaired osseous development with an altered osteocyte morphology and reduced canalicular connectivity. Moreover, analysis of bone mineral density distribution by qBEI indicated an impaired and more heterogeneous matrix mineralization in individuals with MESD mutations than in controls. In contrast to the previously reported phenotypes of individuals with OI XX, the more severe phenotype in the present study is likely explained by a mutation in exon 2, located within the chaperone domain of MESD, that leads to a complete loss of function, which indicates the relevance of MESD in early skeletal development. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR)..
Collapse
Affiliation(s)
- Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Jähn-Rickert
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jozef Zustin
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Floriane Hennig
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany.,Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maximilian M Delsmann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Schoner
- Institute of Pathology, Fetal Pathology, Philipps-University Marburg, Marburg, Germany
| | - Helga Rehder
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Alfons Kreczy
- Department of Pathology, REGIOMED Klinikum Coburg, Coburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uwe Kornak
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany.,Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
158
|
Kondo H, Matsushita I, Nagata T, Fujihara E, Hosono K, Uchio E, Hotta Y, Kusaka S. Retinal Features of Family Members With Familial Exudative Vitreoretinopathy Caused By Mutations in KIF11 Gene. Transl Vis Sci Technol 2021; 10:18. [PMID: 34128965 PMCID: PMC8212440 DOI: 10.1167/tvst.10.7.18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/10/2021] [Indexed: 12/02/2022] Open
Abstract
Purpose To determine the clinical characteristics of patients and family members with familial exudative vitreoretinopathy (FEVR) caused by mutations in the KIF11 gene. Methods Twenty-one patients from 10 FEVR families with mutations in the KIF11 gene were studied. The retinal and systemic features were examined. The genetic analyses performed included Sanger sequencing of the KIF11 gene, whole exome sequencing, as well as array comparative genomic hybridization (CGH) analysis and multiple ligation probe assay (MLPA). Results Sequence analysis revealed seven different KIF11 mutations. Array CGH with MLPA revealed two different exon deletions. All probands had advanced FEVR with retinal detachments (RDs) and microcephaly with or without developmental disabilities. Patients with bilateral RDs were more frequently associated with developmental disabilities (P = 0.023). Multimodal imaging of the family members revealed that six of nine patients without RDs (66%) had varying degrees of chorioretinopathy. The retinal folds in FEVR patients were associated with severe retinal avascularization. However, funduscopic changes in the peripheral retina were unremarkable in family members without RDs. A score representing the peripheral vascular anomalies determined from the fluorescein angiograms was lower than that of control eyes of patients with mutations of the Wnt signaling genes (P = 0.0029). Conclusions The probands with KIF11 mutations were associated with severe ocular and systemic pathologies, whereas affected family members showed highly variable clinical manifestations. Peripheral vascular anomalies can often be unremarkable in eyes without RDs. Translational Relevance These findings highlight more diverse mechanisms that underlie the pathological changes in patients with FEVR.
Collapse
Affiliation(s)
- Hiroyuki Kondo
- Department of Ophthalmology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Itsuka Matsushita
- Department of Ophthalmology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tatsuo Nagata
- Department of Ophthalmology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Etsuko Fujihara
- Division of Ophthalmology, Matsue Red Cross Hospital, Matsue, Japan
| | - Katsuhiro Hosono
- Department of Ophthalmology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Eiichi Uchio
- Department of Ophthalmology, Fukuoka University, Fukuoka, Japan
| | - Yoshihiro Hotta
- Department of Ophthalmology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Shunji Kusaka
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
159
|
Marmor MT, Matz J, McClellan RT, Medam R, Miclau T. Use of Osteobiologics for Fracture Management: The When, What, and How. Injury 2021; 52 Suppl 2:S35-S43. [PMID: 33549314 DOI: 10.1016/j.injury.2021.01.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/02/2023]
Abstract
Osteobiologics are defined as a group of natural and synthetic materials used to augment bone healing. The selection of the most appropriate osteobiologic from the growing list of available options can be a challenging task. In selecting a material, surgeons should weigh a variety of considerations, including the indication for their use (the when), the most suitable substance (the what), and the correct mode of application (the how). This summary reviews these considerations and seeks to provide the surgeon with a basis for informed clinical evidence-based decision-making in their choice of a successful option.
Collapse
Affiliation(s)
- Meir T Marmor
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Jacob Matz
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Robert Trigg McClellan
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| | - Ramapaada Medam
- University of Central Florida College of Medicine, Orlando, FL, USA
| | - Theodore Miclau
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA.
| |
Collapse
|
160
|
Bhardwaj M, Yadav P, Vashishth D, Sharma K, Kumar A, Chahal J, Dalal S, Kataria SK. A Review on Obesity Management through Natural Compounds and a Green Nanomedicine-Based Approach. Molecules 2021; 26:3278. [PMID: 34071722 PMCID: PMC8198321 DOI: 10.3390/molecules26113278] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022] Open
Abstract
Obesity is a serious health complication in almost every corner of the world. Excessive weight gain results in the onset of several other health issues such as type II diabetes, cancer, respiratory diseases, musculoskeletal disorders (especially osteoarthritis), and cardiovascular diseases. As allopathic medications and derived pharmaceuticals are partially successful in overcoming this health complication, there is an incessant need to develop new alternative anti-obesity strategies with long term efficacy and less side effects. Plants harbor secondary metabolites such as phenolics, flavonoids, terpenoids and other specific compounds that have been shown to have effective anti-obesity properties. Nanoencapsulation of these secondary metabolites enhances the anti-obesity efficacy of these natural compounds due to their speculated property of target specificity and enhanced efficiency. These nanoencapsulated and naive secondary metabolites show anti-obesity properties mainly by inhibiting the lipid and carbohydrate metabolizing enzymes, suppression of adipogenesis and appetite, and enhancing energy metabolism. This review focuses on the plants and their secondary metabolites, along with their nanoencapsulation, that have anti-obesity effects, with their possible acting mechanisms, for better human health.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India; (M.B.); (P.Y.); (D.V.)
| | - Poonam Yadav
- Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India; (M.B.); (P.Y.); (D.V.)
| | - Divya Vashishth
- Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India; (M.B.); (P.Y.); (D.V.)
| | - Kavita Sharma
- Department of Zoology, Gaur Brahman Degree College, Rohtak 124001, India;
| | - Ajay Kumar
- Department of Zoology, Maharaja Neempal Singh Government College, Bhiwani 127021, India;
| | - Jyoti Chahal
- Department of Zoology, Hindu Girls College, Sonipat 131001, India;
| | - Sunita Dalal
- Department of Biotechnology, Kurukshetra University, Kurukshetra 136119, India;
| | - Sudhir Kumar Kataria
- Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India; (M.B.); (P.Y.); (D.V.)
| |
Collapse
|
161
|
Song D, He G, Shi Y, Ni J, Long F. Functional interaction between Wnt and Bmp signaling in periosteal bone growth. Sci Rep 2021; 11:10782. [PMID: 34031510 PMCID: PMC8144582 DOI: 10.1038/s41598-021-90324-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/04/2021] [Indexed: 02/05/2023] Open
Abstract
Wnt and Bmp proteins are well known to regulate bone development and homeostasis. Although both signals are extensively studied, their potential interaction in vivo is less well understood. Previous studies have shown that deletion of Bmpr1a, a type I receptor for Bmp signaling, results in excessive trabecular bone formation while diminishing periosteal bone growth. Moreover, forced-expression of the Wnt antagonist Sost suppresses the overgrowth of trabecular bone caused by Bmpr1a deletion, thus implicating hyperactive Wnt signaling in the excessive trabecular bone formation. However, it remains uncertain whether Wnt and Bmp signaling interacts in regulating the periosteal bone growth. Here we show that multiple Wnt genes are markedly suppressed in the cortical bone without Bmpr1a. Importantly, overexpression of Wnt7b fully rescues periosteal bone growth in the Bmpr1a-deficient mice. Thus, pharmacological activation of Wnt signaling can restore normal bone size without intact Bmp signaling.
Collapse
Affiliation(s)
- Deye Song
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Guangxu He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yu Shi
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.,State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiangdong Ni
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Fanxin Long
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, USA. .,Translational Research Program in Pediatric Orthopedics, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA. .,Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
162
|
Hasegawa T, Hirato M, Kobashi C, Yamaguchi A, Takagi R, Tanaka Y, Kaburaki T, Kakehashi A. Evaluation of the Foveal Avascular Zone in Familial Exudative Vitreoretinopathy Using Optical Coherence Tomography Angiography. Clin Ophthalmol 2021; 15:1913-1920. [PMID: 33994778 PMCID: PMC8114822 DOI: 10.2147/opth.s305520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/06/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose To evaluate the foveal avascular zone (FAZ) and retinal structure in familial exudative vitreoretinopathy (FEVR). Patients and Methods Eighteen eyes with stage 1 or 2 FEVR and 20 control eyes were evaluated. The central retinal thickness (CRT), foveal inner retinal thickness (IRT), surface retinal vessel density (SRVD), and deep retinal vessel density (DRVD) were measured using optical coherence tomography. The FAZ area was calculated using ImageJ software. The equivalent spherical value (SE) and axial length (AL) were measured. Results The CRT (232.5±3.086 vs 211±12.6325 μm; p=0.003) and foveal IRT (15.83±13.95 vs 0.9±4.02 μm; p=0.002) were thicker in the FEVR group than in the control group. The surface FAZ area (0.265±0.08 vs 0.364±0.09 mm2; p=0.004) and the deep FAZ area (0.364±0.1 vs 0.484±0.11 mm2; p=0.03) were smaller in the FEVR group than in the control group. The SRVD values did not differ among the sectors, but the DRVD was higher in the FEVR group except for the inferior sector (superior, p=0.027; inferior, p=0.88; temporal, p=0.035; nasal, p=0.027). The SE and AL did not differ between the two groups. There were no correlations between the surface and deep layer FAZ area and age, CRT, SE, and AL. The surface, deep FAZ area, and foveal IRT were correlated negatively (surface, r = -0.47, p=0.033; deep layer FAZ area, r = -0.46, p=0.037). Conclusion Eyes with FEVR have a smaller FAZ because the vascular structure in the inner retina remained in the fovea.
