1
|
van Velsen EFS, Wijnen M, Muradin GSR, Zillikens MC. Incident Vertebral Fractures During Romosozumab Treatment in a Patient With a Pathogenic LRP5 Variant. JCEM CASE REPORTS 2025; 3:luae238. [PMID: 39726666 PMCID: PMC11669863 DOI: 10.1210/jcemcr/luae238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Indexed: 12/28/2024]
Abstract
A defect in the canonical Wnt-β-catenin pathway may lead to reduced bone strength and increased fracture risk. Sclerostin is a key inhibitor of this pathway by binding to low-density lipoprotein (LDL) receptor-related protein (LRP)-5/6, thereby reducing bone formation. The effectiveness of romosozumab, a human monoclonal antibody that binds sclerostin and prevents this inhibitory effect, has been questioned in patients with inactivating genetic variants in LRP5 or LRP6. We present a 67-year-old woman with severe osteoporosis with 4 grade 2 vertebral fractures due to a heterozygous pathogenic variant in LRP5. She was treated with romosozumab for 1 year, after which a routine follow-up spine x-ray revealed 5 new vertebral fractures, despite a strong increase in bone mineral density (BMD) (lumbar spine [LS] + 58%; femur neck [FN] + 23%), although overestimated at LS because of the vertebral fractures. This suggests that in patients with loss-of-function LRP5 variants, romosozumab is able to increase BMD. However, it is unclear whether the progressive vertebral fractures are due to the severe osteoporosis in relation to the start of romosozumab or a diminished responsiveness related to her LRP5 variant. Further evaluation is needed on the effect of romosozumab on BMD and fracture outcomes in patients with a likely defective LRP5/6 receptor.
Collapse
Affiliation(s)
- Evert F S van Velsen
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center, 3015 CE, Rotterdam, the Netherlands
- Erasmus MC Bone Center, Erasmus Medical Center, University Medical Center, 3015 CE, Rotterdam, the Netherlands
| | - Mark Wijnen
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center, 3015 CE, Rotterdam, the Netherlands
- Erasmus MC Bone Center, Erasmus Medical Center, University Medical Center, 3015 CE, Rotterdam, the Netherlands
| | - Galied S R Muradin
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, University Medical Center, 3015 CE, Rotterdam, the Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center, 3015 CE, Rotterdam, the Netherlands
- Erasmus MC Bone Center, Erasmus Medical Center, University Medical Center, 3015 CE, Rotterdam, the Netherlands
| |
Collapse
|
2
|
Sillence DO. A Dyadic Nosology for Osteogenesis Imperfecta and Bone Fragility Syndromes 2024. Calcif Tissue Int 2024; 115:873-890. [PMID: 38942908 PMCID: PMC11607092 DOI: 10.1007/s00223-024-01248-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024]
Abstract
In 2023 following extensive consultation with key stakeholders, the expert Nosology Working Group of the International Skeletal Dysplasia Society (ISDS) published the new Dyadic Nosology for Genetic Disorders of the Skeleton. Some 770 entities were delineated associated with 552 genes. From these entities, over 40 genes resulting in distinct forms of Osteogenesis Imperfecta (OI) and Bone Fragility and/or Familial Osteoporosis were identified. To assist clinicians and lay stake holders and bring the considerable body of knowledge of the matrix biology and genomics to people with OI as well as to clinicians and scientists, a dyadic nosology has been recommended. This combines a genomic co-descriptor with a phenotypic naming based on the widely used Sillence nosology for the OI syndromes and the many other syndromes characterized in part by bone fragility.This review recapitulates and explains the evolution from the simple Congenita and Tarda subclassification of OI in the 1970 nosology, which was replaced by the Sillence types I-IV nosology which was again replaced in 2009 with 5 clinical groups, type 1 to 5. Qualitative and quantitative defects in type I collagen polypeptides were postulated to account for the genetic heterogeneity in OI for nearly 30 years, when OI type 5, a non-collagen disorder was recognized. Advances in matrix biology and genomics since that time have confirmed a surprising complexity both in transcriptional as well as post-translational mechanisms of collagens as well as in the many mechanisms of calcified tissue homeostasis and integrity.
Collapse
Affiliation(s)
- David Owen Sillence
- Specialities of Genomic Medicine and Paediatrics and Adolescent Health, Children's Hospital Westmead, Sydney University Clinical School, Westmead, NSW, 2145, Australia.
- Department of Genetic Medicine, Honorary Emeritus Consultant, Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
3
|
Jovanovic M, Marini JC. Update on the Genetics of Osteogenesis Imperfecta. Calcif Tissue Int 2024; 115:891-914. [PMID: 39127989 PMCID: PMC11607015 DOI: 10.1007/s00223-024-01266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024]
Abstract
Osteogenesis imperfecta (OI) is a heterogeneous heritable skeletal dysplasia characterized by bone fragility and deformity, growth deficiency, and other secondary connective tissue defects. OI is now understood as a collagen-related disorder caused by defects of genes whose protein products interact with collagen for folding, post-translational modification, processing and trafficking, affecting bone mineralization and osteoblast differentiation. This review provides the latest updates on genetics of OI, including new developments in both dominant and rare OI forms, as well as the signaling pathways involved in OI pathophysiology. There is a special emphasis on discoveries of recessive mutations in TENT5A, MESD, KDELR2 and CCDC134 whose causality of OI types XIX, XX, XXI and XXI, respectively, is now established and expends the complexity of mechanisms underlying OI to overlap LRP5/6 and MAPK/ERK pathways. We also review in detail new discoveries connecting the known OI types to each other, which may underlie an eventual understanding of a final common pathway in OI cellular and bone biology.
Collapse
Affiliation(s)
- Milena Jovanovic
- Section on Heritable Disorders of Bone and Extracellular Matrix, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Section on Adolescent Bone and Body Composition, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Joan C Marini
- Section on Heritable Disorders of Bone and Extracellular Matrix, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
4
|
Mancini M, Chapurlat R, Isidor B, Desjonqueres M, Couture G, Guggenbuhl P, Coutant R, El Chehadeh S, Fradin M, Frazier A, Goldenberg A, Guillot P, Koumakis E, Mehsen-Cêtre N, Rossi M, Schaefer É, Sigaudy S, Porquet-Bordes V, Fontanges É, Letard P, Edouard T, Javier RM, Cohen-Solal M, Funck-Brentano T, Collet C. Early-Onset Osteoporosis: Molecular Analysis in Large Cohort and Focus on the PLS3 Gene. Calcif Tissue Int 2024; 115:591-598. [PMID: 39316135 DOI: 10.1007/s00223-024-01288-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024]
Abstract
Osteoporosis is a skeletal disorder characterized by abnormal bone microarchitecture and low bone mineral density (BMD), responsible for an increased risk of fractures and skeletal fragility. It is a common pathology of the aging population. However, when osteoporosis occurs in children or young adults, it strongly suggests an underlying genetic etiology. Over the past two decades, several genes have been identified as responsible for this particular kind of considered monogenic early-onset osteoporosis (EOOP) or juvenile osteoporosis, the main ones being COL1A1, COL1A2, LRP5, LRP6, WNT1, and more recently PLS3. In this study, the objective was to characterize a large cohort of patients diagnosed with primary osteoporosis and to establish its diagnosis yield. The study included 577 patients diagnosed with primary osteoporosis and its diagnosis yield was established. To this end, next-generation sequencing (NGS) of a panel of 21 genes known to play a role in bone fragility was carried out. A genetic etiology was explained in about 18% of cases, while the others remain unexplained. The most frequently identified gene associated with EOOP is LRP5, which was responsible for 8.2% of the positive results (47 patients). As unexpected, 17 patients (2.9%) had a variant in PLS3 which encodes plastin 3. Alterations of PLS3 are associated with dominant X-linked osteoporosis, an extremely rare disease. Given the rarity of this disease, we focused on it. It was observed that males were more affected than females, but it is noteworthy that three females with a particularly severe phenotype were identified. Of these three, two had a variant in an additional gene involved in EOP, illustrating the probable existence of digenism. We significantly increase the number of variants potentially associated with EOOP, especially in PLS3. The results of our study demonstrate that molecular analysis in EOOP is beneficial and useful.
Collapse
Affiliation(s)
- Maxence Mancini
- Biochemistry and Molecular Genetics Department, Lariboisière Hospital, AP-HP, Paris, France
| | - Roland Chapurlat
- Rheumatology and Bone Pathology Department, Inserm UMR 1033, Université de Lyon, Edouard Herriot Hospital, HCL, Lyon, France
| | - Bertrand Isidor
- Medical Genetics Department, CHU de Nantes, Hôtel Dieu Hospital, Nantes, France
| | - Marine Desjonqueres
- Nephrology - Rheumatology - Dermatology Paediatric Department, Edouard Herriot Hospital, HCL, Lyon, France
| | - Guillaume Couture
- Endocrine, Bone Diseases and Genetics Unit, Rheumatology Department, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, University Hospital, RESTORE, INSERM U1301, Toulouse, France
| | | | - Régis Coutant
- Department of Paediatrics and Endocrinology, CHU d'Angers, Angers, France
| | - Salima El Chehadeh
- Medical Genetics Department, Institut de Génétique Médicale d'alsace, CHU de Strasbourg, Strasbourg, France
| | - Mélanie Fradin
- Clinical Genetics Department, CHU Rennes, Sud Hospital, Rennes, France
| | - Aline Frazier
- Rheumatology Department, Inserm 1132, Univsersité Paris-Cité, Lariboisière Hospital, AP-HP, Paris, France
| | - Alice Goldenberg
- Medical Genetics Department, Charles- Nicolle Hospital, CHU de Rouen, Rouen, France
| | - Pascaline Guillot
- Rheumatology Department, CHU de Nantes, Hôpital Hôtel Dieu, Nantes, France
| | | | | | - Massimiliano Rossi
- Medical Genetics Department, Edouard Herriot Hospital, HCL, Lyon, France
| | - Élise Schaefer
- Medical Genetics Department, Institut de Génétique Médicale d'alsace, CHU de Strasbourg, Strasbourg, France
| | - Sabine Sigaudy
- Medical Genetics Department, CHU de Marseille, Timone Hospital, Marseille, France
| | - Valérie Porquet-Bordes
- Endocrine, Bone Diseases and Genetics Unit, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, Paediatric Research Unit, Children's Hospital, Toulouse University Hospital, RESTORE, INSERM U1301, Toulouse, France
| | - Élisabeth Fontanges
- Rheumatology and Bone Pathology Department, Inserm UMR 1033, Université de Lyon, Edouard Herriot Hospital, HCL, Lyon, France
| | - Pauline Letard
- Medical Genetics Department, CHU de Poitiers, Poitiers, France
| | - Thomas Edouard
- Endocrine, Bone Diseases and Genetics Unit, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, Paediatric Research Unit, Children's Hospital, Toulouse University Hospital, RESTORE, INSERM U1301, Toulouse, France
| | - Rose-Marie Javier
- Rheumatology Department, CHU de Strasbourg, Hautepierre Hospital, Strasbourg, France
| | - Martine Cohen-Solal
- Rheumatology Department, Inserm 1132, Univsersité Paris-Cité, Lariboisière Hospital, AP-HP, Paris, France
| | - Thomas Funck-Brentano
- Rheumatology Department, Inserm 1132, Univsersité Paris-Cité, Lariboisière Hospital, AP-HP, Paris, France
| | - Corinne Collet
- Rare Disease Genomic Medicine Department, CHU Necker-Enfants Malades, INSERM UMR1163, Institut Imagine, Université Paris-Cité, Paris, France.
