1
|
Li JX, Dang YM, Liu MC, Gao LQ, Lin H. Fibroblasts in heterotopic ossification: mechanisms and therapeutic targets. Int J Biol Sci 2025; 21:544-564. [PMID: 39781450 PMCID: PMC11705629 DOI: 10.7150/ijbs.102297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/07/2024] [Indexed: 01/12/2025] Open
Abstract
Heterotopic ossification (HO) refers to the abnormal formation of bone in non-skeletal tissues. Fibroblasts have traditionally been viewed as stationary cells primarily responsible for producing extracellular matrix during tissue repair and fibrosis. However, recent discoveries regarding their plasticity-encompassing roles in inflammation, extracellular matrix remodeling, and osteogenesis-highlight their potential as key contributors to the development of HO. In this review, we systematically summarize the diverse phenotypic and functional plasticity of fibroblasts in HO. Furthermore, we evaluate the possible interaction between fibroblasts and macrophages in pathophysiological processes and signaling pathways. Finally, we highlight the potential strategies for preventing and treating HO by targeting fibroblast activities.
Collapse
Affiliation(s)
- Jia-xin Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- First Clinical School, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yan-miao Dang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Meng-chao Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lin-qing Gao
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Hui Lin
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| |
Collapse
|
2
|
Iorio R, Cantagalli MR, Viglietta E, Corsetti F, Gugliotta Y, Previ L, Gagliardo S, Fenucci S, Maffulli N. "Heterotopic ossification after total hip arthroplasty through direct anterior approach without a dedicated orthopaedic table or direct lateral approach: a quasi-randomized single-center study". Arch Orthop Trauma Surg 2024; 144:4225-4232. [PMID: 39311937 PMCID: PMC11564372 DOI: 10.1007/s00402-024-05510-3] [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: 04/04/2024] [Accepted: 08/14/2024] [Indexed: 11/15/2024]
Abstract
INTRODUCTION Heterotopic ossifications (HO) are common after total hip arthroplasty (THA). The invasiveness of surgical approaches plays a relevant role in HO development. The aims of this study were to assess the development of HO 6 months after THA through direct lateral approach (DLA) or direct anterior approach (DAA) without a dedicated orthopaedic table and to assess the clinical impact of HO. METHODS This is a single-center IRB-approved, quasi-randomized prospective cohort, observational imaging study. Fifty patients underwent primary THA through DLA and 50 through DAA. Age, sex, BMI and side of the affected hip were collected. At the 6 post-operative month the Harris Hip Score (HHS) and the presence of HO (scored through the Brooker classification system) were assessed. RESULTS There was no significant difference in the demographic data between groups. Operative time was significantly higher in the DAA group (72 ± 10 min vs. 58 ± 8 min: p < 0.03). At 6 post-operative months the incidence of HO was 14% in the DAA group and 32% in the DLA group (p = 0.02). Severe HO (Brooker 3-4) were significantly more common in the DLA group (p = 0.04). There was no significant difference in the HHS of patients with HO between the DAA and DLA groups. There was no association between poorer clinical outcomes and the severity of HO. CONCLUSION The DAA without a dedicated orthopaedic table is associated with a significant lower incidence of HO than the DLA 6 months after elective THA. Except for the surgical approach, no other factors correlated with the occurrence of HO. Even though a lower HHS was found with severe HO, the correlation between severity of HO and clinical outcomes did not reach statistical significance.
Collapse
Affiliation(s)
- Raffaele Iorio
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy
| | - Matteo Romano Cantagalli
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy
| | - Edoardo Viglietta
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy.
| | - Federico Corsetti
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy
| | - Yuri Gugliotta
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy
| | - Leonardo Previ
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy
| | - Salvatore Gagliardo
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy
| | - Simone Fenucci
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy
| | - Nicola Maffulli
- Orthopaedic Unit, S. Andrea Hospital, University of Rome "La Sapienza", Via di Grottarossa 1035, Rome, Italy
| |
Collapse
|
3
|
Towler OW, Shore EM, Kaplan FS. Molecular Developmental Biology of Fibrodysplasia Ossificans Progressiva: Measuring the Giant by Its Toe. Biomolecules 2024; 14:1009. [PMID: 39199396 PMCID: PMC11353020 DOI: 10.3390/biom14081009] [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: 06/25/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
When a genetic disease is characterized by the abnormal activation of normal molecular pathways and cellular events, it is illuminating to critically examine the places and times of these activities both in health and disease. Therefore, because heterotopic ossification (HO) in fibrodysplasia ossificans progressiva (FOP) is by far the disease's most prominent symptom, attention is also directed toward the pathways and processes of bone formation during skeletal development. FOP is recognizable by effects of the causative mutation on skeletal development even before HO manifests, specifically in the malformation of the great toes. This signature skeletal phenotype is the most highly penetrant, but is only one among several skeletal abnormalities associated with FOP. Patients may present clinically with joint malformation and ankylosis, particularly in the cervical spine and costovertebral joints, as well as characteristic facial features and a litany of less common, non-skeletal symptoms, all stemming from missense mutations in the ACVR1 gene. In the same way that studying the genetic cause of HO advanced our understanding of HO initiation and progression, insight into the roles of ACVR1 signaling during tissue development, particularly in the musculoskeletal system, can be gained from examining altered skeletal development in individuals with FOP. This review will detail what is known about the molecular mechanisms of developmental phenotypes in FOP and the early role of ACVR1 in skeletal patterning and growth, as well as highlight how better understanding these processes may serve to advance patient care, assessments of patient outcomes, and the fields of bone and joint biology.