Collapse
Affiliation(s)
- Tetsuya Hasegawa
- Department of Ophthalmology, Saitama Medical Center Jichi Medical University, Saitama-shi, Saitama-ken, Japan
| | - Misaki Hirato
- Department of Ophthalmology, Saitama Medical Center Jichi Medical University, Saitama-shi, Saitama-ken, Japan
| | - Chieko Kobashi
- Department of Ophthalmology, Saitama Medical Center Jichi Medical University, Saitama-shi, Saitama-ken, Japan
| | - Aya Yamaguchi
- Department of Ophthalmology, Saitama Medical Center Jichi Medical University, Saitama-shi, Saitama-ken, Japan
| | - Rina Takagi
- Department of Ophthalmology, Saitama Medical Center Jichi Medical University, Saitama-shi, Saitama-ken, Japan
| | - Yoshiaki Tanaka
- Department of Ophthalmology, Saitama Medical Center Jichi Medical University, Saitama-shi, Saitama-ken, Japan
| | - Toshikatsu Kaburaki
- Department of Ophthalmology, Saitama Medical Center Jichi Medical University, Saitama-shi, Saitama-ken, Japan
| | - Akihiro Kakehashi
- Department of Ophthalmology, Saitama Medical Center Jichi Medical University, Saitama-shi, Saitama-ken, Japan
| |
Collapse
|
163
|
Zhang S, Li X, Liu W, Zhang X, Huang L, Li S, Yang M, Zhao P, Yang J, Fei P, Zhu X, Yang Z. Whole-Exome Sequencing Identified DLG1 as a Candidate Gene for Familial Exudative Vitreoretinopathy. Genet Test Mol Biomarkers 2021; 25:309-316. [PMID: 33945310 DOI: 10.1089/gtmb.2021.0013] [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: 11/13/2022] Open
Abstract
Purpose: Familial exudative vitreoretinopathy (FEVR) is a blinding retinal vascular disease. Clinically, FEVR is characterized by incomplete vascularization of the peripheral retina and pathological neovascularization. Only about 50% of FEVR cases can be explained by known FEVR disease gene variations. This study aimed to identify novel genes associated with the FEVR phenotype and explore their pathogenic mechanisms. Materials and Methods: Exome sequencing analyses were conducted on one Chinese family with FEVR whose affected members did not exhibit pathogenic variants in the known FEVR genes (verified using Sanger sequencing analysis). Functions of the affected proteins were evaluated using reporter assays. Western blot analysis was used to detect mutant protein expression and the genes' pathogenic mechanisms. Results: A rare novel heterozygous variant in DLG1 (c.1792A>G; p.S598G) was identified. The amino acid residues surrounding the identified variant are highly conserved among vertebrates. A luciferase reporter assay revealed that the mutant DLG1 protein DLG1-S598G lost its ability to activate Wnt signaling. Moreover, a knockdown (KD) of DLG1 in human primary retinal endothelial cells impaired tube formation. Mechanistically, DLG1 KD led to a reduction in phosphorylated VEGFR2, an essential receptor for the angiogenic potency that signals the vascular endothelial growth factor molecule. Conclusions: The data reported here demonstrate that DLG1 is a novel candidate gene for FEVR.
Collapse
Affiliation(s)
- Shanshan Zhang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Xiao Li
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Wenjing Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Xiang Zhang
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulin Huang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Shujin Li
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China.,Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, China
| | - Mu Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China.,Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiyun Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Ping Fei
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianjun Zhu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China.,Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, China
| | - Zhenglin Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Prenatal Diagnosis Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China.,Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, China
| |
Collapse
|
164
|
Caetano da Silva C, Ricquebourg M, Orcel P, Fabre S, Funck‐Brentano T, Cohen‐Solal M, Collet C. More severe phenotype of early-onset osteoporosis associated with recessive form of LRP5 and combination with DKK1 or WNT3A. Mol Genet Genomic Med 2021; 9:e1681. [PMID: 33939331 PMCID: PMC8222848 DOI: 10.1002/mgg3.1681] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Background Early‐onset osteoporosis (EOOP) is defined by low bone mineral density (BMD), which increases the risk of fracture. Although the prevalence of osteoporosis at a young age is unknown, low BMD is highly linked to genetic background. Heterozygous pathogenic variants in low‐density lipoprotein receptor‐related protein 5 (LRP5) are associated with EOOP. This study aimed to investigate the genetic profile in patients with EOOP to better understand the variation in phenotype severity by using a targeted gene sequencing panel associated with bone fragility. Method and Results We used a sequencing panel with 17 genes reported to be related to bone fragility for analysis of 68 patients with EOOP. We found a high positivity rate of EOOP with LRP5 variants (14 patients, 20.6%). The remaining 79.4% of patients with EOOP but without LRP5 variants showed variable disease severity, as observed in patients with at least one variant in this gene. One patient, with multiple fractures and spine L1‐L4 BMD Z‐score −2.9, carried a novel pathogenic homozygous variant, c.2918T>C, p.(Leu973Pro), without any pseudoglioma. In addition to carrying the LRP5 variant, 2 other patients carried a heterozygous variant in Wnt signaling pathway genes: dickkopf WNT signaling pathway inhibitor 1 (DKK1) [NM_012242.4: c.359G>T, p.(Arg120Leu)] and Wnt family member 3A (WNT3A) [NM_033131.3: c.377G>A, p. (Arg126His)]. As compared with single‐variant LRP5 carriers, double‐variant carriers had a significantly lower BMD Z‐score (−4.1 ± 0.8) and higher mean number of fractures (6.0 ± 2.8 vs. 2.2 ± 1.9). Analysis of the family segregation suggests the inheritance of BMD trait. Conclusion Severe forms of EOOP may occur with carriage of 2 pathogenic variants in genes encoding regulators of the Wnt signaling pathway. Two‐variant carriers of Wnt pathway genes had severe EOOP. Moreover, DKK1 and WNT3A genes should be included in next‐generation sequence analyses of bone fragility. Gene association may occur in the same signaling pathway and can generate a severe bone phenotype in early‐onset osteoporosis. Recessive form associated with lipoprotein receptor‐related protein 5 could be responsible for a stronger phenotype. Interestingly this recessive form is not associated with ocular problems as observed in pseudoglioma osteoporosis or vitreoretinopathy. Assessment of genetics based on an next generation sequencing panel should include WNT3A and DKK1.
Collapse
Affiliation(s)
| | - Manon Ricquebourg
- Inserm U1132 and Université de ParisParisFrance
- Department of RheumatologyHôpital Lariboisière, AP‐HPParisFrance
| | - Philippe Orcel
- Inserm U1132 and Université de ParisParisFrance
- Department of RheumatologyHôpital Lariboisière, AP‐HPParisFrance
| | - Stéphanie Fabre
- Inserm U1132 and Université de ParisParisFrance
- Department of RheumatologyHôpital Lariboisière, AP‐HPParisFrance
| | - Thomas Funck‐Brentano
- Inserm U1132 and Université de ParisParisFrance
- Department of RheumatologyHôpital Lariboisière, AP‐HPParisFrance
| | - Martine Cohen‐Solal
- Inserm U1132 and Université de ParisParisFrance
- Department of RheumatologyHôpital Lariboisière, AP‐HPParisFrance
| | - Corinne Collet
- Inserm U1132 and Université de ParisParisFrance
- Functional Unit of Molecular BiologyHôpital Lariboisière, AP‐HPParisFrance
| |
Collapse
|
165
|
Abstract
Skeletal integrity is maintained by a meticulous balance between bone resorption and bone formation, and recent studies have revealed the essential role of canonical Wnt signaling pathways in maintaining skeletal homeostasis. The SOST gene, which encodes sclerostin, a member of Dan family glycoproteins, was originally identified as the gene responsible for two sclerosing bone dysplasias, sclerosteosis and van Buchem disease. Sclerostin is highly expressed by osteocytes, negatively regulates canonical Wnt signaling pathways by binding to low-density lipoprotein receptor-related protein (LRP) 5/6, and suppresses osteoblast differentiation and/or function. Romosozumab, a specific anti-sclerostin antibody, inhibits sclerostin-LRP5/6 interactions and indirectly activates canonical Wnt signaling pathways and bone formation. This review focuses on the mechanism of action of sclerostin and summarizes clinical studies that demonstrated the efficacy of romosozumab to increase bone mineral density and reduce osteoporotic fractures, as well as its cardiovascular safety.
Collapse
Affiliation(s)
- Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
166
|
Twelve years of GWAS discoveries for osteoporosis and related traits: advances, challenges and applications. Bone Res 2021; 9:23. [PMID: 33927194 PMCID: PMC8085014 DOI: 10.1038/s41413-021-00143-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/21/2020] [Indexed: 02/03/2023] Open
Abstract
Osteoporosis is a common skeletal disease, affecting ~200 million people around the world. As a complex disease, osteoporosis is influenced by many factors, including diet (e.g. calcium and protein intake), physical activity, endocrine status, coexisting diseases and genetic factors. In this review, we first summarize the discovery from genome-wide association studies (GWASs) in the bone field in the last 12 years. To date, GWASs and meta-analyses have discovered hundreds of loci that are associated with bone mineral density (BMD), osteoporosis, and osteoporotic fractures. However, the GWAS approach has sometimes been criticized because of the small effect size of the discovered variants and the mystery of missing heritability, these two questions could be partially explained by the newly raised conceptual models, such as omnigenic model and natural selection. Finally, we introduce the clinical use of GWAS findings in the bone field, such as the identification of causal clinical risk factors, the development of drug targets and disease prediction. Despite the fruitful GWAS discoveries in the bone field, most of these GWAS participants were of European descent, and more genetic studies should be carried out in other ethnic populations to benefit disease prediction in the corresponding population.
Collapse
|
167
|
Donsante S, Palmisano B, Serafini M, Robey PG, Corsi A, Riminucci M. From Stem Cells to Bone-Forming Cells. Int J Mol Sci 2021; 22:ijms22083989. [PMID: 33924333 PMCID: PMC8070464 DOI: 10.3390/ijms22083989] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 12/22/2022] Open
Abstract
Bone formation starts near the end of the embryonic stage of development and continues throughout life during bone modeling and growth, remodeling, and when needed, regeneration. Bone-forming cells, traditionally termed osteoblasts, produce, assemble, and control the mineralization of the type I collagen-enriched bone matrix while participating in the regulation of other cell processes, such as osteoclastogenesis, and metabolic activities, such as phosphate homeostasis. Osteoblasts are generated by different cohorts of skeletal stem cells that arise from different embryonic specifications, which operate in the pre-natal and/or adult skeleton under the control of multiple regulators. In this review, we briefly define the cellular identity and function of osteoblasts and discuss the main populations of osteoprogenitor cells identified to date. We also provide examples of long-known and recently recognized regulatory pathways and mechanisms involved in the specification of the osteogenic lineage, as assessed by studies on mice models and human genetic skeletal diseases.