| |
Collapse
|
5
|
Shen X, Feng S, Chen S, Gong B, Wang S, Wang H, Song D, Ni J. Wnt3a-induced LRP6 phosphorylation enhances osteoblast differentiation to alleviate osteoporosis through activation of mTORC1/β-catenin signaling. Arch Biochem Biophys 2024; 761:110169. [PMID: 39362316 DOI: 10.1016/j.abb.2024.110169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
OBJECTIVE Osteoporosis (OP) is a common cause of morbidity and mortality in older individuals. The importance of Wnt3a in osteogenic activity and bone tissue homeostasis is well known. Here, we explored the possible molecular mechanism by which Wnt3a mediates the LRP6/mTORC1/β-catenin axis to regulate osteoblast differentiation in OP. METHODS OP-related key genes were identified through a bioinformatics analysis. A ROS17/2.8 cell differentiation system for rat osteogenic progenitors and a rat model of senile OP were constructed for in vitro and in vivo mechanism verification. RESULTS Bioinformatics analysis revealed that LRP6 was poorly expressed in OP and may play a key role in the occurrence of OP by affecting osteoblast differentiation. LRP6 knockdown inhibited osteoblast differentiation in an in vitro model. In addition, Wnt3a promoted osteoblast differentiation by inducing LRP6 phosphorylation. Moreover, LRP6 promoted mTORC1 expression, which indirectly promoted β-catenin expression, thus promoting osteoblast differentiation. Finally, an in vivo assay revealed that LRP6 inhibition improved OP. CONCLUSION Our study provides evidence that Wnt3a induces phosphorylation of LRP6 to activate the mTORC1/β-catenin axis, thus promoting osteoblast differentiation and ultimately improving OP in aged rats.
Collapse
Affiliation(s)
- Xiang Shen
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, China
| | - Shuolin Feng
- Department of Orthopedics, The Fourth Hospital of Changsha, China
| | - Shanbin Chen
- Department of Orthopedics, The Fourth Hospital of Changsha, China
| | - Bin Gong
- Department of Orthopedics, The Fourth Hospital of Changsha, China
| | - Suiyuan Wang
- Department of Orthopedics, The Fourth Hospital of Changsha, China
| | - Huan Wang
- Department of Orthopedics, The Fourth Hospital of Changsha, China
| | - Deye Song
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, China.
| | - Jiangdong Ni
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, China.
| |
Collapse
|
6
|
Bartosik M, Simon A, Busse B, Barvencik F, Amling M, Oheim R, von Brackel FN. Sex-Specific Association Patterns of Bone Microstructure and Lower Leg Arterial Calcification. Calcif Tissue Int 2024; 115:636-647. [PMID: 39397150 PMCID: PMC11531430 DOI: 10.1007/s00223-024-01299-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
In conversations about bone loss and the importance of calcium homeostasis, patients frequently inquire about the association with arterial calcifications. Although a relationship between bone loss and the occurrence of vascular calcifications is suspected, it is not yet fully investigated and understood. This study aims to analyze associations between bone mineralization, structure, and vascular calcification at the lower leg in patients with low bone mineral density in HR-pQCT. We retrospectively analyzed 774 high-resolution quantitative computed tomography (HR-pQCT) scans of the distal tibia for the presence of vascular calcifications. After sex-specific propensity score matching for age and BMI to account for confounders, 132 patients remained for quantification of bone microstructure, bone density, lower leg arterial calcification (LLAC), and laboratory parameters of bone turnover. The interactions between bone parameters and vascular calcification were quantified by regression analyses. The calcium metabolism was not different between individuals with and without LLAC, nor oral calcium supplementation. Female patients with LLAC had a higher cortical perimeter (p = 0.016) compared to female patients without LLAC, whereas male patients with LLAC had lower cortical pore diameter than male patients without LLAC (p = 0.027). The appearance of LLAC was sex specifically associated with bone parameters. In female patients, only plaque density was associated with HR-pQCT bone parameters and age, whereas in male patients, plaque volume was associated with HR-pQCT parameters of the distal tibia. Female patients exhibit an increasing plaque density depended on age and trabecular thinning. Decreasing cortical pore diameter and trabecular number along with increasing bone mineralization are linked to increasing plaque volume in male patients.
Collapse
Affiliation(s)
- Mikolaj Bartosik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Alexander Simon
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Division of Orthopaedics, Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Interdisciplinary Competence Center for Interface Research (ICCIR), Hamburg, Germany
| | - Florian Barvencik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Felix N von Brackel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany.
- Interdisciplinary Competence Center for Interface Research (ICCIR), Hamburg, Germany.
| |
Collapse
|
7
|
Vaghasia N, Dutta A, Mithal A. LRP5 Variant Without Pseudoglioma in a Young Man With Fragility Fractures. JCEM CASE REPORTS 2024; 2:luae163. [PMID: 39309619 PMCID: PMC11414401 DOI: 10.1210/jcemcr/luae163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Indexed: 09/25/2024]
Abstract
Osteoporosis in children and young adults is relatively rare. Hereditary causes are often overlooked in the absence of a positive family history. We report a 29-year-old male presenting with recurrent fragility fractures since 6 years of age. Secondary causes, such as celiac disease, inflammatory disorders, and hypogonadism, were ruled out. Family history was negative for any bone disease. Exome sequencing revealed 2 variants of LRP5 gene-intron 5 c.1015 + 1G > A and exon 5 c.892C > T. Although the former variant has been described in literature as a cause of osteoporosis in homozygous state only, it manifested as osteoporosis in our patient, in the heterozygous state, in presence of a second variant of uncertain significance. However, eye involvement, which is classically seen in "osteoporosis-pseudoglioma syndrome" homozygote, was absent in our patient. Genetic analysis of the parents revealed father to be a carrier of intron 5 c.1015 + 1G > A and mother exon 5 c.892C > T variants of the LRP5 gene. However, none of them had osteoporosis on bone densitometry. The patient was subsequently treated with IV zoledronic acid (planned to be administered annually) and showed improvement in bone density by 11% at the spine and 9.5% at the left femur; there were no further fractures over 1 year of follow-up.
Collapse
Affiliation(s)
- Nupoor Vaghasia
- Institute of Endocrinology and Diabetes, Max Healthcare, Saket, New Delhi 110017, India
| | - Aditya Dutta
- Institute of Endocrinology and Diabetes, Max Healthcare, Saket, New Delhi 110017, India
| | - Ambrish Mithal
- Institute of Endocrinology and Diabetes, Max Healthcare, Saket, New Delhi 110017, India
| |
Collapse
|
8
|
Wade E, Mulholland K, Shaw I, Cundy T, Robertson S. Idiopathic juvenile osteoporosis-a polygenic disorder? JBMR Plus 2024; 8:ziae099. [PMID: 39193113 PMCID: PMC11347881 DOI: 10.1093/jbmrpl/ziae099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/01/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Idiopathic juvenile osteoporosis (IJO) is a rare condition presenting with vertebral and metaphyseal fractures that affects otherwise healthy prepubertal children. Bone mineral density (BMD) measurements are very low. The primary problem appears to be deficient bone formation, with a failure to accrue bone normally during growth. The onset in childhood suggests IJO is a genetic disorder, and a number of reports indicate that some children carry heterozygous pathogenic variants in genes known to be associated with defective osteoblast function and low bone mass, most commonly LRP5 or PLS3. However, a positive family history is unusual in IJO, suggesting the genetic background can be complex. We describe a young man with classical IJO who was investigated with a bone fragility gene panel and whole genome sequencing. The proband was found to carry four variants in three different genes potentially affecting osteoblast function. From his mother he had inherited mutations in ALPL (p.Asn417Ser) and LRP5 (p.Arg1036Gln), and from his father mutations in LRP5 (p.Asp1551Alsfs*13) and activating transcription factor 4 (ATF4) (p.Leu306Ile). His sister had also inherited the LRP5 (p.Asp1551Alsfs*13) from her father, but not the ATF4 mutation. Their spinal BMD z-scores differed substantially (sister -1.6, father -3.2) pointing to the potential importance of the ATF4 mutation. Activating transcription factor 4 acts downstream from RUNX2 and osterix and plays an important role in osteoblast differentiation and function. This case, together with others recently published, supports the view that IJO can result from clustering of mutations in genes related to osteoblast development and function. Novel genes in these pathways may be involved. Our case also emphasizes the value of detailed study of other family members. After a bone biopsy had excluded a mineralization defect due to hypophosphatasia, the proband was treated with zoledronate infusions with good clinical effect.