Collapse
Affiliation(s)
- O. Will Towler
- Division of Plastic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Eileen M. Shore
- Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frederick S. Kaplan
- Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
4
|
Zhang C, Xiao D, Shu L, Gong M, Liu X, Jiang X. Single-cell RNA sequencing uncovers cellular heterogeneity and the progression of heterotopic ossification of the elbow. Front Pharmacol 2024; 15:1434146. [PMID: 39045052 PMCID: PMC11264200 DOI: 10.3389/fphar.2024.1434146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 07/25/2024] Open
Abstract
Heterotopic ossification of the elbow (HOE) is a complicated pathologic process characterized by extra bone formation in the elbow. Bone formation is a complex developmental process involving the differentiation of mesenchymal stem cells into osteoblasts. The aim of this study was to explore the cellular origin and progression of HOE by single-cell RNA sequencing. We identified 13 clusters of cells in HOE and further analyzed the subclusters for 4 of the main cell types. Six subclusters of osteoblasts, nine subclusters of chondrocytes, six subclusters of fibroblasts, and five subclusters of mononuclear phagocytes (MPs) were identified and analyzed. The new findings on osterix (OSX) and SOX9 expression in osteoblast subclusters and chondrocyte subclusters indicate that HOE is mediated through endochondral ossification. Further identification of the corresponding signature gene sets of distinct subclusters indicated that subclusters of osteoblasts_3, osteoblasts_4, osteoblasts_5, and osteoblasts_6 are relatively more mature during the osteoblastic progression of HOE. The trajectory analysis of the osteoblasts demonstrated that some genes were gradually downregulated, such as CRYAB, CCL3, SFRP4, WIF1, and IGFBP3, while other critical genes were upregulated, such as VCAN, IGFBP4, FSTL1, POSTN, MDK, THBS2, and ALPL, suggesting that these factors may participate in HOE progression. Cell-cell communication networks revealed extensive molecular interactions among the 13 HOE clusters. Ligand-receptor pairs for IL6, COL24A1, COL22A1, VWF, FZD6, FGF2, and NOTCH1 were identified, suggesting that multiple signaling pathways may be involved in HOE. In conclusion, this study provided the cellular atlas for HOE. We have established a greater extent of the heterogeneity of HOE cells than previously known through transcriptomic analysis at the single-cell level. We have observed gradual patterns of signature gene expression during the differentiation and maturation progression of osteoblasts from stem cells in HOE with higher resolution. The cell heterogeneity of HOE deserves further investigation to pave the way for identification of potential targets for HOE early diagnosis and therapeutic treatment.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Orthopedics, Peking University International Hospital, Beijing, China
- Biomedical Engineering Department, Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Dan Xiao
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Beijing, China
| | - Li Shu
- Department of Orthopedics, Peking University International Hospital, Beijing, China
| | - Maoqi Gong
- Department of Orthopedic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xinghua Liu
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Beijing, China
| | - Xieyuan Jiang
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Beijing, China
| |
Collapse
|
5
|
Samuel JT, Nwokeji IN, Ali S, Jakkula NR, Fung J, Campbell A, Singanamala N. Immobile to Ambulation: Complex Multijoint Pathologies in a Patient with Advanced HIV Disease: A Case Report. JBJS Case Connect 2024; 14:01709767-202409000-00053. [PMID: 39270047 DOI: 10.2106/jbjs.cc.24.00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
CASE A 30-year-old man with a history of advanced HIV disease (AHD) presented with bilateral equinocavus, leg, and foot muscle paresis, Brooker grade 4 heterotopic ossification of hips and knee stiffness, and was unable to sit upright, stand independently, or walk. Electromyography showed demyelinating sensorimotor and axonal polyneuropathy of lower extremities. Multiple surgeries of bilateral hips, ankles, and feet enabled joint mobility, plantigrade feet, and independent ambulation. CONCLUSION Patients with AHD may develop multijoint pathologies, secondary to HIV, antiretroviral therapy, or prolonged immobility, resulting in loss of ambulation and independence. Restoring ambulation may necessitate multiple surgeries, with potential for success.
Collapse
Affiliation(s)
- Justin T Samuel
- CUNY School of Medicine, New York, New York
- Department of Orthopedic Surgery, St. Barnabas Hospital, Bronx, New York
| | - Imani N Nwokeji
- CUNY School of Medicine, New York, New York
- Department of Orthopedic Surgery, St. Barnabas Hospital, Bronx, New York
| | - Sehrish Ali
- CUNY School of Medicine, New York, New York
- Department of Orthopedic Surgery, St. Barnabas Hospital, Bronx, New York
| | | | - Jonathan Fung
- Department of Orthopedic Surgery, St. Barnabas Hospital, Bronx, New York
| | - Andrew Campbell
- Department of Orthopedic Surgery, St. Barnabas Hospital, Bronx, New York
| | - Naveen Singanamala
- Department of Orthopedic Surgery, St. Barnabas Hospital, Bronx, New York
| |
Collapse
|
6
|
Vishlaghi N, Guo L, Griswold-Wheeler D, Sun Y, Booker C, Crossley JL, Bancroft AC, Juan C, Korlakunta S, Ramesh S, Pagani CA, Xu L, James AW, Tower RJ, Dellinger M, Levi B. Vegfc-expressing cells form heterotopic bone after musculoskeletal injury. Cell Rep 2024; 43:114049. [PMID: 38573853 DOI: 10.1016/j.celrep.2024.114049] [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: 08/30/2023] [Revised: 02/09/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024] Open
Abstract
Heterotopic ossification (HO) is a challenging condition that occurs after musculoskeletal injury and is characterized by the formation of bone in non-skeletal tissues. While the effect of HO on blood vessels is well established, little is known about its impact on lymphatic vessels. Here, we use a mouse model of traumatic HO to investigate the relationship between HO and lymphatic vessels. We show that injury triggers lymphangiogenesis at the injury site, which is associated with elevated vascular endothelial growth factor C (VEGF-C) levels. Through single-cell transcriptomic analyses, we identify mesenchymal progenitor cells and tenocytes as sources of Vegfc. We demonstrate by lineage tracing that Vegfc-expressing cells undergo osteochondral differentiation and contribute to the formation of HO. Last, we show that Vegfc haploinsufficiency results in a nearly 50% reduction in lymphangiogenesis and HO formation. These findings shed light on the complex mechanisms underlying HO formation and its impact on lymphatic vessels.