Collapse
Affiliation(s)
- Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Ospedale San Gerardo, 20900 Monza, Italy;
| | - Biagio Palmisano
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Ospedale San Gerardo, 20900 Monza, Italy;
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA;
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
- Correspondence:
| |
Collapse
|
168
|
Brommage R, Jeter-Jones S, Xiong W, Liu J. MicroCT analyses of mouse femoral neck architecture. Bone 2021; 145:115040. [PMID: 31437568 DOI: 10.1016/j.bone.2019.115040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 07/24/2019] [Accepted: 08/17/2019] [Indexed: 10/26/2022]
Abstract
Hip fractures at the femoral neck are a major cause of morbidity and mortality, but aside from biomechanical strength testing, little is known about femoral neck architecture in mice. Procedures were optimized to analyze high-resolution (6 μm voxel size) microCT scans of the mouse femoral neck to provide bone mass and architectural information. Similar to histomorphometric observations in rats, the boundary between cortical and trabecular bone is difficult to identify in the mouse femoral mid-neck and these compartments were not analyzed separately. Analyses included total area, mineralized bone area, and bone volume fraction (BV/TV). Femoral neck architecture varies in C57BL/6J, 129/SvEv and BALB/c mouse strains. Bone cross sectional area and BV/TV were low in Lrp5 but elevated in Sost gene knockout mice. Sfrp4 gene knockout resulted in high total area, normal bone area, low BV/TV and, as indicated by BS/BV values, greater trabecularization. Femoral neck BV/TV declined with age and ovariectomy, but increased with teriparatide treatment. These findings demonstrate that the architecture of the mouse femoral neck mimics phenotypes and treatment effects observed at other skeletal sites and is a relevant bone site for translational studies examining osteoporosis therapies.
Collapse
Affiliation(s)
- Robert Brommage
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA.
| | | | - Wendy Xiong
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA
| | - Jeff Liu
- Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, USA
| |
Collapse
|
169
|
Longo JF, Brosius SN, Znoyko I, Alers VA, Jenkins DP, Wilson RC, Carroll AJ, Wolff DJ, Roth KA, Carroll SL. Establishment and genomic characterization of a sporadic malignant peripheral nerve sheath tumor cell line. Sci Rep 2021; 11:5690. [PMID: 33707600 PMCID: PMC7952412 DOI: 10.1038/s41598-021-85055-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 02/17/2021] [Indexed: 12/19/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive Schwann cell-derived neoplasms that occur sporadically or in patients with neurofibromatosis type 1 (NF1). Preclinical research on sporadic MPNSTs has been limited as few cell lines exist. We generated and characterized a new sporadic MPNST cell line, 2XSB, which shares the molecular and genomic features of the parent tumor. These cells have a highly complex karyotype with extensive chromothripsis. 2XSB cells show robust invasive 3-dimensional and clonogenic culture capability and form solid tumors when xenografted into immunodeficient mice. High-density single nucleotide polymorphism array and whole exome sequencing analyses indicate that, unlike NF1-associated MPNSTs, 2XSB cells have intact, functional NF1 alleles with no evidence of mutations in genes encoding components of Polycomb Repressor Complex 2. However, mutations in other genes implicated in MPNST pathogenesis were identified in 2XSB cells including homozygous deletion of CDKN2A and mutations in TP53 and PTEN. We also identified mutations in genes not previously associated with MPNSTs but associated with the pathogenesis of other human cancers. These include DNMT1, NUMA1, NTRK1, PDE11A, CSMD3, LRP5 and ACTL9. This sporadic MPNST-derived cell line provides a useful tool for investigating the biology and potential treatment regimens for sporadic MPNSTs.
Collapse
Affiliation(s)
- Jody Fromm Longo
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425-9080, USA
| | - Stephanie N Brosius
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294-0017, USA
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, 35294-0017, USA
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Iya Znoyko
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425-9080, USA
| | - Victoria A Alers
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425-9080, USA
| | - Dorea P Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425-9080, USA
| | - Robert C Wilson
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425-9080, USA
- Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, 29425-9080, USA
| | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294-0017, USA
| | - Daynna J Wolff
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425-9080, USA
| | - Kevin A Roth
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, MSC 908, Charleston, SC, 29425-9080, USA.
- Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, 29425-9080, USA.
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294-0017, USA.
| |
Collapse
|
170
|
Goel D, Vohora D. Liver X receptors and skeleton: Current state-of-knowledge. Bone 2021; 144:115807. [PMID: 33333244 DOI: 10.1016/j.bone.2020.115807] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/26/2020] [Accepted: 12/11/2020] [Indexed: 12/25/2022]
Abstract
The liver X receptors (LXR) is a nuclear receptor that acts as a prominent regulator of lipid homeostasis and inflammatory response. Its therapeutic effectiveness against various diseases like Alzheimer's disease and atherosclerosis has been investigated in detail. Emerging pieces of evidence now reveal that LXR is also a crucial modulator of bone remodeling. However, the molecular mechanisms underlying the pharmacological actions of LXR on the skeleton and its role in osteoporosis are poorly understood. Therefore, in the current review, we highlight LXR and its actions through different molecular pathways modulating skeletal homeostasis. The studies described in this review propound that LXR in association with estrogen, PTH, PPARγ, RXR hedgehog, and canonical Wnt signaling regulates osteoclastogenesis and bone resorption. It regulates RANKL-induced expression of c-Fos, NFATc1, and NF-κB involved in osteoclast differentiation. Additionally, several studies suggest suppression of RANKL-induced osteoclast differentiation by synthetic LXR ligands. Given the significance of modulation of LXR in various physiological and pathological settings, our findings indicate that therapeutic targeting of LXR might potentially prevent or treat osteoporosis and improve bone quality.
Collapse
Affiliation(s)
- Divya Goel
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
171
|
Li FS, Li PP, Li L, Deng Y, Hu Y, He BC. PTEN Reduces BMP9-Induced Osteogenic Differentiation Through Inhibiting Wnt10b in Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 8:608544. [PMID: 33614622 PMCID: PMC7889951 DOI: 10.3389/fcell.2020.608544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/15/2020] [Indexed: 12/09/2022] Open
Abstract
Bone morphogenetic protein 9 (BMP9) is one of the most efficacious osteogenic cytokines. PTEN and Wnt10b are both implicated in regulating the osteogenic potential of BMP9, but the potential relationship between them is unknown. In this study, we determined whether PTEN could reduce the expression of Wnt10b during the osteogenic process initialized by BMP9 in mesenchymal stem cells (MSCs) and the possible molecular mechanism. We find that PTEN is inhibited by BMP9 in MSCs, but Wnt10b is increased simultaneously. The BMP9-induced osteogenic markers are reduced by PTEN but increased by silencing PTEN. The effects of knockdown PTEN on elevating BMP9-induced osteogenic markers are almost abolished by knockdown of Wnt10b. On the contrary, the BMP9-increased ALP activities and mineralization are both inhibited by PTEN but almost reversed by the combination of Wnt10b. Bone masses induced by BMP9 are enhanced by knockdown of PTEN, which is reduced by knockdown of Wnt10b. The BMP9-increased Wnt10b is decreased by PTEN but enhanced by knockdown of PTEN. Meanwhile, the BMP9-induced Wnt10b is also reduced by a PI3K-specific inhibitor (Ly294002) or rapamycin, respectively. The BMP9-induced phosphorylation of CREB or Smad1/5/9 is also reduced by PTEN, but enhanced by PTEN knockdown. In addition, p-CREB interacts with p-Smad1/5/9 in MSCs, and p-CREB or p-Smad1/5/9 are both enriched at the promoter region of Wnt10b. Our findings indicate that inhibitory effects of PTEN on BMP9's osteogenic potential may be partially mediated through decreasing the expression of Wnt10b via the disturbance of interaction between CREB and BMP/Smad signaling.
Collapse
Affiliation(s)
- Fu-Shu Li
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Pei-Pei Li
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Ling Li
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Yan Deng
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Ying Hu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| | - Bai-Cheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China.,Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| |
Collapse
|
172
|
Stürznickel J, Rolvien T, Delsmann A, Butscheidt S, Barvencik F, Mundlos S, Schinke T, Kornak U, Amling M, Oheim R. Clinical Phenotype and Relevance of LRP5 and LRP6 Variants in Patients With Early-Onset Osteoporosis (EOOP). J Bone Miner Res 2021; 36:271-282. [PMID: 33118644 DOI: 10.1002/jbmr.4197] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Reduced bone mineral density (BMD; ie, Z-score ≤-2.0) occurring at a young age (ie, premenopausal women and men <50 years) in the absence of secondary osteoporosis is considered early-onset osteoporosis (EOOP). Mutations affecting the WNT signaling pathway are of special interest because of their key role in bone mass regulation. Here, we analyzed the effects of relevant LRP5 and LRP6 variants on the clinical phenotype, bone turnover, BMD, and bone microarchitecture. After exclusion of secondary osteoporosis, EOOP patients (n = 372) were genotyped by gene panel sequencing, and segregation analysis of variants in LRP5/LRP6 was performed. The clinical assessment included the evaluation of bone turnover parameters, BMD by dual-energy X-ray absorptiometry, and microarchitecture via high-resolution peripheral quantitative computed tomography (HR-pQCT). In 50 individuals (31 EOOP index patients, 19 family members), relevant variants affecting LRP5 or LRP6 were detected (42 LRP5 and 8 LRP6 variants), including 10 novel variants. Seventeen variants were classified as disease causing, 14 were variants of unknown significance, and 19 were BMD-associated single-nucleotide polymorphisms (SNPs). One patient harbored compound heterozygous LRP5 mutations causing osteoporosis-pseudoglioma syndrome. Fractures were reported in 37 of 50 individuals, consisting of vertebral (18 of 50) and peripheral (29 of 50) fractures. Low bone formation was revealed in all individuals. A Z-score ≤-2.0 was detected in 31 of 50 individuals, and values at the spine were significantly lower than those at the hip (-2.1 ± 1.3 versus -1.6 ± 0.8; p = .003). HR-pQCT analysis (n = 34) showed impaired microarchitecture in trabecular and cortical compartments. Significant differences regarding the clinical phenotype were detectable between index patients and family members but not between different variant classes. Relevant variants in LRP5 and LRP6 contribute to EOOP in a substantial number of individuals, leading to a high number of fractures, low bone formation, reduced Z-scores, and impaired microarchitecture. This detailed skeletal characterization improves the interpretation of known and novel LRP5 and LRP6 variants. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alena Delsmann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastian Butscheidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Barvencik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Mundlos
- Institute of Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany.,Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
173
|
Shen L, Sharma D, Yu Y, Long F, Karner CM. Biphasic regulation of glutamine consumption by WNT during osteoblast differentiation. J Cell Sci 2021; 134:jcs251645. [PMID: 33262314 PMCID: PMC7823158 DOI: 10.1242/jcs.251645] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/19/2020] [Indexed: 01/01/2023] Open
Abstract
Osteoblasts are the principal bone-forming cells. As such, osteoblasts have enhanced demand for amino acids to sustain high rates of matrix synthesis associated with bone formation. The precise systems utilized by osteoblasts to meet these synthetic demands are not well understood. WNT signaling is known to rapidly stimulate glutamine uptake during osteoblast differentiation. Using a cell biology approach, we identified two amino acid transporters, γ(+)-LAT1 and ASCT2 (encoded by Slc7a7 and Slc1a5, respectively), as the primary transporters of glutamine in response to WNT. ASCT2 mediates the majority of glutamine uptake, whereas γ(+)-LAT1 mediates the rapid increase in glutamine uptake in response to WNT. Mechanistically, WNT signals through the canonical β-catenin (CTNNB1)-dependent pathway to rapidly induce Slc7a7 expression. Conversely, Slc1a5 expression is regulated by the transcription factor ATF4 downstream of the mTORC1 pathway. Targeting either Slc1a5 or Slc7a7 using shRNA reduced WNT-induced glutamine uptake and prevented osteoblast differentiation. Collectively, these data highlight the critical nature of glutamine transport for WNT-induced osteoblast differentiation.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Leyao Shen
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Deepika Sharma
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yilin Yu
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopaedics, The Children's Hospital of Philadelphia, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Courtney M Karner
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
174
|
El-Gazzar A, Högler W. Mechanisms of Bone Fragility: From Osteogenesis Imperfecta to Secondary Osteoporosis. Int J Mol Sci 2021; 22:ijms22020625. [PMID: 33435159 PMCID: PMC7826666 DOI: 10.3390/ijms22020625] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Bone material strength is determined by several factors, such as bone mass, matrix composition, mineralization, architecture and shape. From a clinical perspective, bone fragility is classified as primary (i.e., genetic and rare) or secondary (i.e., acquired and common) osteoporosis. Understanding the mechanism of rare genetic bone fragility disorders not only advances medical knowledge on rare diseases, it may open doors for drug development for more common disorders (i.e., postmenopausal osteoporosis). In this review, we highlight the main disease mechanisms underlying the development of human bone fragility associated with low bone mass known to date. The pathways we focus on are type I collagen processing, WNT-signaling, TGF-ß signaling, the RANKL-RANK system and the osteocyte mechanosensing pathway. We demonstrate how the discovery of most of these pathways has led to targeted, pathway-specific treatments.