Collapse
Affiliation(s)
- Emma Wade
- Department of Women’s & Children’s Health, Dunedin School of Medicine, University of Otago, Dunedin, 9016, New Zealand
| | - Katie Mulholland
- Department of Women’s & Children’s Health, Dunedin School of Medicine, University of Otago, Dunedin, 9016, New Zealand
| | - Ian Shaw
- Department of Pediatrics, Southland Hospital, Invercargill, 9812, New Zealand
| | - Tim Cundy
- Department of Medicine, Faculty of Medical & Health Sciences, University of Auckland, Auckland, 1023, New Zealand
| | - Stephen Robertson
- Department of Women’s & Children’s Health, Dunedin School of Medicine, University of Otago, Dunedin, 9016, New Zealand
| |
Collapse
|
9
|
Shimazaki S, Sato J. Idiopathic Juvenile Osteoporosis: A Case Report and Literature Review. Cureus 2024; 16:e68361. [PMID: 39355452 PMCID: PMC11443356 DOI: 10.7759/cureus.68361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 10/03/2024] Open
Abstract
This case report describes the rare occurrence of idiopathic juvenile osteoporosis (IJO) in an 11-year-old boy with bone fragility and fractures, particularly in the thoracic and lumbar vertebrae. After excluding discernible underlying causes, the diagnosis was confirmed using clinical and radiological assessments. Treatment commenced with oral bisphosphonates, leading to notable bone mineral density (BMD) improvements and the absence of subsequent fractures. IJO presents diagnostic challenges owing to its multifaceted nature, necessitating the exclusion of other common causes of pediatric osteoporosis. Although the pathophysiology of IJO remains poorly understood, this case underscores the potential efficacy of bisphosphonate therapy in managing the condition and improving patient outcomes. Notably, the patient's symptoms ameliorated as puberty commenced, aligning with the typical IJO patterns reported in the literature. Although the long-term impact of bisphosphonate treatment in pediatric IJO cases warrants further investigation, this case exemplifies the potential to enhance the quality of life of affected individuals.
Collapse
Affiliation(s)
- Shunsuke Shimazaki
- Department of Paediatrics, Funabashi Municipal Medical Center, Funabashi, JPN
| | - Junichi Sato
- Department of Paediatrics, Funabashi Municipal Medical Center, Funabashi, JPN
| |
Collapse
|
10
|
Zanchetta MB, Corsi A. Bone biopsy for the diagnosis of osteomalacia. Can we avoid it? J Bone Miner Res 2024; 39:515-516. [PMID: 38613689 DOI: 10.1093/jbmr/zjae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 04/15/2024]
Affiliation(s)
- Maria Belen Zanchetta
- Instituto de Diagnostico e Investigaciones Metabólicas (IDIM), Universidad del Salvador, Buenos Aires 1012, Argentina
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| |
Collapse
|
11
|
Schmidt FN, Delsmann J, Yazigi B, Beil FT, Amling M, Oheim R. Approaching virtual osteoid volume estimation and in-depth tissue characterization in patients with tumor-induced osteomalacia. J Bone Miner Res 2024; 39:116-129. [PMID: 38477742 PMCID: PMC11240151 DOI: 10.1093/jbmr/zjae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/08/2023] [Accepted: 01/12/2024] [Indexed: 03/14/2024]
Abstract
Tumor-induced osteomalacia (TIO) poses a significant diagnostic challenge, leading to increased disease duration and patient burden also by missing clinical suspicion. Today, diagnosis of osteomalacia relies on invasive iliac crest biopsy, if needed. Therefore, a noninvasive method would be beneficial for patients with severe osteomalacia, such as TIO, to inform their clinical management and address specific needs, like estimating the regeneration capacity at high osteoid volumes (OVs) or the potential of a hungry bone syndrome after tumor removal. Furthermore, given the lack of comprehensive histological characterization of TIO, there is a need for additional tissue characterization. Therefore, our assessment encompassed iliac crest biopsies that were examined using quantitative electron backscattered microscopy, Raman spectroscopy, micro-computed tomography, and histology to analyze the biopsy tissue. Our clinical assessment encompassed DXA and high-resolution peripheral quantitative computed tomography (HR-pQCT) alongside with biochemical analyses and clinical evaluations. Combining imaging and clinical data, we established a model to predict the OV. We compared 9 TIO patients with 10 osteoporosis (OPO) patients and 10 healthy controls. Histological analyses confirmed a pronounced OV in TIO patients (OPO: 1.20% ± 1.23% vs TIO: 23.55% ± 12.23%, P < .0005), and spectroscopy revealed lower phosphate levels in TIO biopsies. By combining HR-pQCT and laboratory diagnostics, we developed a linear regression model to noninvasively predict the OV revealing significantly higher modeled OV/BVmodel values of 24.46% ± 14.22% for TIO compared to the control group (5.952% ± 3.44%, P ≤ .001). By combining laboratory diagnostics, namely, ALP and Tt.BMDRadius measured by HR-pQCT, we achieved the calculation of the virtual osteoid volume to bone volume ratio (OV/BVmodel) with a significant correlation to histology as well as reliable identification of TIO patients compared to OPO and control. This novel approach is potentially helpful for predicting OV by noninvasive techniques in diagnostic procedures and improving the clinical management of TIO.
Collapse
Affiliation(s)
- Felix N Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Julian Delsmann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Bashar Yazigi
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Frank Timo Beil
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| |
Collapse
|
12
|
Stringer F, Sims NA, Sachithanandan N, Aleksova J. Severe Osteoporosis With Pathogenic LRP5 Variant. JCEM CASE REPORTS 2024; 2:luae021. [PMID: 38404691 PMCID: PMC10888517 DOI: 10.1210/jcemcr/luae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Indexed: 02/27/2024]
Abstract
A 24-year-old female patient was diagnosed with osteoporosis after presenting with numerous fractures throughout her childhood and adolescence. Risk factors included chronic constipation, severe vitamin D deficiency, and long-term high-dose steroid use for severe eczema. Metabolic bone disorder clinical exome screening (limited panel of metabolic bone disorders and gastrointestinal disorders) was undertaken and revealed a class 4 likely pathogenic variant in the LRP5 gene known to cause osteoporosis. Optimal treatment for patients with this variant is not well defined. A literature review of the condition and potential treatment options is discussed.
Collapse
Affiliation(s)
- Felicity Stringer
- Department of Endocrinology, St Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC 3065, Australia
| | - Natalie A Sims
- St Vincent's Institute of Medical Research, Fitzroy, Melbourne, VIC 3065, Australia
- Melbourne Medical School, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Nirupa Sachithanandan
- Department of Endocrinology, St Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC 3065, Australia
- Melbourne Medical School, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Jasna Aleksova
- Department of Endocrinology, St Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC 3065, Australia
- Department of Medicine, Monash University, Clayton, VIC 3168, Australia
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| |
Collapse
|
13
|
Smit A, Meijer O, Winter E. The multi-faceted nature of age-associated osteoporosis. Bone Rep 2024; 20:101750. [PMID: 38566930 PMCID: PMC10985042 DOI: 10.1016/j.bonr.2024.101750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Age-associated osteoporosis (AAOP) poses a significant health burden, characterized by increased fracture risk due to declining bone mass and strength. Effective prevention and early treatment strategies are crucial to mitigate the disease burden and the associated healthcare costs. Current therapeutic approaches effectively target the individual contributing factors to AAOP. Nonetheless, the management of AAOP is complicated by the multitude of variables that affect its development. Main intrinsic and extrinsic factors contributing to AAOP risk are reviewed here, including mechanical unloading, nutrient deficiency, hormonal disbalance, disrupted metabolism, cognitive decline, inflammation and circadian disruption. Furthermore, it is discussed how these can be targeted for prevention and treatment. Although valuable as individual targets for intervention, the interconnectedness of these risk factors result in a unique etiology for every patient. Acknowledgement of the multifaceted nature of AAOP will enable the development of more effective and sustainable management strategies, based on a holistic, patient-centered approach.
Collapse
Affiliation(s)
- A.E. Smit
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - O.C. Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
| | - E.M. Winter
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, the Netherlands
- Department of Medicine, Center for Bone Quality, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
14
|
Formosa MM, Christou MA, Mäkitie O. Bone fragility and osteoporosis in children and young adults. J Endocrinol Invest 2024; 47:285-298. [PMID: 37668887 PMCID: PMC10859323 DOI: 10.1007/s40618-023-02179-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023]
Abstract
Osteoporosis is a metabolic bone disorder which increases fragility fracture risk. Elderly individuals, especially postmenopausal women, are particularly susceptible to osteoporosis. Although rare, osteoporosis in children and young adults is becoming increasingly evident, highlighting the need for timely diagnosis, management and follow-up. Early-onset osteoporosis is defined as the presence of a low BMD (Z-score of ≤ -2.0 in individuals aged < 20 years; T-score of ≤ -2.5 in those aged between 20 to 50 years) accompanied by a clinically significant fracture history, or the presence of low-energy vertebral compression fractures even in the absence of osteoporosis. Affected children and young adults should undergo a thorough diagnostic workup, including collection of clinical history, radiography, biochemical investigation and possibly bone biopsy. Once secondary factors and comorbidities are excluded, genetic testing should be considered to determine the possibility of an underlying monogenic cause. Defects in genes related to type I collagen biosynthesis are the commonest contributors of primary osteoporosis, followed by loss-of-function variants in genes encoding key regulatory proteins of canonical WNT signalling (specifically LRP5 and WNT1), the actin-binding plastin-3 protein (encoded by PLS3) resulting in X-linked osteoporosis, and the more recent sphingomyelin synthase 2 (encoded by SGMS2) which is critical for signal transduction affecting sphingomyelin metabolism. Despite these discoveries, genetic causes and underlying mechanisms in early-onset osteoporosis remain largely unknown, and if no causal gene is identified, early-onset osteoporosis is deemed idiopathic. This calls for further research to unravel the molecular mechanisms driving early-onset osteoporosis that consequently will aid in patient management and individualised targeted therapy.