Collapse
Affiliation(s)
- Neda Vishlaghi
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Lei Guo
- Department of Population and Data Sciences, University of Texas Southwestern, Dallas, TX, USA
| | | | - Yuxiao Sun
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Cori Booker
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Janna L Crossley
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Alec C Bancroft
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Conan Juan
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Sneha Korlakunta
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Sowmya Ramesh
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Chase A Pagani
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Lin Xu
- Department of Population and Data Sciences, University of Texas Southwestern, Dallas, TX, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Robert J Tower
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Michael Dellinger
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA.
| | - Benjamin Levi
- Department of Surgery, University of Texas Southwestern, Dallas, TX, USA.
| |
Collapse
|
7
|
Sancilio C, Mastroianni M, Mousad A, Jesus ND, McCormick F. Heterotopic Ossification Secondary to Motor Vehicle Collision Trauma Leading to Femoroacetabular Impingement Syndrome: A Case Report. J Orthop Case Rep 2024; 14:67-72. [PMID: 38681919 PMCID: PMC11043993 DOI: 10.13107/jocr.2024.v14.i04.4362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/11/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction A patient presented for recalcitrant right hip pain secondary to femoroacetabular impingement (FAI) after blunt motor vehicle trauma and following the development of a 12 cm heterotopic ossification (HO). FAI is an increasingly recognized diagnosis where the hip joint is exposed to repeated femoral microtrauma from high-level physical activity or trauma, often causing labral ossification, and perhaps underlying a similar biological mechanism to HO. Case Report In this case report, we have an otherwise healthy 49-year-old male who was involved in a high-speed motor vehicle collision who was diagnosed with right hip FAI secondary to HO (Brooker's Class IV) and indicated for surgical excision of the HO anterior to the right proximal femur. The care team and patient initially trialed non-operative conservative treatment with non-steroidal anti-inflammatories drugs (NSAIDs) and hypothesized therapeutic success using a non-surgical approach. Surgical resection was pursued with the patient after a failure of conservative measures. The patient reported a zero out of ten on a ten-point numerical rating scale for pain, he also stated improved quality of life, satisfaction with the procedure, and subsequent rehabilitation at 1-month post-operative follow-up. Conclusion HO with near complete ankylosis of the hip joint may be causative of FAI when untreated. Although this case demonstrates a rarely studied traumatic etiology of impingement secondary to HO, initial standard conservative anti-inflammatory treatment can still be pursued. By analyzing the periarticular impact of HO secondary to non-surgical trauma, we can utilize and make inferential correlations from the literature, studying HO and impingement in the setting of prior hip surgery to guide treatment and prognosis in those presenting with FAI symptoms secondary to blunt force trauma.
Collapse
Affiliation(s)
| | - Michael Mastroianni
- Columbia University Irving Medical Center/New York Presbyterian Hospital, New York, United States
| | - Albert Mousad
- Department of Biology,Tufts University School of Medicine, Boston, Massachusetts
| | - Nicholas De Jesus
- Department of Biology, Florida Atlantic University, Boca Raton, Florida
| | - Frank McCormick
- Beth Israel Deaconess Medical Center at Signature Healthcare, Brockton, Massachusetts
| |
Collapse
|
8
|
Ni H, Tang S, Zhang Y. A fibrodysplasia ossificans progressiva patient with a rare missense mutation in ACVR1 detected on 18F-FDG PET/CT. Joint Bone Spine 2024; 91:105682. [PMID: 38159793 DOI: 10.1016/j.jbspin.2023.105682] [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: 11/06/2023] [Revised: 12/03/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an exceedingly rare human genetic disorder characterized by the progressive and incapacitating formation of ectopic bone outside the skeleton. We report a case of FOP patient with mutations within the ACVR1 gene (c.982G>A; p.G328R). 18F-FDG positron emission tomography/computed tomography (PET/CT) was carried out for disease assessment. Previous studies have shown increased FDG uptake in regions of heterotopic ossification (HO) in FOP. However, in our study, the PET/CT features demonstrate that active ossificans exhibit increased 18F-FDG uptake, whereas end-stage ossifications do not. Collectively, 18F-FDG PET/CT emerges as a prospective approach to evaluate medication efficacy in the early stages, directing early intervention and pharmacological management of FOP before ossifications formation.
Collapse
Affiliation(s)
- Haopeng Ni
- Department of Nuclear Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
| | - Songhan Tang
- Department of Nuclear Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China
| | - Ying Zhang
- Department of Nuclear Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, 310000 Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Fan H, Cheng Q, Lin K, Gong L, Kan C, Wang S, Zheng H. Metformin alleviates genetic and traumatic heterotopic ossification by inhibiting infiltration and mitochondrial metabolism of myeloid cells. Am J Transl Res 2024; 16:255-271. [PMID: 38322576 PMCID: PMC10839392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/15/2023] [Indexed: 02/08/2024]
Abstract
OBJECTIVES Heterotopic ossification (HO), whether hereditary or traumatic, refers to the abnormal formation of bone in extraskeletal sites, often triggered by inflammation or flare-ups. Unfortunately, there are currently no effective treatments for HO. Metformin is well-known for its anti-diabetic, anti-inflammatory, anti-aging, and anti-cancer effects. However, its potential role in treating HO remains uncertain. METHODS Metformin was dissolved into water and given to mice. All the mice in this study were examined by microCT and myeloid cell quantification using flow cytometry. Complex activity kit was used to examine the activity of mitochondrial complexes of myeloid cells. RESULTS In this study, we discovered that metformin effectively inhibits genetic and traumatic HO formation and progression. Additionally, we observed a significant increase in myeloid cells in the genetic and traumatic HO mouse model compared to uninjured mice. Notably, metformin specifically reduced the infiltration of myeloid cells into the injured sites of the genetic and traumatic HO model mice. Further investigations revealed that metformin targets mitochondrial complex I and suppresses mitochondrial metabolism in myeloid cells. CONCLUSION These findings suggest that metformin suppresses HO development by potentially downregulating the mitochondrial metabolism of myeloid cells, offering a promising therapeutic option for HO treatment.