Collapse
Affiliation(s)
| | - Wolfgang Högler
- Correspondence: ; Tel.: +43-(0)5-7680-84-22001; Fax: +43-(0)5-7680-84-22004
| |
Collapse
|
175
|
Abstract
The WNT/β-catenin signalling pathway is a rich and complex network of cellular proteins that orchestrates diverse short-range cell-to-cell communication in metazoans and is essential for both embryonic development and adult homeostasis. Due to its fundamental importance in controlling cell behaviour at multiple levels, its deregulation is associated with a wide range of diseases in humans and identification of drugs targeting the pathway has attracted strong interest in the pharmaceutical sector. Transduction of WNT signals across the plasma membrane of cells involves a staggering degree of complexity and variety with respect to ligand-receptor, receptor-receptor and receptor-co-receptor interactions (Niehrs, Nat Rev Mol Cell Biol 13:767-779, 2012). Although the low-density-lipoprotein-receptor-related-protein (LRP) family is best known for its role in binding and endocytosis of lipoproteins, specific members appear to have additional roles in cellular communication. Indeed, for WNT/β-catenin signalling one apparently universal requirement is the presence of either LRP5 or LRP6 in combination with one of the ten Frizzled (FZD) WNT receptors (FZD1-10). In the 20 years since their discovery as WNT/FZD co-receptors, research on the LRP family has contributed greatly to our understanding of WNT signalling and LRPs have emerged as central players in WNT/β-catenin signalling. LRP5/6 are highly similar and represent the least redundant class of WNT receptor that transduce WNT/β-catenin signalling from a wide range of different WNT and FZD subtypes. This apparent simplicity however belies the complex arrangement of binding sites in the extracellular domain (ECD) of LRP5/6, which regulate interaction not only with WNTs but also with several inhibitors of WNT signalling. This chapter provides a historical overview, chronologically charting this remarkable progress in the field during the last 20 years of research on LRPs and their role in WNT/-catenin signalling. A more focused overview of the structural, functional and mechanistic aspects of LRP biology is also provided, together with the implications this has for pharmacological targeting of this notoriously intractable pathway.
Collapse
Affiliation(s)
- Gary Davidson
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBSC-FMS), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
176
|
Aaron N, Kraakman MJ, Zhou Q, Liu Q, Costa S, Yang J, Liu L, Yu L, Wang L, He Y, Fan L, Hirakawa H, Ding L, Lo J, Wang W, Zhao B, Guo E, Sun L, Rosen CJ, Qiang L. Adipsin promotes bone marrow adiposity by priming mesenchymal stem cells. eLife 2021; 10:69209. [PMID: 34155972 PMCID: PMC8219379 DOI: 10.7554/elife.69209] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/07/2021] [Indexed: 01/12/2023] Open
Abstract
Background Marrow adipose tissue (MAT) has been shown to be vital for regulating metabolism and maintaining skeletal homeostasis in the bone marrow (BM) niche. As a reflection of BM remodeling, MAT is highly responsive to nutrient fluctuations, hormonal changes, and metabolic disturbances such as obesity and diabetes mellitus. Expansion of MAT has also been strongly associated with bone loss in mice and humans. However, the regulation of BM plasticity remains poorly understood, as does the mechanism that links changes in marrow adiposity with bone remodeling. Methods We studied deletion of Adipsin, and its downstream effector, C3, in C57BL/6 mice as well as the bone-protected PPARγ constitutive deacetylation 2KR mice to assess BM plasticity. The mice were challenged with thiazolidinedione treatment, calorie restriction, or aging to induce bone loss and MAT expansion. Analysis of bone mineral density and marrow adiposity was performed using a μCT scanner and by RNA analysis to assess adipocyte and osteoblast markers. For in vitro studies, primary bone marrow stromal cells were isolated and subjected to osteoblastogenic or adipogenic differentiation or chemical treatment followed by morphological and molecular analyses. Clinical data was obtained from samples of a previous clinical trial of fasting and high-calorie diet in healthy human volunteers. Results We show that Adipsin is the most upregulated adipokine during MAT expansion in mice and humans in a PPARγ acetylation-dependent manner. Genetic ablation of Adipsin in mice specifically inhibited MAT expansion but not peripheral adipose depots, and improved bone mass during calorie restriction, thiazolidinedione treatment, and aging. These effects were mediated through its downstream effector, complement component C3, to prime common progenitor cells toward adipogenesis rather than osteoblastogenesis through inhibiting Wnt/β-catenin signaling. Conclusions Adipsin promotes new adipocyte formation and affects skeletal remodeling in the BM niche. Our study reveals a novel mechanism whereby the BM sustains its own plasticity through paracrine and endocrine actions of a unique adipokine. Funding This work was supported by the National Institutes of Health T32DK007328 (NA), F31DK124926 (NA), R01DK121140 (JCL), R01AR068970 (BZ), R01AR071463 (BZ), R01DK112943 (LQ), R24DK092759 (CJR), and P01HL087123 (LQ).
Collapse
Affiliation(s)
- Nicole Aaron
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pharmacology, Columbia UniversityNew YorkUnited States
| | - Michael J Kraakman
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Medicine, Columbia UniversityNew YorkUnited States
| | - Qiuzhong Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical SchoolSingaporeSingapore
| | - Qiongming Liu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Samantha Costa
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States,School of Medicine, Tufts UniversityBostonUnited States,Graduate School of Biomedical Science and Engineering, University of MaineOronoUnited States
| | - Jing Yang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Longhua Liu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Lexiang Yu
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Liheng Wang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Medicine, Columbia UniversityNew YorkUnited States
| | - Ying He
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Lihong Fan
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| | - Hiroyuki Hirakawa
- Department of Microbiology and Immunology, Columbia UniversityNew YorkUnited States,Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Lei Ding
- Department of Microbiology and Immunology, Columbia UniversityNew YorkUnited States,Department of Rehabilitation and Regenerative Medicine, Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - James Lo
- Weill Center for Metabolic Health, Cardiovascular Research Institute, and Division of Cardiology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science CenterOklahoma CityUnited States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and The David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, Department of Medicine, Weill Cornell Medical College; Graduate Program in Cell & Developmental Biology, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Edward Guo
- Department of Biomedical Engineering, Columbia UniversityNew YorkUnited States
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical SchoolSingaporeSingapore
| | - Cliff J Rosen
- Center for Molecular Medicine, Maine Medical Center Research InstituteScarboroughUnited States
| | - Li Qiang
- Naomi Berrie Diabetes Cente, Columbia UniversityNew YorkUnited States,Department of Pathology and Cellular Biology, Columbia UniversityNew YorkUnited States
| |
Collapse
|
177
|
Negri S, Wang Y, Sono T, Qin Q, Hsu GCY, Cherief M, Xu J, Lee S, Tower RJ, Yu V, Piplani A, Meyers CA, Broderick K, Lee M, James AW. Systemic DKK1 neutralization enhances human adipose-derived stem cell mediated bone repair. Stem Cells Transl Med 2020; 10:610-622. [PMID: 33377628 PMCID: PMC7980212 DOI: 10.1002/sctm.20-0293] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/26/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022] Open
Abstract
Progenitor cells from adipose tissue are able to induce bone repair; however, inconsistent or unreliable efficacy has been reported across preclinical and clinical studies. Soluble inhibitory factors, such as the secreted Wnt signaling antagonists Dickkopf-1 (DKK1), are expressed to variable degrees in human adipose-derived stem cells (ASCs), and may represent a targetable "molecular brake" on ASC mediated bone repair. Here, anti-DKK1 neutralizing antibodies were observed to increase the osteogenic differentiation of human ASCs in vitro, accompanied by increased canonical Wnt signaling. Human ASCs were next engrafted into a femoral segmental bone defect in NOD-Scid mice, with animals subsequently treated with systemic anti-DKK1 or isotype control during the repair process. Human ASCs alone induced significant but modest bone repair. However, systemic anti-DKK1 induced an increase in human ASC engraftment and survival, an increase in vascular ingrowth, and ultimately improved bone repair outcomes. In summary, anti-DKK1 can be used as a method to augment cell-mediated bone regeneration, and could be particularly valuable in the contexts of impaired bone healing such as osteoporotic bone repair.