Collapse
Affiliation(s)
- M M Formosa
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
- Center for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M A Christou
- Department of Endocrinology, School of Medicine, University of Ioannina, Ioannina, Greece
- Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, Ioannina, Greece
| | - O Mäkitie
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Folkhälsan Research Centre, Folkhälsan Institute of Genetics, Helsinki, Finland.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
15
|
Bartosik M, Simon A, Strahl A, Oheim R, Amling M, Schmidt FN. Comparison of Motion Grading in 1,000 Patients by First- and Second-Generation HR-pQCT: A Propensity Score Matched Cohort Study. Calcif Tissue Int 2023; 113:597-608. [PMID: 37880520 PMCID: PMC10673987 DOI: 10.1007/s00223-023-01143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
In-vivo bone microstructure measured by high-resolution peripheral quantitative computed tomography (HR-pQCT) is gaining importance in research and clinical practice. Second-generation HR-pQCT (XCT2) shows improved image quality and shorter measurement duration compared to the first generation (XCT1). Predicting and understanding the occurrence of motion artifacts is crucial for clinical practice. We retrospectively analyzed data from HR-pQCT measurements at the distal radius and tibia of 1,000 patients (aged 20 to 89) evenly distributed between both generations of HR-pQCT. Motion artifacts were graded between 1 (no motion) and 5 (severe motion), with grades greater 3 considered unusable. Additionally, baseline characteristics and patients' muscle performance and balance were measured. Various group comparisons between the two generations of HR-pQCT and regression analyses between patient characteristics and motion grading were performed. The study groups of XCT1 and XCT2 did not differ by age (XCT1: 64.9 vs. XCT2: 63.8 years, p = 0.136), sex (both 74.5% females, p > 0.999), or BMI (both 24.2 kg/m2, p = 0.911) after propensity score matching. XCT2 scans exhibited significantly lower motion grading in both extremities compared to XCT1 (Radius: p < 0.001; Tibia: p = 0.002). In XCT2 motion-corrupted scans were more than halved at the radius (XCT1: 35.3% vs. XCT2: 15.5%, p < 0.001), and at the tibia the frequency of best image quality scans was increased (XCT1: 50.2% vs. XCT2: 63.7%, p < 0.001). The strongest independent predictor for motion-corrupted images is the occurrence of high motion grading at the other scanning site during the same consultation. The association between high motion grading in one scan and a corresponding high motion grading in another scan within the same session suggests a non-resting patient. Additionally, aged, female, and patients with smaller stature tend towards higher motion grading, requiring special attention to a correct extremity fixation.
Collapse
Affiliation(s)
- Mikolaj Bartosik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Simon
- Division of Orthopaedics, Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - André Strahl
- Division of Orthopaedics, Department of Trauma and Orthopaedic Surgery, 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
| | - Felix N Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
16
|
Heidari A, Homaei A, Saffari F. Novel Homozygous Nonsense Mutation in the LRP5 Gene in Two Siblings with Osteoporosis-pseudoglioma Syndrome. J Clin Res Pediatr Endocrinol 2023; 15:318-323. [PMID: 34965700 PMCID: PMC10448547 DOI: 10.4274/jcrpe.galenos.2021.2021.0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 11/25/2021] [Indexed: 12/01/2022] Open
Abstract
Osteoporosis-pseudoglioma syndrome (OPPG) is a rare autosomal recessive disorder characterized by severe osteoporosis and eye abnormalities that lead to vision loss. In this study, clinical findings and genetic study of two siblings with OPPG are presented. Whole exome sequencing of DNA enriched for exonic regions was performed with SureSelect 38Mbp all exon kit v. 7.0. The two siblings presented with different clinical manifestations of OPPG. The younger female sibling had blindness and severe osteoporosis with multiple fractures, while her older brother was also blind but with less severe osteoporosis and no fractures. On analysis, a novel homozygous nonsense mutation (c.351G>A) in exon 2 of LRP5 (NM_002335) was found, predicted to change a tryptophan at 117 to a stop codon (p. Trp117Ter). Thus, a variable phenotype was associated with an identical variant in these two siblings. The novel mutation reported herein expands the spectrum of the underlying genetic pathology of OPPG.
Collapse
Affiliation(s)
- Abolfazl Heidari
- Reference Laboratory of Qazvin Medical University, Iran Sana Medical Genetics Laboratory, Qazvin, Iran
| | - Ali Homaei
- Shahid Beheshti University of Medical Sciences, Department of General Surgery, Tehran, Iran
| | - Fatemeh Saffari
- Qazvin University of Medical Sciences, Children Growth Research Center, Research Institute for Prevention of Non-Communicable Diseases, Department of Pediatric Endocrinology, Qazvin, Iran
| |
Collapse
|
17
|
Zhou W, van Rooij JGJ, van de Laarschot DM, Zervou Z, Bruggenwirth H, Appelman‐Dijkstra NM, Ebeling PR, Demirdas S, Verkerk AJMH, Zillikens MC. Prevalence of Monogenic Bone Disorders in a Dutch Cohort of Atypical Femur Fracture Patients. J Bone Miner Res 2023; 38:896-906. [PMID: 37076969 PMCID: PMC10946469 DOI: 10.1002/jbmr.4801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 02/12/2023] [Accepted: 03/07/2023] [Indexed: 04/21/2023]
Abstract
Atypical femur fractures (AFFs), considered rare associations of bisphosphonates, have also been reported in patients with monogenic bone disorders without bisphosphonate use. The exact association between AFFs and monogenic bone disorders remains unknown. Our aim was to determine the prevalence of monogenic bone disorders in a Dutch AFF cohort. AFF patients were recruited from two specialist bone centers in the Netherlands. Medical records of the AFF patients were reviewed for clinical features of monogenic bone disorders. Genetic variants identified by whole-exome sequencing in 37 candidate genes involved in monogenic bone disorders were classified based on the American College of Medical Genetics and Genomics (ACMG) classification guidelines. Copy number variations overlapping the candidate genes were also evaluated using DNA array genotyping data. The cohort comprises 60 AFF patients (including a pair of siblings), with 95% having received bisphosphonates. Fifteen AFF patients (25%) had clinical features of monogenic bone disorders. Eight of them (54%), including the pair of siblings, had a (likely) pathogenic variant in either PLS3, COL1A2, LRP5, or ALPL. One patient carried a likely pathogenic variant in TCIRG1 among patients not suspected of monogenic bone disorders (2%). In total, nine patients in this AFF cohort (15%) had a (likely) pathogenic variant. In one patient, we identified a 12.7 Mb deletion in chromosome 6, encompassing TENT5A. The findings indicate a strong relationship between AFFs and monogenic bone disorders, particularly osteogenesis imperfecta and hypophosphatasia, but mainly in individuals with symptoms of these disorders. The high yield of (likely) pathogenic variants in AFF patients with a clinical suspicion of these disorders stresses the importance of careful clinical evaluation of AFF patients. Although the relevance of bisphosphonate use in this relationship is currently unclear, clinicians should consider these findings in medical management of these patients. © 2023 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)
- Wei Zhou
- Department of Internal MedicineErasmus MCRotterdamThe Netherlands
| | | | | | - Zografia Zervou
- Department of Internal MedicineErasmus MCRotterdamThe Netherlands
| | | | - Natasha M Appelman‐Dijkstra
- Department of Internal Medicine, Division of EndocrinologyLeiden University Medical CenterLeidenThe Netherlands
| | - Peter R Ebeling
- Department of MedicineSchool of Clinical Sciences, Monash UniversityClaytonAustralia
| | - Serwet Demirdas
- Department of Clinical GeneticsErasmus MCRotterdamThe Netherlands
| | | | | |
Collapse
|
18
|
Zikán V. Osteoporosis in adults in clinical practice (1): diagnosis and differential diagnosis. VNITRNI LEKARSTVI 2023; 69:4-15. [PMID: 37468317 DOI: 10.36290/vnl.2023.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Osteoporosis is a systemic metabolic disease of the skeleton characterized by low bone strength that results in an increased risk of fracture. Fractures are associated with serious clinical consequences, including pain, disability, loss of independence, and death, as well as high healthcare costs. Early identification and intervention with patients at high risk for fracture is needed to reduce the burden of osteoporotic fractures. The identification of a patient at high risk of fracture should be followed by evaluation for factors contributing to low bone mineral density (BMD) and/or low bone quality, falls, and fractures. Components of the osteological evaluation include an assessment of BMD by dual-energy X-ray absorptiometry, osteoporosis-directed medical history and physical exam, laboratory studies, and possibly skeletal imaging. Disorders other than osteoporosis, requiring other types of treatment, may be found. This overview summarizes the basic procedures for the diagnosis and differential diagnosis of osteoporosis, which are necessary before starting treatment.
Collapse
|
19
|
Puente N, Vega AI, Hernandez JL, Fernandez-Luna JL, Riancho JA. An LRP6 mutation (Arg360His) associated with low bone mineral density but not cardiovascular events in a Caucasian family. Osteoporos Int 2022; 33:2445-2448. [PMID: 35840698 PMCID: PMC9568478 DOI: 10.1007/s00198-022-06494-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022]
Abstract
UNLABELLED We present a family with a rare mutation of the LRP6 gene and for the first time provide evidence for its association with low bone mineral density. INTRODUCTION The Wnt pathway plays a critical role in bone homeostasis. Pathogenic variants of the Wnt co-receptor LRP6 have been associated with abnormal skeletal phenotypes or increased risk of cardiovascular events. PATIENT AND METHODS Here we report an index premenopausal patient and her family carrying a rare missense LRP6 pathogenic variant (rs141212743; 0.0002 frequency among Europeans). This variant has been previously associated with metabolic syndrome and atherosclerosis, in the presence of normal bone mineral density. However, the LRP6 variant was associated with low bone mineral density in this family, without evidence for association with serum lipid levels or cardiovascular events. CONCLUSION Thus, this novel association shows that LRP6 pathogenic variants may be involved in some cases of early-onset osteoporosis, but the predominant effect, either skeletal or cardiovascular, may vary depending on the genetic background or other acquired factors.