Collapse
Affiliation(s)
- Haitao Fan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical UniversityNo. 81 Meishan Road, Hefei 230022, Anhui, China
- Department of Neurospinal Surgery, The First Affiliated Hospital of Ningbo UniversityNingbo 315010, Zhejiang, China
| | - Qirong Cheng
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical UniversityNo. 81 Meishan Road, Hefei 230022, Anhui, China
| | - Keqiong Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical UniversityNo. 81 Meishan Road, Hefei 230022, Anhui, China
| | - Liangju Gong
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical UniversityNo. 81 Meishan Road, Hefei 230022, Anhui, China
| | - Chen Kan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical UniversityNo. 81 Meishan Road, Hefei 230022, Anhui, China
| | - Siying Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical UniversityNo. 81 Meishan Road, Hefei 230022, Anhui, China
| | - Hong Zheng
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical UniversityNo. 81 Meishan Road, Hefei 230022, Anhui, China
| |
Collapse
|
10
|
Mierzejewski B, Pulik Ł, Grabowska I, Sibilska A, Ciemerych MA, Łęgosz P, Brzoska E. Coding and noncoding RNA profile of human heterotopic ossifications - Risk factors and biomarkers. Bone 2023; 176:116883. [PMID: 37597797 DOI: 10.1016/j.bone.2023.116883] [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: 07/06/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Heterotopic ossification (HO) means the formation of bone in muscles and soft tissues, such as ligaments or tendons. HO could have a genetic history or develop after a traumatic event, as a result of muscle injury, fractures, burns, surgery, or neurological disorders. Many lines of evidence suggest that the formation of HO is related to the pathological differentiation of stem or progenitor cells present within soft tissues or mobilized from the bone marrow. The cells responsible for the initiation and progression of HO are generally called HO precursor cells. The exact mechanisms behind the development of HO are not fully understood. However, several factors have been identified as potential contributors. For example, local tissue injury and inflammation disturb soft tissue homeostasis. Inflammatory cells release growth factors and cytokines that promote osteogenic or chondrogenic differentiation of HO precursor cells. The bone morphogenetic protein (BMP) is one of the main factors involved in the development of HO. In this study, next-generation sequencing (NGS) and RT-qPCR were performed to analyze the differences in mRNA, miRNA, and lncRNA expression profiles between muscles, control bone samples, and HO samples coming from patients who underwent total hip replacement (THR). As a result, crucial changes in the level of gene expression between HO and healthy tissues were identified. The bioinformatic analysis allowed to describe the processes most severely impacted, as well as genes which level differed the most significantly between HO and control samples. Our analysis showed that the level of transcripts involved in leukocyte migration, differentiation, and activation, as well as markers of chronic inflammatory diseases, that is, miR-148, increased in HO, as compared to muscle. Furthermore, the levels of miR-195 and miR-143, which are involved in angiogenesis, were up-regulated in HO, as compared to bone. Thus, we suggested that inflammation and angiogenesis play an important role in HO formation. Importantly, we noticed that HO is characterized by a higher level of TLR3 expression, compared to muscle and bone. Thus, we suggest that infection may also be a risk factor in HO development. Furthermore, an increased level of transcripts coding proteins involved in osteogenesis and signaling pathways, such as ALPL, SP7, BGLAP, BMP8A, BMP8B, SMPD3 was noticed in HO, as compared to muscles. Interestingly, miR-99b, miR-146, miR-204, and LINC00320 were up-regulated in HO, comparing to muscles and bone. Therefore, we suggested that these molecules could be important biomarkers of HO formation and a potential target for therapies.
Collapse
Affiliation(s)
- Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Łukasz Pulik
- Department of Orthopedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Aleksandra Sibilska
- Department of Orthopedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland
| | - Maria Anna Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland
| | - Paweł Łęgosz
- Department of Orthopedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland.
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland.
| |
Collapse
|
11
|
Yea JH, Gomez-Salazar M, Onggo S, Li Z, Thottappillil N, Cherief M, Negri S, Xing X, Qin Q, Tower RJ, Fan CM, Levi B, James AW. Tppp3 + synovial/tendon sheath progenitor cells contribute to heterotopic bone after trauma. Bone Res 2023; 11:39. [PMID: 37479686 PMCID: PMC10361999 DOI: 10.1038/s41413-023-00272-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 07/23/2023] Open
Abstract
Heterotopic ossification (HO) is a pathological process resulting in aberrant bone formation and often involves synovial lined tissues. During this process, mesenchymal progenitor cells undergo endochondral ossification. Nonetheless, the specific cell phenotypes and mechanisms driving this process are not well understood, in part due to the high degree of heterogeneity of the progenitor cells involved. Here, using a combination of lineage tracing and single-cell RNA sequencing (scRNA-seq), we investigated the extent to which synovial/tendon sheath progenitor cells contribute to heterotopic bone formation. For this purpose, Tppp3 (tubulin polymerization-promoting protein family member 3)-inducible reporter mice were used in combination with either Scx (Scleraxis) or Pdgfra (platelet derived growth factor receptor alpha) reporter mice. Both tendon injury- and arthroplasty-induced mouse experimental HO models were utilized. ScRNA-seq of tendon-associated traumatic HO suggested that Tppp3 is an early progenitor cell marker for either tendon or osteochondral cells. Upon HO induction, Tppp3 reporter+ cells expanded in number and partially contributed to cartilage and bone formation in either tendon- or joint-associated HO. In double reporter animals, both Pdgfra+Tppp3+ and Pdgfra+Tppp3- progenitor cells gave rise to HO-associated cartilage. Finally, analysis of human samples showed a substantial population of TPPP3-expressing cells overlapping with osteogenic markers in areas of heterotopic bone. Overall, these data demonstrate that synovial/tendon sheath progenitor cells undergo aberrant osteochondral differentiation and contribute to HO after trauma.