Collapse
Affiliation(s)
- Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA.,Orthopaedic and Trauma Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seungyong Lee
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Robert J Tower
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Victoria Yu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Abhi Piplani
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Carolyn A Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kristen Broderick
- Department of Plastic Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Min Lee
- School of Dentistry, University of California Los Angeles, Los Angeles, California, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
178
|
Singla A, Wang J, Yang R, Geller DS, Loeb DM, Hoang BH. Wnt Signaling in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:125-139. [PMID: 32767238 DOI: 10.1007/978-3-030-43085-6_8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Wnt molecules are a class of cysteine-rich secreted glycoproteins that participate in various developmental events during embryogenesis and adult tissue homeostasis. Since its discovery in 1982, the roles of Wnt signaling have been established in various key regulatory systems in biology. Wnt signals exert pleiotropic effects, including mitogenic stimulation, cell fate specification, and differentiation. The Wnt signaling pathway in humans has been shown to be involved in a wide variety of disorders including colon cancer, sarcoma, coronary artery disease, tetra-amelia, Mullerian duct regression, eye vascular defects, and abnormal bone mass. The canonical Wnt pathway functions by regulating the function of the transcriptional coactivator β-catenin, whereas noncanonical pathways function independent of β-catenin. Although the role of Wnt signaling is well established in epithelial malignancies, its role in mesenchymal tumors is more controversial. Some studies have suggested that Wnt signaling plays a pro-oncogenic role in various sarcomas by driving cell proliferation and motility; however, others have reported that Wnt signaling acts as a tumor suppressor by committing tumor cells to differentiate into a mature lineage. Wnt signaling pathway also plays an important role in regulating cancer stem cell function. In this review, we will discuss Wnt signaling pathway and its role in osteosarcoma.
Collapse
Affiliation(s)
- Amit Singla
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jichuan Wang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Musculoskeletal Tumor Center, Beijing Key Laboratory for Musculoskeletal Tumors, Peking University People's Hospital, Beijing, China
| | - Rui Yang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David S Geller
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David M Loeb
- Departments of Pediatrics and Developmental and Molecular Biology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bang H Hoang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
179
|
Takita S, Seko Y. eys +/- ; lrp5 +/- Zebrafish Reveals Lrp5 Can Be the Receptor of Retinol in the Visual Cycle. iScience 2020; 23:101762. [PMID: 33251495 PMCID: PMC7683268 DOI: 10.1016/j.isci.2020.101762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/17/2020] [Accepted: 10/30/2020] [Indexed: 12/04/2022] Open
Abstract
Vision is essential for vertebrates including humans. Sustained vision is accomplished by retinoid metabolism, the “visual cycle,” where all-trans retinol (atROL) is incorporated into the retinal pigment epithelium (RPE) from photoreceptors presumably through decade-long missing receptor(s). Here, we show that the LDL-related receptor-5 (Lrp5) protein is linked to the retinol binding protein 1a (Rbp1a), the transporter of atROL in the visual cycle, by generating and analyzing the digenic eyes shut homolog+/-; lrp5+/− zebrafish, the same form of gene defect detected in a human case of inherited retinal degeneration. Global gene expression analysis followed by genetic study clarified that rbp1a played a role downstream of lrp5. Rbp1a protein was colocalized with Lrp5 protein at microvilli of RPE cells. Furthermore, Rbp1a directly bound to the C-terminal intracellular region of Lrp5 in vitro. Collectively, these results strongly suggest that Lrp5 is a potent candidate of the receptor of atROL in the visual cycle. eys+/-; lrp5+/− zebrafish showed mild photoreceptor degeneration Microarray analysis identified dramatical decrease of rbp1a expression Rbp1a protein was colocalized with Lrp5 protein at the microvilli of the RPE Rbp1a directly bound to the C-terminal intracellular region of Lrp5 in vitro
Collapse
Affiliation(s)
- Shimpei Takita
- Visual Functions Section, Department of Rehabilitation for Sensory Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, 4-1 Namiki, Tokorozawa, Saitama 359-8555, Japan
| | - Yuko Seko
- Visual Functions Section, Department of Rehabilitation for Sensory Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, 4-1 Namiki, Tokorozawa, Saitama 359-8555, Japan
| |
Collapse
|
180
|
Kaur M, Nagpal M, Singh M. Osteoblast-n-Osteoclast: Making Headway to Osteoporosis Treatment. Curr Drug Targets 2020; 21:1640-1651. [DOI: 10.2174/1389450121666200731173522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/02/2020] [Accepted: 07/15/2020] [Indexed: 12/18/2022]
Abstract
Background:
Bone is a dynamic tissue that continuously undergoes the modeling and remodeling
process to maintain its strength and firmness. Bone remodeling is determined by the functioning
of osteoblast and osteoclast cells. The imbalance between the functioning of osteoclast and osteoblast
cells leads to osteoporosis. Osteoporosis is divided into primary and secondary osteoporosis.
Generally, osteoporosis is diagnosed by measuring bone mineral density (BMD) and various osteoblast
and osteoclast cell markers.
Methods:
Relevant literature reports have been studied and data has been collected using various
search engines like google scholar, scihub, sciencedirect, pubmed, etc. A thorough understanding of
the mechanism of bone targeting strategies has been discussed and related literature has been studied
and compiled.
Results:
Bone remodeling process has been described in detail including various approaches for targeting
bone. Several bone targeting moieties have been stated in detail along with their mechanisms.
Targeting of osteoclasts and osteoblasts using various nanocarriers has been discussed in separate sections.
The toxicity issues or Biosafety related to the use of nanomaterials have been covered.
Conclusion:
The treatment of osteoporosis targets the inhibition of bone resorption and the use of
agents that promote bone mineralization to slow disease progression. Current osteoporosis therapy involves
the use of targeting moieties such as bisphosphonates and tetracyclines for targeting various
drugs. Nanotechnology has been used for targeting various drug molecules such as RANKLinhibitors,
parathyroid hormone analogues, estrogen agonists and antagonists, Wnt signaling enhancer
and calcitonin specifically to bone tissue (osteoclast and osteoblasts). So, a multicomponent treatment
strategy targeting both the bone cells will be more effective rather than targeting only osteoclasts and
it will be a potential area of research in bone targeting used to treat osteoporosis.
The first section of the review article covers various aspects of bone targeting. Another section comprises
details of various targeting moieties such as bisphosphonates, tetracyclines; and various
nanocarriers developed to target osteoclast and osteoblast cells and summarized data on in vivo models
has been used for assessment of bone targeting, drawbacks of current strategies and future perspectives.
Collapse
Affiliation(s)
- Malkiet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
181
|
Kim KT, Lee YS, Han I. The Role of Epigenomics in Osteoporosis and Osteoporotic Vertebral Fracture. Int J Mol Sci 2020; 21:E9455. [PMID: 33322579 PMCID: PMC7763330 DOI: 10.3390/ijms21249455] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022] Open
Abstract
Osteoporosis is a complex multifactorial condition of the musculoskeletal system. Osteoporosis and osteoporotic vertebral fracture (OVF) are associated with high medical costs and can lead to poor quality of life. Genetic factors are important in determining bone mass and structure, as well as any predisposition for bone degradation and OVF. However, genetic factors are not enough to explain osteoporosis development and OVF occurrence. Epigenetics describes a mechanism for controlling gene expression and cellular processes without altering DNA sequences. The main mechanisms in epigenetics are DNA methylation, histone modifications, and non-coding RNAs (ncRNAs). Recently, alterations in epigenetic mechanisms and their activity have been associated with osteoporosis and OVF. Here, we review emerging evidence that epigenetics contributes to the machinery that can alter DNA structure, gene expression, and cellular differentiation during physiological and pathological bone remodeling. A progressive understanding of normal bone metabolism and the role of epigenetic mechanisms in multifactorial osteopathy can help us better understand the etiology of the disease and convert this information into clinical practice. A deep understanding of these mechanisms will help in properly coordinating future individual treatments of osteoporosis and OVF.
Collapse
Affiliation(s)
- Kyoung-Tae Kim
- Department of Neurosurgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (K.-T.K.); (Y.-S.L.)
- Department of Neurosurgery, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Young-Seok Lee
- Department of Neurosurgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (K.-T.K.); (Y.-S.L.)
- Department of Neurosurgery, Kyungpook National University Chilgok Hospital, Daegu 41944, Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do 13496, Korea
| |
Collapse
|
182
|
Long noncoding RNA expression profiles in intermittent parathyroid hormone induced cementogenesis. Genomics 2020; 113:217-228. [PMID: 33309767 DOI: 10.1016/j.ygeno.2020.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/01/2020] [Accepted: 12/06/2020] [Indexed: 11/22/2022]
Abstract
The aim of this study was to explore the involvement of long noncoding RNAs (lncRNAs) during intermittent parathyroid hormone (PTH) induced cementogenesis. Expression profiles of lncRNAs and mRNAs were obtained using high-throughput microarray. Gene Ontology enrichment analysis, Kyoto Encyclopedia of Genes and Genomes pathway analysis, and coding-noncoding gene coexpression networks construction were performed. We identified 190 lncRNAs and 135 mRNAs that were differentially expressed during intermittent PTH-induced cementogenesis. In this process, the Wnt signaling pathway was negatively regulated, and eight lncRNAs were identified as possible core regulators of Wnt signaling. Based on the results of microarrray analysis, we further verified the repressed expression of Wnt signaling crucial components β-catenin, APC and Axin2. Above all, we speculated that lncRNAs may play important roles in PTH-induced cementogenesis via the negative regulation of Wnt pathway.