Collapse
Affiliation(s)
- Nuria Puente
- Servicio de Medicina Interna, Hospital UM Valdecilla, Universidad de Cantabria, IDIVAL, Avda Valdecilla sn, 39008, Santander, Spain
| | - Ana I Vega
- Servicio de Genética, Hospital UM Valdecilla, IDIVAL, Santander, Spain
| | - José L Hernandez
- Servicio de Medicina Interna, Hospital UM Valdecilla, Universidad de Cantabria, IDIVAL, Avda Valdecilla sn, 39008, Santander, Spain
| | | | - Jose A Riancho
- Servicio de Medicina Interna, Hospital UM Valdecilla, Universidad de Cantabria, IDIVAL, Avda Valdecilla sn, 39008, Santander, Spain.
| |
Collapse
|
20
|
Amling M. [Research: from the army surgeon to the academic trauma surgeon of the twenty-first century]. UNFALLCHIRURGIE (HEIDELBERG, GERMANY) 2022; 125:767-770. [PMID: 36040513 DOI: 10.1007/s00113-022-01229-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/20/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Michael Amling
- Institut für Osteologie und Biomechanik, Universitätsklinikum Hamburg Eppendorf, Lottestr. 59, 22529, Hamburg, Deutschland.
| |
Collapse
|
21
|
Lin B, Xu P, Zheng J, Deng X, Ye Q, Huang Z, Wang N. Effects and mechanisms of natural alkaloids for prevention and treatment of osteoporosis. Front Pharmacol 2022; 13:1014173. [PMID: 36210805 PMCID: PMC9539536 DOI: 10.3389/fphar.2022.1014173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/31/2022] [Indexed: 11/20/2022] Open
Abstract
Natural alkaloids are polycyclic, nitrogen-containing, and basic compounds obtained from plants. In this review, the advances in bioactive alkaloids with respect to their chemical structures, herbal sources, and effects for the prevention and treatment of osteoporosis are discussed. Anti-osteoporosis alkaloids are classified into six categories based on the chemical structure, namely, isoquinoline alkaloids, quinolizidine alkaloids, piperidine alkaloids, indole alkaloids, pyrrolizidine alkaloids and steroidal alkaloids. They promote mesenchymal stem cells differentiation, improve osteoblast proliferation, stimulate osteoblast autophagy and suppress osteoclast formation. These natural alkaloids can regulate multiple signaling pathways, including interrupting the tumor necrosis factor receptor associated factor 6- receptor activator of nuclear factor kappa B interaction, inhibiting the nuclear factor kappa B pathway in osteoclasts, activating the p38 mitogen-activated protein kinases pathway in osteoblasts, and triggering the wingless and int-1 pathway in mesenchymal stem cells. This review provides evidence and support for novel drug and clinical treatment of osteoporosis using natural alkaloids.
Collapse
Affiliation(s)
- Bingfeng Lin
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Pingcui Xu
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Juan Zheng
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Xuehui Deng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qitao Ye
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhongping Huang
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, China
| | - Nani Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
- *Correspondence: Nani Wang,
| |
Collapse
|
22
|
Costantini A, Mäkitie RE, Hartmann MA, Fratzl-Zelman N, Zillikens MC, Kornak U, Søe K, Mäkitie O. Early-Onset Osteoporosis: Rare Monogenic Forms Elucidate the Complexity of Disease Pathogenesis Beyond Type I Collagen. J Bone Miner Res 2022; 37:1623-1641. [PMID: 35949115 PMCID: PMC9542053 DOI: 10.1002/jbmr.4668] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 12/05/2022]
Abstract
Early-onset osteoporosis (EOOP), characterized by low bone mineral density (BMD) and fractures, affects children, premenopausal women and men aged <50 years. EOOP may be secondary to a chronic illness, long-term medication, nutritional deficiencies, etc. If no such cause is identified, EOOP is regarded primary and may then be related to rare variants in genes playing a pivotal role in bone homeostasis. If the cause remains unknown, EOOP is considered idiopathic. The scope of this review is to guide through clinical and genetic diagnostics of EOOP, summarize the present knowledge on rare monogenic forms of EOOP, and describe how analysis of bone biopsy samples can lead to a better understanding of the disease pathogenesis. The diagnostic pathway of EOOP is often complicated and extensive assessments may be needed to reliably exclude secondary causes. Due to the genetic heterogeneity and overlapping features in the various genetic forms of EOOP and other bone fragility disorders, the genetic diagnosis usually requires the use of next-generation sequencing to investigate several genes simultaneously. Recent discoveries have elucidated the complexity of disease pathogenesis both regarding genetic architecture and bone tissue-level pathology. Two rare monogenic forms of EOOP are due to defects in genes partaking in the canonical WNT pathway: LRP5 and WNT1. Variants in the genes encoding plastin-3 (PLS3) and sphingomyelin synthase 2 (SGMS2) have also been found in children and young adults with skeletal fragility. The molecular mechanisms leading from gene defects to clinical manifestations are often not fully understood. Detailed analysis of patient-derived transiliac bone biopsies gives valuable information to understand disease pathogenesis, distinguishes EOOP from other bone fragility disorders, and guides in patient management, but is not widely available in clinical settings. Despite the great advances in this field, EOOP remains an insufficiently explored entity and further research is needed to optimize diagnostic and therapeutic approaches. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Alice Costantini
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Paris Cité University, INSERM UMR1163, Institut Imagine, Paris, France
| | - Riikka E Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Otorhinolaryngology-Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Markus A Hartmann
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria.,Vienna Bone and Growth Center, Vienna, Austria
| | - Nadja Fratzl-Zelman
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria.,Vienna Bone and Growth Center, Vienna, Austria
| | - M Carola Zillikens
- Bone Center, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Uwe Kornak
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark.,Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Outi Mäkitie
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Folkhälsan Institute of Genetics, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Children's Hospital and Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
23
|
Stürznickel J, Heider F, Delsmann A, Gödel M, Grünhagen J, Huber TB, Kornak U, Amling M, Oheim R. Clinical Spectrum of Hereditary Hypophosphatemic Rickets With Hypercalciuria (HHRH). J Bone Miner Res 2022; 37:1580-1591. [PMID: 35689455 DOI: 10.1002/jbmr.4630] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/19/2022] [Accepted: 06/04/2022] [Indexed: 11/11/2022]
Abstract
Hereditary hypophosphatemic rickets with hypercalciuria (HHRH) represents an FGF23-independent disease caused by biallelic variants in the solute carrier family 34-member 3 (SLC34A3) gene. HHRH is characterized by chronic hypophosphatemia and an increased risk for nephrocalcinosis and rickets/osteomalacia, muscular weakness, and secondary limb deformity. Biochemical changes, but no relevant skeletal changes, have been reported for heterozygous SLC34A3 carriers. Therefore, we assessed the characteristics of individuals with biallelic and monoallelic SLC34A3 variants. In 8 index patients and 5 family members, genetic analysis was performed using a custom gene panel. The skeletal assessment comprised biochemical parameters, areal bone mineral density (aBMD), and bone microarchitecture. Pathogenic SLC34A3 variants were revealed in 7 of 13 individuals (2 homozygous, 5 heterozygous), whereas 3 of 13 carried monoallelic variants of unknown significance. Whereas both homozygous individuals had nephrocalcinosis, only one displayed a skeletal phenotype consistent with HHRH. Reduced to low-normal phosphate levels, decreased tubular reabsorption of phosphate (TRP), and high-normal to elevated values of 1,25-OH2 -D3 accompanied by normal cFGF23 levels were revealed independently of mutational status. Interestingly, individuals with nephrocalcinosis showed significantly increased calcium excretion and 1,25-OH2 -D3 levels but normal phosphate reabsorption. Furthermore, aBMD Z-score <-2.0 was revealed in 4 of 8 heterozygous carriers, and HR-pQCT analysis showed a moderate decrease in structural parameters. Our findings highlight the clinical relevance also of monoallelic SLC34A3 variants, including their potential skeletal impairment. Calcium excretion and 1,25-OH2 -D3 levels, but not TRP, were associated with nephrocalcinosis. Future studies should investigate the effects of distinct SLC34A3 variants and optimize treatment and monitoring regimens to prevent nephrocalcinosis and skeletal deterioration. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fiona Heider
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alena Delsmann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Gödel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Grünhagen
- Labor Berlin Charité Vivantes GmbH-corporate member of Institute for Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uwe Kornak
- Institute for Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 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.,Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
24
|
Oheim R, Tsourdi E, Seefried L, Beller G, Schubach M, Vettorazzi E, Stürznickel J, Rolvien T, Ehmke N, Delsmann A, Genest F, Krüger U, Zemojtel T, Barvencik F, Schinke T, Jakob F, Hofbauer LC, Mundlos S, Kornak U. Genetic Diagnostics in Routine Osteological Assessment of Adult Low Bone Mass Disorders. J Clin Endocrinol Metab 2022; 107:e3048-e3057. [PMID: 35276006 PMCID: PMC9202726 DOI: 10.1210/clinem/dgac147] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Indexed: 12/17/2022]
Abstract
CONTEXT Many different inherited and acquired conditions can result in premature bone fragility/low bone mass disorders (LBMDs). OBJECTIVE We aimed to elucidate the impact of genetic testing on differential diagnosis of adult LBMDs and at defining clinical criteria for predicting monogenic forms. METHODS Four clinical centers broadly recruited a cohort of 394 unrelated adult women before menopause and men younger than 55 years with a bone mineral density (BMD) Z-score < -2.0 and/or pathological fractures. After exclusion of secondary causes or unequivocal clinical/biochemical hallmarks of monogenic LBMDs, all participants were genotyped by targeted next-generation sequencing. RESULTS In total, 20.8% of the participants carried rare disease-causing variants (DCVs) in genes known to cause osteogenesis imperfecta (COL1A1, COL1A2), hypophosphatasia (ALPL), and early-onset osteoporosis (LRP5, PLS3, and WNT1). In addition, we identified rare DCVs in ENPP1, LMNA, NOTCH2, and ZNF469. Three individuals had autosomal recessive, 75 autosomal dominant, and 4 X-linked disorders. A total of 9.7% of the participants harbored variants of unknown significance. A regression analysis revealed that the likelihood of detecting a DCV correlated with a positive family history of osteoporosis, peripheral fractures (> 2), and a high normal body mass index (BMI). In contrast, mutation frequencies did not correlate with age, prevalent vertebral fractures, BMD, or biochemical parameters. In individuals without monogenic disease-causing rare variants, common variants predisposing for low BMD (eg, in LRP5) were overrepresented. CONCLUSION The overlapping spectra of monogenic adult LBMD can be easily disentangled by genetic testing and the proposed clinical criteria can help to maximize the diagnostic yield.