Collapse
Affiliation(s)
- Ji-Hye Yea
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mario Gomez-Salazar
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Sharon Onggo
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Orthopaedic and Trauma Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology of the University of Verona, Verona, Italy
| | - Xin Xing
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Robert Joel Tower
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Chen-Ming Fan
- Carnegie Institution for Science, Baltimore, MD, USA
| | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
12
|
Boncompagni A, Lucas-Herald AK, Beattie P, McDevitt H, Iughetti L, Constantinou P, Kinning E, Ahmed SF, Mason A. Progressive osseous heteroplasia: A case report with an unexpected trigger. Bone Rep 2023; 18:101665. [PMID: 36936194 PMCID: PMC10015177 DOI: 10.1016/j.bonr.2023.101665] [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: 11/10/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Progressive osseous heteroplasia (POH) is a rare genetic disorder characterised by progressive heterotopic ossification (HO) within the skin and subcutaneous tissues. The condition is caused by heterozygous inactivating mutations of the GNAS gene and usually presents in infancy. We describe the case of a white male ex-preterm who was first referred because of subcutaneous calcium deposits along the right arm after extravasation of parenteral nutrition. As these lesions progressed, a skin biopsy was undertaken which revealed intramembranous ossification. Genetic testing revealed a constitutional, de novo, heterozygous, nonsense variant in the GNAS gene that has not previously been described, but which is consistent with patient's clinical diagnosis of POH. No endocrine abnormalities or other signs congruent with overlapping conditions were detected. To the best of our knowledge, this is the first case describing an inflammatory trigger in POH. Trials with intravenous bisphosphonate and glucocorticoid as well as with topical sodium thiosulphate were attempted without clinical improvement. Excision of the calcifications and physiotherapy seem to have provided a partial improvement on mobility of the elbow. This case widens the spectrum of phenotypes seen in GNAS mutation disorders and suggests that alternative anti-inflammatory treatments may be effective. Mutations in GNAS should be considered in cases of significant progressive calcium deposition after extravasation injury.
Collapse
Affiliation(s)
- Alessandra Boncompagni
- Developmental Endocrinology Research Group, Royal Hospital for Children, University of Glasgow, Glasgow, United Kingdom
- Postgraduate School of Paediatrics, Department of Medical and Surgical Sciences of Mothers, Children and Adults, University of Modena & Reggio Emilia, Paediatric Unit, Modena, Italy
| | - Angela K. Lucas-Herald
- Developmental Endocrinology Research Group, Royal Hospital for Children, University of Glasgow, Glasgow, United Kingdom
| | - Paula Beattie
- Department of Paediatric Dermatology, Royal Hospital for Children, Glasgow, United Kingdom
| | - Helen McDevitt
- Developmental Endocrinology Research Group, Royal Hospital for Children, University of Glasgow, Glasgow, United Kingdom
| | - Lorenzo Iughetti
- Postgraduate School of Paediatrics, Department of Medical and Surgical Sciences of Mothers, Children and Adults, University of Modena & Reggio Emilia, Paediatric Unit, Modena, Italy
| | - Panayiotis Constantinou
- Department of Clinical Genetics, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Esther Kinning
- Department of Clinical Genetics, Queen Elizabeth University Hospital, Glasgow, United Kingdom
- Department of Clinical Genetics, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
| | - S. Faisal Ahmed
- Developmental Endocrinology Research Group, Royal Hospital for Children, University of Glasgow, Glasgow, United Kingdom
| | - Avril Mason
- Developmental Endocrinology Research Group, Royal Hospital for Children, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
13
|
Hou J, Chen J, Fan J, Tang Z, Zhou W, Lin H. Inhibition of NF-κB Signaling-Mediated Crosstalk Between Macrophages and Preosteoblasts by Metformin Alleviates Trauma-Induced Heterotopic Ossification. Inflammation 2023:10.1007/s10753-023-01817-2. [PMID: 37115368 DOI: 10.1007/s10753-023-01817-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023]
Abstract
Heterotopic ossification (HO) is a pathological condition that occurs in soft tissues following severe trauma. The exact pathogenesis of HO remains unclear. Studies have shown that inflammation predisposes patients to the development of HO and triggers ectopic bone formation. Macrophages are crucial mediators of inflammation and are involved in HO development. The present study investigated the inhibitory effect and underlying mechanism of metformin on macrophage infiltration and traumatic HO in mice. Our results found that abundant levels of macrophages were recruited to the injury site during early HO progression and that early administration of metformin prevented traumatic HO in mice. Furthermore, we found that metformin attenuated macrophage infiltration and the NF-κB signaling pathway in injured tissue. The monocyte-to-macrophage transition in vitro was suppressed by metformin and this event was mediated by AMPK. Finally, we showed that inflammatory mediator's regulation by macrophages targeted preosteoblasts, leading to elevated BMP signaling, and osteogenic differentiation and driving HO formation, and this effect was blocked after the activation of AMPK in macrophages. Collectively, our study suggests that metformin prevents traumatic HO by inhibiting of NF-κB signaling in macrophages and subsequently attenuating BMP signaling and osteogenic differentiation in preosteoblasts. Therefore, metformin may serve as a therapeutic drug for traumatic HO by targeting NF-κB signaling in macrophages.