Collapse
|
183
|
Alcorta-Sevillano N, Macías I, Infante A, Rodríguez CI. Deciphering the Relevance of Bone ECM Signaling. Cells 2020; 9:E2630. [PMID: 33297501 PMCID: PMC7762413 DOI: 10.3390/cells9122630] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Bone mineral density, a bone matrix parameter frequently used to predict fracture risk, is not the only one to affect bone fragility. Other factors, including the extracellular matrix (ECM) composition and microarchitecture, are of paramount relevance in this process. The bone ECM is a noncellular three-dimensional structure secreted by cells into the extracellular space, which comprises inorganic and organic compounds. The main inorganic components of the ECM are calcium-deficient apatite and trace elements, while the organic ECM consists of collagen type I and noncollagenous proteins. Bone ECM dynamically interacts with osteoblasts and osteoclasts to regulate the formation of new bone during regeneration. Thus, the composition and structure of inorganic and organic bone matrix may directly affect bone quality. Moreover, proteins that compose ECM, beyond their structural role have other crucial biological functions, thanks to their ability to bind multiple interacting partners like other ECM proteins, growth factors, signal receptors and adhesion molecules. Thus, ECM proteins provide a complex network of biochemical and physiological signals. Herein, we summarize different ECM factors that are essential to bone strength besides, discussing how these parameters are altered in pathological conditions related with bone fragility.
Collapse
Affiliation(s)
| | | | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, Barakaldo, 48903 Bizkaia, Spain; (N.A.-S.); (I.M.)
| | - Clara I. Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, Barakaldo, 48903 Bizkaia, Spain; (N.A.-S.); (I.M.)
| |
Collapse
|
184
|
|
185
|
Wnt signaling: An attractive target for periodontitis treatment. Biomed Pharmacother 2020; 133:110935. [PMID: 33227711 DOI: 10.1016/j.biopha.2020.110935] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Periodontitis is the most common chronic inflammatory disease, and a leading cause of tooth loss. Characterized by resorption of alveolar process and destruction of periodontal ligaments, periodontitis can impact not only periodontal tissues but also systemic diseases, such as diabetes, cardiovascular diseases, and respiratory infections. Currently, it is a hotspot to manage destruction and gain regeneration of periodontal tissues. Increasing evidence indicates that the Wnt signaling plays an important role in homeostasis of periodontal tissues, functions of periodontal derived cells, and progression of periodontitis. Its molecule expressions were abnormal in periodontitis. As such, modulators targeting the Wnt signaling may be an adjuvant therapy for periodontitis treatment. This review elucidates the role of Wnt signaling and its molecules, with a view to develop a potential application of drugs targeting the Wnt signaling for periodontitis treatment.
Collapse
|
186
|
Bosakova M, Abraham SP, Nita A, Hruba E, Buchtova M, Taylor SP, Duran I, Martin J, Svozilova K, Barta T, Varecha M, Balek L, Kohoutek J, Radaszkiewicz T, Pusapati GV, Bryja V, Rush ET, Thiffault I, Nickerson DA, Bamshad MJ, University of Washington Center for Mendelian Genomics, Rohatgi R, Cohn DH, Krakow D, Krejci P. Mutations in GRK2 cause Jeune syndrome by impairing Hedgehog and canonical Wnt signaling. EMBO Mol Med 2020; 12:e11739. [PMID: 33200460 PMCID: PMC7645380 DOI: 10.15252/emmm.201911739] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
Mutations in genes affecting primary cilia cause ciliopathies, a diverse group of disorders often affecting skeletal development. This includes Jeune syndrome or asphyxiating thoracic dystrophy (ATD), an autosomal recessive skeletal disorder. Unraveling the responsible molecular pathology helps illuminate mechanisms responsible for functional primary cilia. We identified two families with ATD caused by loss-of-function mutations in the gene encoding adrenergic receptor kinase 1 (ADRBK1 or GRK2). GRK2 cells from an affected individual homozygous for the p.R158* mutation resulted in loss of GRK2, and disrupted chondrocyte growth and differentiation in the cartilage growth plate. GRK2 null cells displayed normal cilia morphology, yet loss of GRK2 compromised cilia-based signaling of Hedgehog (Hh) pathway. Canonical Wnt signaling was also impaired, manifested as a failure to respond to Wnt ligand due to impaired phosphorylation of the Wnt co-receptor LRP6. We have identified GRK2 as an essential regulator of skeletogenesis and demonstrate how both Hh and Wnt signaling mechanistically contribute to skeletal ciliopathies.
Collapse
Affiliation(s)
- Michaela Bosakova
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Sara P Abraham
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Alexandru Nita
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Eva Hruba
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Marcela Buchtova
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - S Paige Taylor
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Ivan Duran
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Jorge Martin
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Katerina Svozilova
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Tomas Barta
- Department of Histology and EmbryologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Miroslav Varecha
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Lukas Balek
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | | | - Tomasz Radaszkiewicz
- Institute of Experimental BiologyFaculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Ganesh V Pusapati
- Department of BiochemistryStanford UniversityPalo AltoCAUSA
- Department of MedicineStanford UniversityPalo AltoCAUSA
| | - Vitezslav Bryja
- Institute of Experimental BiologyFaculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Eric T Rush
- Children's Mercy Kansas City, Center for Pediatric Genomic MedicineKansas CityMOUSA
- Department of PediatricsUniversity of MissouriKansas CityMOUSA
| | - Isabelle Thiffault
- Children's Mercy Kansas City, Center for Pediatric Genomic MedicineKansas CityMOUSA
- Department of PediatricsUniversity of MissouriKansas CityMOUSA
| | | | - Michael J Bamshad
- Department of Genome SciencesUniversity of WashingtonSeattleWAUSA
- Department of PediatricsUniversity of WashingtonSeattleWAUSA
- Division of Genetic MedicineSeattle Children's HospitalSeattleWAUSA
| | | | - Rajat Rohatgi
- Department of BiochemistryStanford UniversityPalo AltoCAUSA
- Department of MedicineStanford UniversityPalo AltoCAUSA
| | - Daniel H Cohn
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Molecular Cell and Developmental BiologyUniversity of California at Los AngelesLos AngelesCAUSA
| | - Deborah Krakow
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Human GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Obstetrics and GynecologyDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Pavel Krejci
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| |
Collapse
|
187
|
Effects of Estrogen Receptor and Wnt Signaling Activation on Mechanically Induced Bone Formation in a Mouse Model of Postmenopausal Bone Loss. Int J Mol Sci 2020; 21:ijms21218301. [PMID: 33167497 PMCID: PMC7663944 DOI: 10.3390/ijms21218301] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022] Open
Abstract
In the adult skeleton, bone remodeling is required to replace damaged bone and functionally adapt bone mass and structure according to the mechanical requirements. It is regulated by multiple endocrine and paracrine factors, including hormones and growth factors, which interact in a coordinated manner. Because the response of bone to mechanical signals is dependent on functional estrogen receptor (ER) and Wnt/β-catenin signaling and is impaired in postmenopausal osteoporosis by estrogen deficiency, it is of paramount importance to elucidate the underlying mechanisms as a basis for the development of new strategies in the treatment of osteoporosis. The present study aimed to investigate the effectiveness of the activation of the ligand-dependent ER and the Wnt/β-catenin signal transduction pathways on mechanically induced bone formation using ovariectomized mice as a model of postmenopausal bone loss. We demonstrated that both pathways interact in the regulation of bone mass adaption in response to mechanical loading and that the activation of Wnt/β-catenin signaling considerably increased mechanically induced bone formation, whereas the effects of estrogen treatment strictly depended on the estrogen status in the mice.
Collapse
|
188
|
Exosomal arrow (Arr)/lipoprotein receptor protein 6 (LRP6) in Drosophila melanogaster increases the extracellular level of Sol narae (Sona) in a Wnt-independent manner. Cell Death Dis 2020; 11:944. [PMID: 33139721 PMCID: PMC7608652 DOI: 10.1038/s41419-020-02850-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/30/2020] [Indexed: 12/28/2022]
Abstract
Wg/Wnt as a signaling protein binds to Frizzled (Fz) and Arrow (Arr), two Wg co-receptors essential for Wg signaling for cell proliferation, differentiation, and cell survival. Arr has a long extracellular region, a single transmembrane domain and an intracellular region. Here, we report that a new arrm7 mutant is identified in a genetic screen as a suppressor of lethality induced by overexpression of Sol narae (Sona), a secreted metalloprotease in ADAMTS family involved in Wg signaling. arrm7 allele has a premature stop codon, which encodes Arrm7 protein missing the intracellular region. arrm7 clones show cell death phenotype and overexpression of Arrm7 protein also induces cell death. Levels of extracellular Sona were decreased in both arrm7 and arr2 null clones, demonstrating that Arr increases the level of extracellular Sona. Indeed, Arr but not Arrm7, increased levels of Sona in cytoplasm and exosome fraction by inhibiting the lysosomal degradation pathway. Interestingly, Arr itself was identified in the exosome fraction, demonstrating that Arr is secreted to extracellular space. When Sona-expressing S2 cells were treated with exosomal Arr, the extracellular level of active Sona was increased. These results show that exosomal Arr dictates Sona-expressing cells to increase the level of extracellular Sona. This new function of Arr occurred in the absence of Wg because S2 cells do not express Wg. We propose that Arr plays two distinct roles, one as an exosomal protein to increase the level of extracellular Sona in a Wnt-independent manner and the other as a Wg co-receptor in a Wnt-dependent manner.
Collapse
|
189
|
Fenofibrate prevents iron induced activation of canonical Wnt/β-catenin and oxidative stress signaling in the retina. NPJ Aging Mech Dis 2020; 6:12. [PMID: 33145027 PMCID: PMC7599211 DOI: 10.1038/s41514-020-00050-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/09/2020] [Indexed: 12/20/2022] Open
Abstract
Accumulating evidence strongly implicates iron in the pathogenesis of aging and disease. Iron levels have been found to increase with age in both the human and mouse retinas. We and others have shown that retinal diseases such as age-related macular degeneration and diabetic retinopathy are associated with disrupted iron homeostasis, resulting in retinal iron accumulation. In addition, hereditary disorders due to mutation in one of the iron regulatory genes lead to age dependent retinal iron overload and degeneration. However, our knowledge on whether iron toxicity contributes to the retinopathy is limited. Recently, we reported that iron accumulation is associated with the upregulation of retinal and renal renin-angiotensin system (RAS). Evidences indicate that multiple genes/components of the RAS are targets of Wnt/β-catenin signaling. Interestingly, aberrant activation of Wnt/β-catenin signaling is observed in several degenerative diseases. In the present study, we explored whether iron accumulation regulates canonical Wnt signaling in the retina. We found that in vitro and in vivo iron treatment resulted in the upregulation of Wnt/β-catenin signaling and its downstream target genes including renin-angiotensin system in the retina. We confirmed further that iron activates canonical Wnt signaling in the retina using TOPFlash T-cell factor/lymphoid enhancer factor promoter assay and Axin2-LacZ reporter mouse. The presence of an iron chelator or an antioxidant reversed the iron-mediated upregulation of Wnt/β-catenin signaling in retinal pigment epithelial (RPE) cells. In addition, treatment of RPE cells with peroxisome proliferator-activated receptor (PPAR) α-agonist fenofibrate prevented iron-induced activation of oxidative stress and Wnt/β-catenin signaling by chelating the iron. The role of fenofibrate, an FDA-approved drug for hyperlipidemia, as an iron chelator has potentially significant therapeutic impact on iron associated degenerative diseases.