Collapse
Affiliation(s)
- Ralf Oheim
- Ralf Oheim, MD, Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestraße 59, 22529 Hamburg, Germany.
| | - Elena Tsourdi
- Department of Medicine III, Technische Universität Dresden Medical Center, 01307 Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden Medical Center, 01307 Dresden, Germany
| | - Lothar Seefried
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, 97070 Würzburg, Germany
| | - Gisela Beller
- Centre of Muscle and Bone Research, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Max Schubach
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Eik Vettorazzi
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
- Department of Orthopaedics and Trauma Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
- Department of Orthopaedics and Trauma Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nadja Ehmke
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Alena Delsmann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Franca Genest
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, 97070 Würzburg, Germany
| | - Ulrike Krüger
- Core Facility Genomics, Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Tomasz Zemojtel
- Core Facility Genomics, Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Florian Barvencik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, 97070 Würzburg, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden Medical Center, 01307 Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden Medical Center, 01307 Dresden, Germany
| | - Stefan Mundlos
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10178 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Uwe Kornak
- Correspondence: Uwe Kornak, PhD, Institute of Human Genetics, Universitätsmedizin Göttingen, Heinrich-Düker-Weg 12, 37073 Göttingen, Germany.
| |
Collapse
|
25
|
Caetano da Silva C, Edouard T, Fradin M, Aubert-Mucca M, Ricquebourg M, Raman R, Salles JP, Charon V, Guggenbuhl P, Muller M, Cohen-Solal M, Collet C. WNT11, a new gene associated with early onset osteoporosis, is required for osteoblastogenesis. Hum Mol Genet 2022; 31:1622-1634. [PMID: 34875064 PMCID: PMC9122655 DOI: 10.1093/hmg/ddab349] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 11/12/2022] Open
Abstract
Monogenic early onset osteoporosis (EOOP) is a rare disease defined by low bone mineral density (BMD) that results in increased risk of fracture in children and young adults. Although several causative genes have been identified, some of the EOOP causation remains unresolved. Whole-exome sequencing revealed a de novo heterozygous loss-of-function mutation in Wnt family member 11 (WNT11) (NM_004626.2:c.677_678dup p.Leu227Glyfs*22) in a 4-year-old boy with low BMD and fractures. We identified two heterozygous WNT11 missense variants (NM_004626.2:c.217G > A p.Ala73Thr) and (NM_004626.2:c.865G > A p.Val289Met) in a 51-year-old woman and in a 61-year-old woman, respectively, both with bone fragility. U2OS cells with heterozygous WNT11 mutation (NM_004626.2:c.690_721delfs*40) generated by CRISPR-Cas9 showed reduced cell proliferation (30%) and osteoblast differentiation (80%) as compared with wild-type U2OS cells. The expression of genes in the Wnt canonical and non-canonical pathways was inhibited in these mutant cells, but recombinant WNT11 treatment rescued the expression of Wnt pathway target genes. Furthermore, the expression of RSPO2, a WNT11 target involved in bone cell differentiation, and its receptor leucine-rich repeat containing G protein-coupled receptor 5 (LGR5), was decreased in WNT11 mutant cells. Treatment with WNT5A and WNT11 recombinant proteins reversed LGR5 expression, but Wnt family member 3A (WNT3A) recombinant protein treatment had no effect on LGR5 expression in mutant cells. Moreover, treatment with recombinant RSPO2 but not WNT11 or WNT3A activated the canonical pathway in mutant cells. In conclusion, we have identified WNT11 as a new gene responsible for EOOP, with loss-of-function variant inhibiting bone formation via Wnt canonical and non-canonical pathways. WNT11 may activate Wnt signaling by inducing the RSPO2-LGR5 complex via the non-canonical Wnt pathway.
Collapse
Affiliation(s)
- Caroline Caetano da Silva
- INSERM U1132 and Université de Paris, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris F-75010, France
| | - Thomas Edouard
- Endocrine Bone Diseases and Genetics Unit, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, Pediatric Clinical Research Unit, Children’s Hospital, RESTORE INSERM U1301, Toulouse University Hospital, Toulouse 31300, France
| | - Melanie Fradin
- Service de Génétique Clinique, Centre de Référence des Anomalies du Développement de l'Ouest, Hôpital Sud de Rennes, Rennes F-35033, France
| | - Marion Aubert-Mucca
- Endocrine Bone Diseases and Genetics Unit, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, Pediatric Clinical Research Unit, Children’s Hospital, RESTORE INSERM U1301, Toulouse University Hospital, Toulouse 31300, France
| | - Manon Ricquebourg
- INSERM U1132 and Université de Paris, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris F-75010, France
| | - Ratish Raman
- Laboratory for Organogenesis and Regeneration (LOR), GIGA-Research, Liège University, Liège 4000, Belgium
| | - Jean Pierre Salles
- Endocrine Bone Diseases and Genetics Unit, Reference Centre for Rare Diseases of Calcium and Phosphate Metabolism, ERN BOND, OSCAR Network, Pediatric Clinical Research Unit, Children’s Hospital, RESTORE INSERM U1301, Toulouse University Hospital, Toulouse 31300, France
| | - Valérie Charon
- Department of Radiology, CHU de Rennes, Rennes F-35000, France
| | | | - Marc Muller
- Laboratory for Organogenesis and Regeneration (LOR), GIGA-Research, Liège University, Liège 4000, Belgium
| | - Martine Cohen-Solal
- INSERM U1132 and Université de Paris, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris F-75010, France
| | - Corinne Collet
- INSERM U1132 and Université de Paris, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris F-75010, France
- Département de Génétique, UF de Génétique Moléculaire, Hôpital Robert Debré, APHP, Paris F-75019, France
| |
Collapse
|
26
|
Abstract
Osteoporosis is a skeletal disorder with enhanced bone fragility, usually affecting the elderly. It is very rare in children and young adults and the definition is not only based on a low BMD (a Z-score < - 2.0 in growing children and a Z-score ≤ - 2.0 or a T-score ≤ - 2.5 in young adults) but also on the occurrence of fragility fractures and/or the existence of underlying chronic diseases or secondary factors such as use of glucocorticoids. In the absence of a known chronic disease, fragility fractures and low BMD should prompt extensive screening for secondary causes, which can be found in up to 90% of cases. When fragility fractures occur in childhood or young adulthood without an evident secondary cause, investigations should explore the possibility of an underlying monogenetic bone disease, where bone fragility is caused by a single variant in a gene that has a major role in the skeleton. Several monogenic forms relate to type I collagen, but other forms also exist. Loss-of-function variants in LRP5 and WNT1 may lead to early-onset osteoporosis. The X-chromosomal osteoporosis caused by PLS3 gene mutations affects especially males. Another recently discovered form relates to disturbed sphingolipid metabolism due to SGMS2 mutations, underscoring the complexity of molecular pathology in monogenic early-onset osteoporosis. Management of young patients consists of treatment of secondary factors, optimizing lifestyle factors including calcium and vitamin D and physical exercise. Treatment with bone-active medication should be discussed on a personalized basis, considering the severity of osteoporosis and underlying disease versus the absence of evidence on anti-fracture efficacy and potential harmful effects in pregnancy.
Collapse
Affiliation(s)
- Outi Mäkitie
- Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Folkhälsan Research Center, Biomedicum Helsinki, P.O. Box 63, FI-00014, Helsinki, Finland.
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus University Medical Center, 3015, Rotterdam, The Netherlands
| |
Collapse
|
27
|
Yu S, Li D, Zhang N, Ni S, Sun M, Wang L, Xiao H, Liu D, Liu J, Yu Y, Zhang Z, Yeung STY, Zhang S, Lu A, Zhang Z, Zhang B, Zhang G. Drug discovery of sclerostin inhibitors. Acta Pharm Sin B 2022; 12:2150-2170. [PMID: 35646527 PMCID: PMC9136615 DOI: 10.1016/j.apsb.2022.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/03/2021] [Accepted: 12/16/2021] [Indexed: 12/18/2022] Open
Abstract
Sclerostin, a protein secreted from osteocytes, negatively regulates the WNT signaling pathway by binding to the LRP5/6 co-receptors and further inhibits bone formation and promotes bone resorption. Sclerostin contributes to musculoskeletal system-related diseases, making it a promising therapeutic target for the treatment of WNT-related bone diseases. Additionally, emerging evidence indicates that sclerostin contributes to the development of cancers, obesity, and diabetes, suggesting that it may be a promising therapeutic target for these diseases. Notably, cardiovascular diseases are related to the protective role of sclerostin. In this review, we summarize three distinct types of inhibitors targeting sclerostin, monoclonal antibodies, aptamers, and small-molecule inhibitors, from which monoclonal antibodies have been developed. As the first-in-class sclerostin inhibitor approved by the U.S. FDA, the monoclonal antibody romosozumab has demonstrated excellent effectiveness in the treatment of postmenopausal osteoporosis; however, it conferred high cardiovascular risk in clinical trials. Furthermore, romosozumab could only be administered by injection, which may cause compliance issues for patients who prefer oral therapy. Considering these above safety and compliance concerns, we therefore present relevant discussion and offer perspectives on the development of next-generation sclerostin inhibitors by following several ways, such as concomitant medication, artificial intelligence-based strategy, druggable modification, and bispecific inhibitors strategy.