Collapse
Affiliation(s)
- Jia Hou
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jie Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jingjing Fan
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhimin Tang
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenwen Zhou
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
14
|
Tu B, Li J, Sun Z, Zhang T, Liu H, Yuan F, Fan C. Macrophage-Derived TGF-β and VEGF Promote the Progression of Trauma-Induced Heterotopic Ossification. Inflammation 2023; 46:202-216. [PMID: 35986177 DOI: 10.1007/s10753-022-01723-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
Abstract
Heterotopic ossification (HO) is a pathological bone formation process caused by musculoskeletal trauma. HO is characterized by aberrant endochondral ossification and angiogenesis. Our previous studies have indicated that macrophage inflammation is involved in traumatic HO formation. In this study, we found that macrophage infiltration and TGF-β signaling activation are presented in human HO. Depletion of macrophages effectively suppressed traumatic HO formation in a HO mice model, and macrophage depletion significantly inhibited the activation of TGF-β/Smad2/3 signaling. In addition, the TGF-β blockade created by a neutralizing antibody impeded ectopic bone formation in vivo. Notably, endochondral ossification and angiogenesis are attenuated following macrophage depletion or TGF-β inhibition. Furthermore, our observations on macrophage polarization revealed that M2 macrophages, rather than M1 macrophages, play a critical role in supporting HO development by enhancing the osteogenic and chondrogenic differentiation of mesenchymal stem cells. Our findings on ectopic bone formation in HO patients and the mice model indicate that M2 macrophages are an important contributor for HO development, and that inhibition of M2 polarization or TGF-β activity may be a potential method of therapy for traumatic HO.
Collapse
Affiliation(s)
- Bing Tu
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Ziyang Sun
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Tongtong Zhang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Hang Liu
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Feng Yuan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
15
|
Pagani CA, Bancroft AC, Tower RJ, Livingston N, Sun Y, Hong JY, Kent RN, Strong AL, Nunez JH, Medrano JMR, Patel N, Nanes BA, Dean KM, Li Z, Ge C, Baker BM, James AW, Weiss SJ, Franceschi RT, Levi B. Discoidin domain receptor 2 regulates aberrant mesenchymal lineage cell fate and matrix organization. SCIENCE ADVANCES 2022; 8:eabq6152. [PMID: 36542719 PMCID: PMC9770942 DOI: 10.1126/sciadv.abq6152] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/05/2022] [Indexed: 06/17/2023]
Abstract
Extracellular matrix (ECM) interactions regulate both the cell transcriptome and proteome, thereby determining cell fate. Traumatic heterotopic ossification (HO) is a disorder characterized by aberrant mesenchymal lineage (MLin) cell differentiation, forming bone within soft tissues of the musculoskeletal system following traumatic injury. Recent work has shown that HO is influenced by ECM-MLin cell receptor signaling, but how ECM binding affects cellular outcomes remains unclear. Using time course transcriptomic and proteomic analyses, we identified discoidin domain receptor 2 (DDR2), a cell surface receptor for fibrillar collagen, as a key MLin cell regulator in HO formation. Inhibition of DDR2 signaling, through either constitutive or conditional Ddr2 deletion or pharmaceutical inhibition, reduced HO formation in mice. Mechanistically, DDR2 perturbation alters focal adhesion orientation and subsequent matrix organization, modulating Focal Adhesion Kinase (FAK) and Yes1 Associated Transcriptional Regulator and WW Domain Containing Transcription Regulator 1 (YAP/TAZ)-mediated MLin cell signaling. Hence, ECM-DDR2 interactions are critical in driving HO and could serve as a previously unknown therapeutic target for treating this disease process.
Collapse
Affiliation(s)
- Chase A. Pagani
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Alec C. Bancroft
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Robert J. Tower
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Nicholas Livingston
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Yuxiao Sun
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Jonathan Y. Hong
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Robert N. Kent
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Amy L. Strong
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Johanna H. Nunez
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Jessica Marie R. Medrano
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| | - Nicole Patel
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Benjamin A. Nanes
- Department of Dermatology, University of Texas Southwestern, Dallas, TX, USA
- Lydia Hill Department of Bioinformatics, University of Texas Southwestern, Dallas, TX, USA
| | - Kevin M. Dean
- Lydia Hill Department of Bioinformatics, University of Texas Southwestern, Dallas, TX, USA
- Cecil H. and The Ida Green Center for Systems Biology, University of Texas Southwestern, Dallas, TX, USA
| | - Zhao Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Chunxi Ge
- School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Brendon M. Baker
- Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen J. Weiss
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
| |
Collapse
|
16
|
Pulik Ł, Mierzejewski B, Sibilska A, Grabowska I, Ciemerych MA, Łęgosz P, Brzóska E. The role of miRNA and lncRNA in heterotopic ossification pathogenesis. Stem Cell Res Ther 2022; 13:523. [PMID: 36522666 PMCID: PMC9753082 DOI: 10.1186/s13287-022-03213-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Heterotopic ossification (HO) is the formation of bone in non-osseous tissues, such as skeletal muscles. The HO could have a genetic or a non-genetic (acquired) background, that is, it could be caused by musculoskeletal trauma, such as burns, fractures, joint arthroplasty (traumatic HO), or cerebral or spinal insult (neurogenetic HO). HO formation is caused by the differentiation of stem or progenitor cells induced by local or systemic imbalances. The main factors described so far in HO induction are TGFβ1, BMPs, activin A, oncostatin M, substance P, neurotrophin-3, and WNT. In addition, dysregulation of noncoding RNAs, such as microRNA or long noncoding RNA, homeostasis may play an important role in the development of HO. For example, decreased expression of miRNA-630, which is responsible for the endothelial-mesenchymal transition, was observed in HO patients. The reduced level of miRNA-421 in patients with humeral fracture was shown to be associated with overexpression of BMP2 and a higher rate of HO occurrence. Down-regulation of miRNA-203 increased the expression of runt-related transcription factor 2 (RUNX2), a crucial regulator of osteoblast differentiation. Thus, understanding the various functions of noncoding RNAs can reveal potential targets for the prevention or treatment of HO.