Collapse
|
190
|
Abstract
The phenotypic trait of high bone mass (HBM) is an excellent example of the nexus between common and rare disease genetics. HBM may arise from carriage of many 'high bone mineral density [BMD]'-associated alleles, and certainly the genetic architecture of individuals with HBM is enriched with high BMD variants identified through genome-wide association studies of BMD. HBM may also arise as a monogenic skeletal disorder, due to abnormalities in bone formation, bone resorption, and/or bone turnover. Individuals with monogenic disorders of HBM usually, though not invariably, have other skeletal abnormalities (such as mandible enlargement) and thus are best regarded as having a skeletal dysplasia rather than just isolated high BMD. A binary etiological division of HBM into polygenic vs. monogenic, however, would be excessively simplistic: the phenotype of individuals carrying rare variants of large effect can still be modified by their common variant polygenic background, and by the environment. HBM disorders-whether predominantly polygenic or monogenic in origin-are not only interesting clinically and genetically: they provide insights into bone processes that can be exploited therapeutically, with benefits both for individuals with these rare bone disorders and importantly for the many people affected by the commonest bone disease worldwide-i.e., osteoporosis. In this review we detail the genetic architecture of HBM; we provide a conceptual framework for considering HBM in the clinical context; and we discuss monogenic and polygenic causes of HBM with particular emphasis on anabolic causes of HBM.
Collapse
Affiliation(s)
- Celia L. Gregson
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Emma L. Duncan
- Department of Twin Research & Genetic Epidemiology, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
191
|
Sharma M, Pruitt K. Wnt Pathway: An Integral Hub for Developmental and Oncogenic Signaling Networks. Int J Mol Sci 2020; 21:E8018. [PMID: 33126517 PMCID: PMC7663720 DOI: 10.3390/ijms21218018] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
The Wnt pathway is an integral cell-to-cell signaling hub which regulates crucial development processes and maintenance of tissue homeostasis by coordinating cell proliferation, differentiation, cell polarity, cell movement, and stem cell renewal. When dysregulated, it is associated with various developmental diseases, fibrosis, and tumorigenesis. We now better appreciate the complexity and crosstalk of the Wnt pathway with other signaling cascades. Emerging roles of the Wnt signaling in the cancer stem cell niche and drug resistance have led to development of therapeutics specifically targeting various Wnt components, with some agents currently in clinical trials. This review highlights historical and recent findings on key mediators of Wnt signaling and how they impact antitumor immunity and maintenance of cancer stem cells. This review also examines current therapeutics being developed that modulate Wnt signaling in cancer and discusses potential shortcomings associated with available therapeutics.
Collapse
Affiliation(s)
| | - Kevin Pruitt
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| |
Collapse
|
192
|
Zheng S, Wei Y, Jiang Y, Hao Y. LRP8 activates STAT3 to induce PD-L1 expression in osteosarcoma. TUMORI JOURNAL 2020; 107:238-246. [PMID: 33054597 DOI: 10.1177/0300891620952872] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE Targeting programmed death-ligand 1 (PD-L1) may be an effective intervention for osteosarcoma and PD-L1 expression is controlled by diverse regulatory factors. Low-density lipoprotein receptor-related protein 8 (LRP8) regulates osteoblast differentiation and it is unclear whether and how LRP8 could contribute to osteosarcoma pathogenesis. In this study, we investigated the LRP8/signal transducer and activator of transcription 3 (STAT3)/PD-L1 network in osteosarcoma. METHODS The expression of LRP8, STAT3, and PD-L1 was measured in osteosarcoma tissues and paired normal tissues. The effects of LRP8 on STAT3 and PD-L1 expression were investigated in an osteosarcoma cell line. The effects on immunosuppression were investigated in an in vitro co-culture system with Jurkat cell line and osteosarcoma cell line. The effects of LRP8 were blocked by a LRP8 neutralizing antibody, dominant-negative STAT3, or STAT3 inhibitor. RESULTS LRP8 was overexpressed in osteosarcoma compared to normal tissues and its level was correlated with phospho-STAT3 (p-STAT3) level in osteosarcoma tissues. In osteosarcoma cell lines, LRP8 increased p-STAT3 level and promoted nuclear translocation of STAT3. STAT3 activation also increased PD-L1 mRNA, protein, and promoter activity. In addition, LRP8 enhanced PD-L1 expression via STAT3. In a co-culture system, LRP8 overexpression in an osteosarcoma cell line impaired viability and interleukin-2 secretion of Jurkat cells and induced apoptosis of Jurkat cells. The effects of LRP8 could be blocked by neutralizing LRP8 antibody or STAT3 inhibitor. Blocking LRP8 inhibits proliferation and induces apoptosis of osteosarcoma cells. CONCLUSIONS Our results provide evidence for a novel regulation network of LRP8/STAT3/PD-L1 in osteosarcoma and LRP8 may be a potential therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Shiqin Zheng
- Department of Osteoarthrosis, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yuxi Wei
- Department of Osteoarthrosis, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yu Jiang
- Department of Osteoarthrosis, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yi Hao
- Department of Osteoarthrosis, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
193
|
Trajanoska K, Rivadeneira F. Genomic Medicine: Lessons Learned From Monogenic and Complex Bone Disorders. Front Endocrinol (Lausanne) 2020; 11:556610. [PMID: 33162933 PMCID: PMC7581702 DOI: 10.3389/fendo.2020.556610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022] Open
Abstract
Current genetic studies of monogenic and complex bone diseases have broadened our understanding of disease pathophysiology, highlighting the need for medical interventions and treatments tailored to the characteristics of patients. As genomic research progresses, novel insights into the molecular mechanisms are starting to provide support to clinical decision-making; now offering ample opportunities for disease screening, diagnosis, prognosis and treatment. Drug targets holding mechanisms with genetic support are more likely to be successful. Therefore, implementing genetic information to the drug development process and a molecular redefinition of skeletal disease can help overcoming current shortcomings in pharmaceutical research, including failed attempts and appalling costs. This review summarizes the achievements of genetic studies in the bone field and their application to clinical care, illustrating the imminent advent of the genomic medicine era.
Collapse
|
194
|
Zhu X, Sun K, Huang L, Ma S, Hao F, Yang Z, Sundaresan P, Zhu X. Identification of Novel Mutations in the FZD4 and NDP Genes in Patients with Familial Exudative Vitreoretinopathy in South India. Genet Test Mol Biomarkers 2020; 24:92-98. [PMID: 31999491 DOI: 10.1089/gtmb.2019.0212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background: Familial exudative vitreoretinopathy (FEVR) is an inheritable retinal vascular disease, which often leads to severe vision loss and blindness in children. However, reported mutations can only account for 50-60% of patients with FEVR. The purpose of this study was to identify novel frizzled class receptor 4 (FZD4) and Norrin cystine knot growth factor NDP (NDP) mutations in a cohort of Indian patients with FEVR by whole-exome sequencing. Methods: We performed data filtering and bioinformatic analyses. Results: Two novel heterozygous mutations in FZD4 gene were identified, each in two different families: c.1499_1500del [p.500_500del] and c.G296C [p.C99S]. One novel mutation in NDP in another family was identified: c.A256G [p.K86E]. All FZD4 mutations affected conserved amino acid residues and were absent in 1000 control individuals. To assess the effect of these FZD4 mutations on the biological activity of the protein, we introduced each FZD4 mutation into FZD4 cDNA by the site-directed mutagenesis techniques. A Norrin/beta-catenin pathway-based luciferase reporter assay revealed that the c.1499_1500del failed to activate the luciferase reporter; in contrast, compared with the wild-type FZD4 protein, the, c.G296C [p.C99S] mutation exhibited increased luciferase reporter activity. Conclusion: Our study found two novel FZD4 mutations, with opposite effects regarding functional expression levels in Indian patients with FEVR and expands on the mutational spectrum of FZD4 in Indian FEVR patients.
Collapse
Affiliation(s)
- Xiong Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Kuanxiang Sun
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lulin Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shi Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Fang Hao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Periasamy Sundaresan
- Department of Genetics, Aravind Medical Research Foundation, Aravind Eye Hospital, Madurai, India
| | - Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
195
|
Bertrand AA, Malapati SH, Yamaguchi DT, Lee JC. The Intersection of Mechanotransduction and Regenerative Osteogenic Materials. Adv Healthc Mater 2020; 9:e2000709. [PMID: 32940024 PMCID: PMC7864218 DOI: 10.1002/adhm.202000709] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/14/2020] [Indexed: 12/23/2022]
Abstract
Mechanical signals play a central role in cell fate determination and differentiation in both physiologic and pathologic circumstances. Such signals may be delivered using materials to generate discrete microenvironments for the purposes of tissue regeneration and have garnered increasing attention in recent years. Unlike the addition of progenitor cells or growth factors, delivery of a microenvironment is particularly attractive in that it may reduce the known untoward consequences of the former two strategies, such as excessive proliferation and potential malignant transformation. Additionally, the ability to spatially modulate the fabrication of materials allows for the creation of multiple microenvironments, particularly attractive for regenerating complex tissues. While many regenerative materials have been developed and tested for augmentation of specific cellular responses, the intersection between cell biology and material interactions have been difficult to dissect due to the complexity of both physical and chemical interactions. Specifically, modulating materials to target individual signaling pathways is an avenue of interdisciplinary research that may lead to a more effective method of optimizing regenerative materials. In this work, the aim is to summarize the major mechanotransduction pathways for osteogenic differentiation and to consolidate the known materials and material properties that activate such pathways.