Collapse
|
28
|
Rahlf AL, Hoenig T, Stürznickel J, Cremans K, Fohrmann D, Sanchez-Alvarado A, Rolvien T, Hollander K. A machine learning approach to identify risk factors for running-related injuries: study protocol for a prospective longitudinal cohort trial. BMC Sports Sci Med Rehabil 2022; 14:75. [PMID: 35473813 PMCID: PMC9040327 DOI: 10.1186/s13102-022-00426-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/23/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Running is a very popular sport among both recreational and competitive athletes. However, participating in running is associated with a comparably high risk of sustaining an exercise-related injury. Due to the often multifactorial and individual reasons for running injuries, a shift in thinking is required to account for the dynamic process of the various risk factors. Therefore, a machine learning approach will be used to comprehensively analyze biomechanical, biological, and loading parameters in order to identify risk factors and to detect risk patterns in runners. METHODS The prospective longitudinal cohort study will include competitive adult athletes, running at least 20 km per week and being free of injuries three months before the start of the study. At baseline and the end of the study period, subjective questionnaires (demographics, injury history, sports participation, menstruation, medication, psychology), biomechanical measures (e.g., stride length, cadence, kinematics, kinetics, tibial shock, and tibial acceleration) and a medical examination (BMI, laboratory: blood count, creatinine, calcium, phosphate, parathyroid hormone, vitamin D, osteocalcin, bone-specific alkaline phosphatase, DPD cross-links) will be performed. During the study period (one season), continuous data collection will be performed for biomechanical parameters, injuries, internal and external load. Statistical analysis of the data is performed using machine learning (ML) methods. For this purpose, the correlation of the collected data to possible injuries is automatically learned by an ML model and from this, a ranking of the risk factors can be determined with the help of sensitivity analysis methods. DISCUSSION To achieve a comprehensive risk reduction of injuries in runners, a multifactorial and individual approach and analysis is necessary. Recently, the use of ML processes for the analysis of risk factors in sports was discussed and positive results have been published. This study will be the first prospective longitudinal cohort study in runners to investigate the association of biomechanical, bone health, and loading parameters as well as injuries via ML models. The results may help to predict the risk of sustaining an injury and give way for new analysis methods that may also be transferred to other sports. TRIAL REGISTRATION DRKS00026904 (German Clinical Trial Register DKRS), date of registration 18.10.2021.
Collapse
Affiliation(s)
- A L Rahlf
- Department of Sports Science, Institute of Health, Nutrition and Sports Science, Europa-Universität Flensburg, Campusallee 2, 24943, Flensburg, Germany.
| | - T Hoenig
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - J Stürznickel
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.,Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - K Cremans
- Department of Mechanical Engineering, Institute of Modelling and High-Performance Computing, Niederrhein University of Applied Sciences, Reinarzstraße 49, 47805, Krefeld, Germany
| | - D Fohrmann
- Institute of Interdisciplinary Exercise Science and Sports Medicine, MSH Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany
| | - A Sanchez-Alvarado
- Department of Sports and Exercise Medicine, Institute of Human Movement Science, University of Hamburg, Turmweg 2, 20148, Hamburg, Germany
| | - T Rolvien
- Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - K Hollander
- Institute of Interdisciplinary Exercise Science and Sports Medicine, MSH Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany
| |
Collapse
|
29
|
Homaei A, Chegini V, Saffari F. Clinical Response to Treatment with Teriparatide in an Adolescent with Osteoporosis-Pseudoglioma Syndrome (OPPG): A Case Report. Int J Endocrinol Metab 2022; 20:e121031. [PMID: 35993038 PMCID: PMC9383542 DOI: 10.5812/ijem-121031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/27/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Osteoporosis-pseudoglioma syndrome (OPPG) is a rare autosomal recessive disorder characterized by severe osteoporosis and eye abnormalities that leads to vision loss. In this study, we report the outcome of a short period of treatment with teriparatide in one patient with OPPG. CASE PRESENTATION The patient was a 17-year-old girl who suffered a bone fracture at the age of two and was diagnosed with OPPG at the age of three. Genetic testing was performed for the patient, and a novel homozygous nonsense mutation (c.351G>A) in exon 2 of the LRP5 gene was reported. She was treated with pamidronate, but the bone fracture increased, and the disability progressed. Therefore, at the age of 11 years and nine months, teriparatide was administered subcutaneously at a dose of 20 micrograms per day for four consecutive months. After the treatment with teriparatide, physical activity was achieved, and no further fractures were observed besides the gradual rise in bone mineral density (BMD) (from 0.532 to 0.711 gr/cm2 in lumbar spine and 0.372 to 0.635 gr/cm2 in femur neck). CONCLUSIONS In children and adolescents diagnosed with OPPG who do not respond to other conventional therapies, short courses of teriparatide therapy may be helpful.
Collapse
Affiliation(s)
- Ali Homaei
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Victoria Chegini
- Department of Pediatrics, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Saffari
- Children Growth Research Center, Research Institute for Prevention of Non-communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
- Clinical Research Development Unit, Qods Hospital, Qazvin University of Medical Sciences, Qazvin, Iran
- Corresponding Author: Children Growth Research Center, Research Institute for Prevention of Non-communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
30
|
Zhao M, Wu D, Yu K, Shen M. Clinical and Genetic Features of Chinese Adult Patients With Chronic Non-Bacterial Osteomyelitis: A Single Center Report. Front Immunol 2022; 13:860646. [PMID: 35422809 PMCID: PMC9002012 DOI: 10.3389/fimmu.2022.860646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
Objectives Chronic non-bacterial osteomyelitis (CNO) is a rare polygenic autoinflammatory bone disease. We aimed to characterize the clinical manifestations and gene variants of Chinese adult patients with CNO. Methods By reviewing data of all CNO patients being diagnosed and followed up at the Center for Adult Autoinflammation Diseases, Department of Rheumatology, Peking Union Medical College Hospital, clinical and genetic features of these patients were evaluated and concluded. Results The median age of disease onset was 19 (6-64) years old, and adult-onset was observed in 6 (60%) patients. The mean time of diagnosis delay was 92 ± 78 months. The common symptoms were bone pain (10, 100%), fever (9, 90%), and arthritis (6, 60%). In total, there were 54 skeletal lesions, and each patient had no less than 2 lesions. The most frequently affected sites included lower limbs (20.5%), mandible, vertebrae and pelvis (17.5%, separately). Variants of 4 genes were detected in our study including COL1A1, PSTPIP1, LRP5 and CLCN7. In seven patients who were treated with combination therapy containing tumor necrosis factor (TNF) α inhibitors, five (55.6%) had a complete response and 2 (44.4%) had a partial response. Conclusion This is the first and largest case series of CNO in the Chinese adult patients. Four novel genetic mutations potentially associated with CNO were identified. Notably, CNO should be considered in the differential diagnosis of adult patients with long disease course and recurrent multifocal osteomyelitis of unknown cause, and these patients might benefit from combination therapy containing TNFα inhibitors.
Collapse
Affiliation(s)
- Mengzhu Zhao
- Department of Rheumatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Di Wu
- Department of Rheumatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Keyi Yu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Min Shen
- Department of Rheumatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
31
|
Song S, Guo Y, Yang Y, Fu D. Advances in pathogenesis and therapeutic strategies for osteoporosis. Pharmacol Ther 2022; 237:108168. [PMID: 35283172 DOI: 10.1016/j.pharmthera.2022.108168] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 02/07/2023]
Abstract
Osteoporosis, is the most common bone disorder worldwide characterized by low bone mineral density, leaving affected bones vulnerable to fracture. Bone homeostasis depends on the precise balance between bone resorption by osteoclasts and bone matrix formation by mesenchymal lineage osteoblasts, and involves a series of complex and highly regulated steps. Bone homeostasis will be disrupted when the speed of bone resorption is faster than bone formation. Based on various regulatory mechanisms of bone homeostasis, a series of drugs targeting osteoporosis have emerged in clinical practice, including bisphosphonates, selective estrogen receptor modulators, calcitonin, molecular-targeted drugs and so on. However, many drugs have major adverse effects or are unsuitable for long-term use. Therefore, it is very urgent to find more effective therapeutic drugs based on the new pathogenesis of osteoporosis. In this review, we summarize novel mechanisms involved in the pathological process of osteoporosis, including the roles of gut microbiome, autophagy, iron balance and cellular senescence. Based on the above pathological mechanism, we found promising drugs for osteoporosis treatment, such as: probiotics, alpha-ketoglutarate, senolytics and hydrogen sulfide. This new finding may provide an important basis for elucidating the complex pathological mechanisms of osteoporosis and provide promising drugs for clinical osteoporosis treatment.
Collapse
Affiliation(s)
- Shasha Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, PR China
| | - Yuanyuan Guo
- Department of Pharmacy, Liyuan Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei 430077, PR China
| | - Yuehua Yang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Dehao Fu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China.
| |
Collapse
|
32
|
Rouleau C, Malorie M, Collet C, Porquet-Bordes V, Gennero I, Eddiry S, Laroche M, Salles JP, Couture G, Edouard T. Diagnostic yield of bone fragility gene panel sequencing in children and young adults referred for idiopathic primary osteoporosis at a single regional reference centre. Bone Rep 2022; 16:101176. [PMID: 35252483 PMCID: PMC8892094 DOI: 10.1016/j.bonr.2022.101176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/22/2022] Open
Abstract
Aim To describe the presenting features, bone characteristics and molecular genetics in a large monocentric cohort of children and young adults with idiopathic primary osteoporosis. Methods Sixty-six patients (19 children, 47 adults; 28 males, 38 females; age at referral: 3.8 to 65 years) diagnosed with primary osteoporosis were included in this study; patients with features of osteogenesis imperfecta or other known syndromes associated with osteoporosis were excluded. For each patient, the following data were collected by retrospective chart review: family and personal history of fracture and osteoporosis, mineral homeostasis parameters and markers of bone formation and resorption, bone mineral density (BMD) of the lumbar spine (LS-BMD), the total body less head (TB-BMD), and total hip levels (TH-BMD) measured by DXA. As part of the initial assessment process, a bone fragility gene panel sequencing was performed in all of these patients. Results There was a higher predominance of males in the children (63%) and of females in the adults (66%) (p = 0.030). Compared to the adults, the children had a significantly lower frequency of vertebral fractures (26 vs 57%, p = 0.022) and a higher frequency of peripheral fractures (84 vs 53%; p = 0.019). Bone fragility gene panel sequencing allowed the identification of the heterozygous pathogenic variant in 27% of patients (most frequently in LRP5, WNT1 and COL1A1 or 2 genes) and the heterozygous p.(Val667Met) LRP5 variant in 11% of them. The frequency of pathogenic variants tended to be higher in the children compared to the adults without reaching statistical significance (42 vs 19%; p = 0.053). The frequency of the p.(Val667Met) LRP5 variant was similar in children and adults. No significant differences were found regarding the various clinical, biological and radiological characteristics of the patients according to genotype. Conclusion In this study, we reported the presenting features and bone characteristics in a large cohort of children and young adults with idiopathic primary osteoporosis. Bone fragility gene panel sequencing allowed the identification of genetic variants in a significant proportion of these patients. Molecular diagnosis in these patients is important in order to be able to offer genetic counselling and organise patient management.