Collapse
Affiliation(s)
- Łukasz Pulik
- Department of Orthopaedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005, Warsaw, Poland.
| | - Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Aleksandra Sibilska
- Department of Orthopaedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005, Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Maria Anna Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Paweł Łęgosz
- Department of Orthopaedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005, Warsaw, Poland
| | - Edyta Brzóska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| |
Collapse
|
17
|
Yamamoto M, Stoessel SJ, Yamamoto S, Goldhamer DJ. Overexpression of Wild-Type ACVR1 in Fibrodysplasia Ossificans Progressiva Mice Rescues Perinatal Lethality and Inhibits Heterotopic Ossification. J Bone Miner Res 2022; 37:2077-2093. [PMID: 35637634 PMCID: PMC9708949 DOI: 10.1002/jbmr.4617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/22/2022] [Accepted: 05/28/2022] [Indexed: 11/07/2022]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a devastating disease of progressive heterotopic bone formation for which effective treatments are currently unavailable. FOP is caused by dominant gain-of-function mutations in the receptor ACVR1 (also known as ALK2), which render the receptor inappropriately responsive to activin ligands. In previous studies, we developed a genetic mouse model of FOP that recapitulates most clinical aspects of the disease. In this model, genetic loss of the wild-type Acvr1 allele profoundly exacerbated heterotopic ossification, suggesting the hypothesis that the stoichiometry of wild-type and mutant receptors dictates disease severity. Here, we tested this model by producing FOP mice that conditionally overexpress human wild-type ACVR1. Injury-induced heterotopic ossification (HO) was completely blocked in FOP mice when expression of both the mutant and wild-type receptor were targeted to Tie2-positive cells, which includes fibro/adipogenic progenitors (FAPs). Perinatal lethality of Acvr1R206H/+ mice was rescued by constitutive ACVR1 overexpression, and these mice survived to adulthood at predicted Mendelian frequencies. Constitutive overexpression of ACVR1 also provided protection from spontaneous abnormal skeletogenesis, and the incidence and severity of injury-induced HO in these mice was dramatically reduced. Analysis of pSMAD1/5/8 signaling both in cultured cells and in vivo indicates that ACVR1 overexpression functions cell-autonomously by reducing osteogenic signaling in response to activin A. We propose that ACVR1 overexpression inhibits HO by decreasing the abundance of ACVR1(R206H)-containing signaling complexes at the cell surface while increasing the representation of activin-A-bound non-signaling complexes comprised of wild-type ACVR1. © 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)
- Masakazu Yamamoto
- Department of Molecular and Cell BiologyUniversity of Connecticut Stem Cell Institute, University of ConnecticutStorrsCTUSA
| | - Sean J Stoessel
- Department of Molecular and Cell BiologyUniversity of Connecticut Stem Cell Institute, University of ConnecticutStorrsCTUSA
| | - Shoko Yamamoto
- Department of Molecular and Cell BiologyUniversity of Connecticut Stem Cell Institute, University of ConnecticutStorrsCTUSA
| | - David J Goldhamer
- Department of Molecular and Cell BiologyUniversity of Connecticut Stem Cell Institute, University of ConnecticutStorrsCTUSA
| |
Collapse
|
18
|
Severe Heterotopic Ossification After Revision Total Knee Arthroplasty: A Case Report and Review of the Literature. J Am Acad Orthop Surg Glob Res Rev 2022; 6:01979360-202211000-00009. [PMID: 36733984 PMCID: PMC9663138 DOI: 10.5435/jaaosglobal-d-22-00053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 09/08/2022] [Indexed: 12/04/2022]
Abstract
The incidence of primary and revision total knee arthroplasty (TKA) is increasing worldwide. Heterotopic ossification is a common and concerning complication of TKA. There are few described cases of severe heterotopic ossification after revision TKA and no known cases of heterotopic ossification causing functional ankylosis after revision TKA. We describe a case of extensive heterotopic ossification in a patient who underwent right TKA for extensive adhesions and stiffness. After early range of motion improvement postoperatively, the patient discontinued a physical therapy regimen. The patient presented 13 years after revision TKA with radiographically evidenced severe heterotopic ossification resulting in a functional ankylosis. The patient elected for nonsurgical management. This case demonstrates a delayed finding of severe heterotopic ossification. The case prompted an applied literature review of several topics: heterotopic ossification as a complication of revision arthroplasty, the contribution of autoimmune and inflammatory conditions to heterotopic ossification; the use of medication, radiation, and physical therapy as prophylaxis against heterotopic ossification; and the range of treatment strategies for severe heterotopic ossification at the knee joint. Consent by the patient involved in this case report was obtained.
Collapse
|
19
|
Fetuin-A is an immunomodulator and a potential therapeutic option in BMP4-dependent heterotopic ossification and associated bone mass loss. Bone Res 2022; 10:62. [PMID: 36289197 PMCID: PMC9605967 DOI: 10.1038/s41413-022-00232-x] [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: 02/20/2021] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 11/08/2022] Open
Abstract
Heterotopic ossification (HO) is the abnormal formation of bone in extraskeletal sites. However, the mechanisms linking HO pathogenesis with bone mass dysfunction remain unclear. Here, we showed that mice harboring injury-induced and BMP4-dependent HO exhibit bone mass loss similar to that presented by patients with HO. Moreover, we found that injury-induced hyperinflammatory responses at the injury site triggered HO initiation but did not result in bone mass loss at 1 day post-injury (dpi). In contrast, a suppressive immune response promoted HO propagation and bone mass loss by 7 dpi. Correcting immune dysregulation by PD1/PDL1 blockade dramatically alleviated HO propagation and bone mass loss. We further demonstrated that fetuin-A (FetA), which has been frequently detected in HO lesions but rarely observed in HO-adjacent normal bone, acts as an immunomodulator to promote PD1 expression and M2 macrophage polarization, leading to immunosuppression. Intervention with recombinant FetA inhibited hyperinflammation and prevented HO and associated bone mass loss. Collectively, our findings provide new insights into the osteoimmunological interactions that occur during HO formation and suggest that FetA is an immunosuppressor and a potential therapeutic option for the treatment of HO.