Collapse
Affiliation(s)
- Anthony A. Bertrand
- Division of Plastic and Reconstructive Surgery, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Sri Harshini Malapati
- Division of Plastic and Reconstructive Surgery, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Dean T. Yamaguchi
- Division of Plastic and Reconstructive Surgery, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, California
| | - Justine C. Lee
- Division of Plastic and Reconstructive Surgery, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, California
- UCLA Molecular Biology Institute, Los Angeles, California
| |
Collapse
|
196
|
Lee SJ, Lehar A, Meir JU, Koch C, Morgan A, Warren LE, Rydzik R, Youngstrom DW, Chandok H, George J, Gogain J, Michaud M, Stoklasek TA, Liu Y, Germain-Lee EL. Targeting myostatin/activin A protects against skeletal muscle and bone loss during spaceflight. Proc Natl Acad Sci U S A 2020; 117:23942-23951. [PMID: 32900939 PMCID: PMC7519220 DOI: 10.1073/pnas.2014716117] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Among the physiological consequences of extended spaceflight are loss of skeletal muscle and bone mass. One signaling pathway that plays an important role in maintaining muscle and bone homeostasis is that regulated by the secreted signaling proteins, myostatin (MSTN) and activin A. Here, we used both genetic and pharmacological approaches to investigate the effect of targeting MSTN/activin A signaling in mice that were sent to the International Space Station. Wild type mice lost significant muscle and bone mass during the 33 d spent in microgravity. Muscle weights of Mstn-/- mice, which are about twice those of wild type mice, were largely maintained during spaceflight. Systemic inhibition of MSTN/activin A signaling using a soluble form of the activin type IIB receptor (ACVR2B), which can bind each of these ligands, led to dramatic increases in both muscle and bone mass, with effects being comparable in ground and flight mice. Exposure to microgravity and treatment with the soluble receptor each led to alterations in numerous signaling pathways, which were reflected in changes in levels of key signaling components in the blood as well as their RNA expression levels in muscle and bone. These findings have implications for therapeutic strategies to combat the concomitant muscle and bone loss occurring in people afflicted with disuse atrophy on Earth as well as in astronauts in space, especially during prolonged missions.
Collapse
Affiliation(s)
- Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032;
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam Lehar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | - Jessica U Meir
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Christina Koch
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Andrew Morgan
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Lara E Warren
- Center for the Advancement of Science in Space, Houston, TX 77058
| | - Renata Rydzik
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, CT 06030
| | | | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | | | - Michael Michaud
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | | | - Yewei Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | - Emily L Germain-Lee
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030
- Connecticut Children's Center for Rare Bone Disorders, Farmington, CT 06032
| |
Collapse
|
197
|
Yorgan TA, Rolvien T, Stürznickel J, Vollersen N, Lange F, Zhao W, Baranowsky A, Rosenthal L, Hermans-Borgmeyer I, Sharaf A, Karsak M, David JP, Oheim R, Amling M, Schinke T. Mice Carrying a Ubiquitous R235W Mutation of Wnt1 Display a Bone-Specific Phenotype. J Bone Miner Res 2020; 35:1726-1737. [PMID: 32369212 DOI: 10.1002/jbmr.4043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Since a key function of Wnt1 in brain development was established early on through the generation of non-viable Wnt1-deficient mice, it was initially surprising that WNT1 mutations were found to cause either early-onset osteoporosis (EOOP) or osteogenesis imperfecta type XV (OI-XV). The deduced function of Wnt1 as an osteoanabolic factor has been confirmed in various mouse models with bone-specific inactivation or overexpression, but mice carrying disease-causing Wnt1 mutations have not yet been described. Triggered by the clinical analysis of EOOP patients carrying a heterozygous WNT1 mutation (p.R235W), we introduced this mutation into the murine Wnt1 gene to address the question of whether this would cause a skeletal phenotype. We observed that Wnt1+/R235W and Wnt1R235W/R235W mice were born at the expected Mendelian ratio and that they did not display postnatal lethality or obvious nonskeletal phenotypes. At 12 weeks of age, the homozygous presence of the Wnt1 mutation was associated with reduced trabecular and cortical bone mass, explained by a lower bone formation rate compared with wild-type littermates. At 52 weeks of age, we also observed a moderate bone mass reduction in heterozygous Wnt1+/R235W mice, thereby underscoring their value as a model of WNT1-dependent EOOP. Importantly, when we treated wild-type and Wnt1+/R235W mice by daily injection of parathyroid hormone (PTH), we detected the same osteoanabolic influence in both groups, together with an increased cortical thickness in the mutant mice. Our data demonstrate the pathogenicity of the WNT1-R235W mutation, confirm that controlling skeletal integrity is the primary physiological function of Wnt1, and suggest that osteoanabolic treatment with teriparatide should be applicable for individuals with WNT1-dependent EOOP. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Orthopedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nele Vollersen
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabiola Lange
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wenbo Zhao
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Baranowsky
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lana Rosenthal
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ahmed Sharaf
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meliha Karsak
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jean-Pierre David
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
198
|
Liang K, Du Y, Chen L, Wang L, Li R, Yan Z, Liu Y. Contrary Roles of Wnt/β-Catenin Signaling in BMP9-Induced Osteogenic and Adipogenic Differentiation of 3T3-L1 Preadipocytes. Cell Biochem Biophys 2020; 78:347-356. [PMID: 32720112 DOI: 10.1007/s12013-020-00935-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 07/17/2020] [Indexed: 12/31/2022]
Abstract
Our previous study revealed that 3T3-L1 preadipocytes can differentiate to either osteoblasts or adipocytes in response to bone morphogenic protein 9 (BMP9). In the present study, we try to further investigate whether the Wnt/β-catenin signaling plays a crucial role in this process. It was found that BMP9 effectively activated the Wnt/β-catenin signaling, and induced the expression levels of certain canonical Wnt ligands and their receptors in preadipocytes. Exogenous expression of β-catenin, Wnt1, Wnt3a, and Wnt10b potentiated BMP9-induced alkaline phosphatase (ALP) activity, while β-catenin knockdown or Dickkopf 1 (Dkk1) diminished BMP9-induced ALP activity. Moreover, it was demonstrated that β-catenin overexpression promoted BMP9-induced mineralization, and increased the expression levels of late osteogenic markers osteopontin and osteocalcin. Furthermore, β-catenin inhibited BMP9-induced lipid accumulation and the adipogenic marker adipocyte fatty acid binding protein (aP2). The cell-implantation assay results identified that β-catenin not only augmented BMP9-induced ectopic bone formation, but also blocked adipogenesis in vivo. Mechanistically, it was found that β-catenin and BMP9 synergistically stimulated the osteogenic transcription factors runt-related transcription factor 2 (Runx2) and Osterix (OSX). However, BMP9-induced adipogenic transcription factors, peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT enhancer-binding protein α (C/EBPα), were inhibited by β-catenin. Therefore, these findings suggested that the Wnt/β-catenin signaling, potentially via the modulation of osteogenic and adipogenic transcriptional factors, exerts an opposite effect on BMP9-induced osteogenic and adipogenic differentiation in preadipocytes.
Collapse
Affiliation(s)
- Kailu Liang
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Yu Du
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Liang Chen
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Liyuan Wang
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Ruidong Li
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Zhengjian Yan
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Yang Liu
- Department of Orthopaedic Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
199
|
Schupbach D, Comeau-Gauthier M, Harvey E, Merle G. Wnt modulation in bone healing. Bone 2020; 138:115491. [PMID: 32569871 DOI: 10.1016/j.bone.2020.115491] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
Abstract
Genetic studies have been instrumental in the field of orthopaedics for finding tools to improve the standard management of fractures and delayed unions. The Wnt signaling pathway that is crucial for development and maintenance of many organs also has a very promising pathway for enhancement of bone regeneration. The Wnt pathway has been shown to have a direct effect on stem cells during bone regeneration, making Wnt a potential target to stimulate bone repair after trauma. A more complete view of how Wnt influences animal bone regeneration has slowly come to light. This review article provides an overview of studies done investigating the modulation of the canonical Wnt pathway in animal bone regeneration models. This not only includes a summary of the recent work done elucidating the roles of Wnt and β-catenin in fracture healing, but also the results of thirty transgenic studies, and thirty-eight pharmacological studies. Finally, we discuss the discontinuation of sclerostin clinical trials, ongoing clinical trials with lithium, the results of Dkk antibody clinical trials, the shift into combination therapies and the future opportunities to enhance bone repair and regeneration through the modulation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Drew Schupbach
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Experimental Surgery, Faculty of Medicine, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A7-117, Montreal, Québec H3G 1A4, Canada.
| | - Marianne Comeau-Gauthier
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Experimental Surgery, Faculty of Medicine, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A7-117, Montreal, Québec H3G 1A4, Canada.
| | - Edward Harvey
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada.
| | - Geraldine Merle
- Department of Surgery, Division of Orthopedic Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room A10-110, Montreal, Québec H3G 1A4, Canada; Department of Chemical Engineering, Polytechnique Montreal, 2500, chemin de Polytechnique, Montréal, Québec H3T 1J4, Canada.
| |
Collapse
|
200
|
Min Swe NM, Kobayashi Y, Kamimoto H, Moriyama K. Aberrantly activated Wnt/β-catenin pathway co-receptors LRP5 and LRP6 regulate osteoblast differentiation in the developing coronal sutures of an Apert syndrome (Fgfr2 S252W /+ ) mouse model. Dev Dyn 2020; 250:465-476. [PMID: 32822074 DOI: 10.1002/dvdy.239] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Apert syndrome is an autosomal, dominant inherited disorder characterized by craniosynostosis and syndactyly caused by gain-of-function mutations in the fibroblast growth factor receptor 2 (FGFR2) gene. Wnt/β-catenin signaling plays critical roles in regulating the skeletal development. Here, we analyzed the role of this pathway in the developing coronal sutures (CS) of a murine Apert syndrome model (Fgfr2S252W/+ ). RESULTS We observed aberrantly increased mRNA expression of Lrp5 and Lrp6 in CS of Fgfr2S252W/+ mice, whereas both wild type (WT) and Fgfr2S252W/+ mice showed similar expression of other Wnt/β-catenin-related genes, such as Wnt3, Wnt3a, Fzd4, Fzd6, Axin2, and Dkk1 as evidenced by in situ hybridization. Significantly increased Lrp5 and Lrp6 mRNA expression was observed by quantitative PCR analysis of cultured cells isolated from CS of Fgfr2S252W/+ mice. Phospho-LRP5, phospho-LRP6, and non-phospho-β-catenin were upregulated in Fgfr2S252W/+ CS compared with that in WT CS. Short-interfering RNA targeting Lrp5 and Lrp6 significantly reduced runt-related transcription factor 2, collagen type 1 alpha 1, and osteocalcin mRNA expression, and alkaline phosphatase activity in cultured cells. CONCLUSIONS The Wnt/β-catenin pathway was activated in the CS of Fgfr2S252W/+ mice during craniofacial development, suggesting the involvement of the Wnt/β-catenin pathway in the pathogenesis of CS synostosis in Fgfr2S252W/+ mice.
Collapse
Affiliation(s)
- Nay Myo Min Swe
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yukiho Kobayashi
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hiroyuki Kamimoto
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|