Collapse
|
33
|
Herath M, Cohen A, Ebeling PR, Milat F. Dilemmas in the Management of Osteoporosis in Younger Adults. JBMR Plus 2022; 6:e10594. [PMID: 35079682 PMCID: PMC8771004 DOI: 10.1002/jbm4.10594] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 12/02/2021] [Accepted: 12/12/2021] [Indexed: 11/16/2022] Open
Abstract
Osteoporosis in premenopausal women and men younger than 50 years is challenging to diagnose and treat. There are many barriers to optimal management of osteoporosis in younger adults, further enhanced by a limited research focus on this cohort. Herein we describe dilemmas commonly encountered in diagnosis, investigation, and management of osteoporosis in younger adults. We also provide a suggested framework, based on the limited available evidence and supported by clinical experience, for the diagnosis, assessment, and management of osteoporosis in this cohort. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Madhuni Herath
- Department of Endocrinology Monash Health Clayton Victoria Australia
- Centre for Endocrinology & Metabolism Hudson Institute of Medical Research Clayton Victoria Australia
- Department of Medicine, School of Clinical Sciences Monash University Clayton Victoria Australia
| | - Adi Cohen
- Department of Medicine Columbia University College of Physicians & Surgeons New York NY USA
| | - Peter R. Ebeling
- Department of Endocrinology Monash Health Clayton Victoria Australia
- Department of Medicine, School of Clinical Sciences Monash University Clayton Victoria Australia
| | - Frances Milat
- Department of Endocrinology Monash Health Clayton Victoria Australia
- Centre for Endocrinology & Metabolism Hudson Institute of Medical Research Clayton Victoria Australia
- Department of Medicine, School of Clinical Sciences Monash University Clayton Victoria Australia
| |
Collapse
|
34
|
Dabas A, Malhotra R, Kumar R, Khadgawat R. Idiopathic juvenile osteoporosis in a child: a four-year follow-up with review of literature. J Pediatr Endocrinol Metab 2021; 34:1487-1490. [PMID: 34331423 DOI: 10.1515/jpem-2021-0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 07/09/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Childhood osteoporosis is an uncommon condition that usually develops secondary to underlying disease states. Idiopathic juvenile osteoporosis or early onset osteoporosis is a rare cause of primary osteoporosis in childhood associated with mutations in "bone fragility" genes. CASE PRESENTATION The index case presented with upper back pain and was detected to have multiple vertebral fractures. Further workup for the cause revealed a homozygous benign mutation in low-density lipoprotein receptor-related protein 5, which was also detected in the mother who remained asymptomatic till presentation. The child was successfully treated with intravenous zoledronate. CONCLUSIONS The case report describes the management approach and four-year follow-up of the child.
Collapse
Affiliation(s)
- Aashima Dabas
- Department of Pediatrics, Maulana Azad Medical College and Lok Nayak Hospital, New Delhi, India
| | - Rakhi Malhotra
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Ravindra Kumar
- Department of Pediatrics, Hindu Rao Hospital, New Delhi, India
| | - Rajesh Khadgawat
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
35
|
Mandair GS, Akhter MP, Esmonde-White FWL, Lappe JM, Bare SP, Lloyd WR, Long JP, Lopez J, Kozloff KM, Recker RR, Morris MD. Altered collagen chemical compositional structure in osteopenic women with past fractures: A case-control Raman spectroscopic study. Bone 2021; 148:115962. [PMID: 33862262 PMCID: PMC8259347 DOI: 10.1016/j.bone.2021.115962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/25/2021] [Accepted: 04/08/2021] [Indexed: 12/17/2022]
Abstract
Incidences of low-trauma fractures among osteopenic women may be related to changes in bone quality. In this blinded, prospective-controlled study, compositional and heterogeneity contributors of bone quality to fracture risk were examined. We hypothesize that Raman spectroscopy can differentiate between osteopenic women with one or more fractures (cases) from women without fractures (controls). This study involved the Raman spectroscopic analysis of cortical and cancellous bone composition using iliac crest biopsies obtained from 59-cases and 59-controls, matched for age (62.0 ± 7.5 and 61.7 ± 7.3 years, respectively, p = 0.38) and hip bone mineral density (BMD, 0.827 ± 0.083 and 0.823 ± 0.072 g/cm3, respectively, p = 0.57). Based on aggregate univariate case-control and odds ratio based logistic regression analyses, we discovered two Raman ratiometric parameters that were predictive of past fracture risk. Specifically, 1244/1268 and 1044/959 cm-1 ratios, were identified as the most differential aspects of bone quality in cortical cases with odds ratios of 0.617 (0.406-0.938 95% CI, p = 0.024) and 1.656 (1.083-2.534 95% CI, p = 0.020), respectively. Both 1244/1268 and 1044/959 cm-1 ratios exhibited moderate sensitivity (59.3-64.4%) but low specificity (49.2-52.5%). These results suggest that the organization of mineralized collagen fibrils were significantly altered in cortical cases compared to controls. In contrast, compositional and heterogeneity parameters related to mineral/matrix ratios, B-type carbonate substitutions, and mineral crystallinity, were not significantly different between cases and controls. In conclusion, a key outcome of this study is the significant odds ratios obtained for two Raman parameters (1244/1268 and 1044/959 cm-1 ratios), which from a diagnostic perspective, may assist in the screening of osteopenic women with suspected low-trauma fractures. One important implication of these findings includes considering the possibility that changes in the organization of collagen compositional structure plays a far greater role in postmenopausal women with osteopenic fractures.
Collapse
Affiliation(s)
- Gurjit S Mandair
- School of Dentistry, Departments of Biologic and Materials, University of Michigan, Ann Arbor, MI, USA.
| | | | | | - Joan M Lappe
- Osteoporosis Research Center, Creighton University, Omaha, NE, USA
| | - Susan P Bare
- Osteoporosis Research Center, Creighton University, Omaha, NE, USA
| | - William R Lloyd
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Jason P Long
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jessica Lopez
- School of Dentistry, Departments of Biologic and Materials, University of Michigan, Ann Arbor, MI, USA
| | - Kenneth M Kozloff
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Robert R Recker
- Osteoporosis Research Center, Creighton University, Omaha, NE, USA
| | - Michael D Morris
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
36
|
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
|
37
|
Butscheidt S, Tsourdi E, Rolvien T, Delsmann A, Stürznickel J, Barvencik F, Jakob F, Hofbauer LC, Mundlos S, Kornak U, Seefried L, Oheim R. Relevant genetic variants are common in women with pregnancy and lactation-associated osteoporosis (PLO) and predispose to more severe clinical manifestations. Bone 2021; 147:115911. [PMID: 33716164 DOI: 10.1016/j.bone.2021.115911] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/19/2021] [Accepted: 03/08/2021] [Indexed: 12/17/2022]
Abstract
Pregnancy and lactation-associated osteoporosis (PLO) is a rare skeletal disorder characterized by early-onset osteoporosis typically manifestating with vertebral compression fractures or transient osteoporosis of the hip. We hypothesized that genetic variants may play a role in the development of PLO. This study aimed to analyze the presence of genetic variants and a potential association with the clinical presentation in PLO. 42 women with PLO were included from 2013 to 2019 in a multicenter study in Germany. All cases underwent comprehensive genetic analysis based on a custom-designed gene panel including genes relevant for skeletal disorders. The skeletal status was assessed using dual-energy X-ray absorptiometry (DXA). Subgroups were further analyzed by serum bone turnover markers (n = 31) and high-resolution peripheral computed tomography (HR-pQCT; n = 23). We detected relevant genetic variants in 21 women (50%), with LRP5, WNT1 and COL1A1/A2 being the most commonly involved genes. The mean number of vertebral compression fractures was 3.3 ± 3.4 per case with a significantly higher occurrence in the subgroup with genetic variants (4.8 ± 3.7 vs. 1.8 ± 2.3, p = 0.02). Among the total cohort, DXA Z-scores were significantly lower at the lumbar spine compared to the femoral neck (p = 0.002). HR-pQCT revealed a pronounced reduction of trabecular and cortical thickness, while trabecular number was within the reference range. Eighteen women (43%) received a bone-specific therapy (primarily teriparatide). Overall, a steep increase in bone mass (+37.7%) was observed after 3 years. In conclusion, pregnancy and lactation represent skeletal risk factors, which may unmask hereditary bone disorders leading to PLO. These cases were affected more severely. Nevertheless, a timely diagnosis and adequate treatment can ensure a substantial recovery potential even without specific therapy. Patients with genetically induced low bone turnover (e.g.; LRP5, WNT1) may especially benefit from osteo-anabolic medication.
Collapse
Affiliation(s)
- Sebastian Butscheidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Division of Orthopaedics, Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Tsourdi
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany; Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Division of Orthopaedics, Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alena Delsmann
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Barvencik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany; Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| | - Stefan Mundlos
- Institute of Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany; Institute of Human Genetics, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Lothar Seefried
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
38
|
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
|