Collapse
|
20
|
Singh S, Morshed S, Motamedi D, Kidane J, Paul A, Hsiao EC, Wentworth KL. Identification of Risk Factors in the Development of Heterotopic Ossification After Primary Total Hip Arthroplasty. J Clin Endocrinol Metab 2022; 107:e3944-e3952. [PMID: 35451005 PMCID: PMC9387692 DOI: 10.1210/clinem/dgac249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Indexed: 11/19/2022]
Abstract
PURPOSE Heterotopic ossification (HO) is a process by which bone forms abnormally in soft tissues. Known risk factors for developing HO include male sex, spinal cord injury, trauma, and surgery. We investigated additional risk factors in the development of HO after hip arthroplasty. METHODS We performed a retrospective review of electronic medical records of 4070 individuals who underwent hip arthroplasty from September 2010 to October 2019 at the University of California, San Francisco Hospital. Demographics, anthropometrics, medications, and comorbid conditions were used in logistic regression analysis to identify factors associated with the development of HO. RESULTS A total of 2541 patients underwent primary hip arthroplasty in the analyzed timeframe (46.04% men, mean age at procedure: 62.13 ± 13.29 years). The incidence of postsurgical HO was 3% (n = 80). A larger proportion of individuals who developed HO had underlying osteoporosis (P < 0.001), vitamin D deficiency (P < 0.001), spine disease (P < 0.001), type 1 or 2 diabetes (P < 0.001), amenorrhea (P = 0.037), postmenopausal status (P < 0.001), parathyroid disorders (P = 0.011), and history of pathologic fracture (P = 0.005). Significant predictors for HO development were Black/African American race [odds ratio (OR) 2.97, P = 0.005], preexisting osteoporosis (OR 2.72, P = 0.001), spine disease (OR 2.04, P = 0.036), and low estrogen states (OR 1.99, P = 0.025). In the overall group, 75.64% received perioperative nonsteroidal anti-inflammatory drugs (NSAIDs), which negatively correlated with HO formation (OR 0.39, P = 0.001). CONCLUSIONS We identified new factors potentially associated with an increased risk of developing HO after primary hip arthroplasty, including African American race, osteoporosis, and low estrogen states. These patients may benefit from HO prophylaxis, such as perioperative NSAIDs.
Collapse
Affiliation(s)
- Sukhmani Singh
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Saam Morshed
- Departments of Orthopedic Surgery, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Daria Motamedi
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph Kidane
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Alexandra Paul
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Edward C Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- The Institute for Human Genetics, The Program in Craniofacial Biology, and the Robert L. Kroc Chair in Rheumatic and Connective Tissue Diseases III, University of California-San Francisco, San Francisco, CA, USA
| | - Kelly L Wentworth
- Correspondence: Kelly Wentworth, MD, University of California, San Francisco, Zuckerberg San Francisco General Hospital, 1001 Potrero Ave, Room 3501A, UCSF Box 0874, San Francisco, CA 94110, USA.
| |
Collapse
|
21
|
Eekhoff EM, de Ruiter RD, Smilde BJ, Schoenmaker T, de Vries TJ, Netelenbos C, Hsiao EC, Scott C, Haga N, Grunwald Z, De Cunto CL, di Rocco M, Delai PLR, Diecidue RJ, Madhuri V, Cho TJ, Morhart R, Friedman CS, Zasloff M, Pals G, Shim JH, Gao G, Kaplan F, Pignolo RJ, Micha D. Gene Therapy for Fibrodysplasia Ossificans Progressiva: Feasibility and Obstacles. Hum Gene Ther 2022; 33:782-788. [PMID: 35502479 PMCID: PMC9419966 DOI: 10.1089/hum.2022.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/06/2022] [Indexed: 02/02/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare and devastating genetic disease, in which soft connective tissue is converted into heterotopic bone through an endochondral ossification process. Patients succumb early as they gradually become trapped in a second skeleton of heterotopic bone. Although the underlying genetic defect is long known, the inherent complexity of the disease has hindered the discovery of effective preventions and treatments. New developments in the gene therapy field have motivated its consideration as an attractive therapeutic option for FOP. However, the immune system's role in FOP activation and the as-yet unknown primary causative cell, are crucial issues which must be taken into account in the therapy design. While gene therapy offers a potential therapeutic solution, more knowledge about FOP is needed to enable its optimal and safe application.
Collapse
Affiliation(s)
- Elisabeth M.W. Eekhoff
- Section Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Ruben D. de Ruiter
- Section Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Bernard J. Smilde
- Section Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam, Vrije Universiteit, Amsterdam, The Netherlands
| | - Teun J. de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam, Vrije Universiteit, Amsterdam, The Netherlands
| | - Coen Netelenbos
- Section Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Edward C. Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, Institute for Human Genetics, Program in Craniofacial Biology, the Institute for Regeneration Medicine, University of California, San Francisco, California, USA
| | - Christiaan Scott
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Heath, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Nobuhiko Haga
- Rehabilitation Services Bureau, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Saitama, Japan
| | - Zvi Grunwald
- Department of Anesthesiology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, USA
| | - Carmen L. De Cunto
- Department of Pediatrics, Section Pediatric Rheumatology, Hospital Italiano Buenos Aires, Argentina
| | - Maja di Rocco
- Department of Pediatrics, Unit of Rare Diseases, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Patricia L. R. Delai
- Teaching and Research Institute of the Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Robert J. Diecidue
- Department of Oral and Maxillofacial Surgery, Sidney Kimmel Medical College, Philadelphia, USA
| | - Vrisha Madhuri
- Department of Paediatric Orthopaedics and Center for Stem Cell Research, Christian Medical College and Hospital, Vellore, India
| | - Tae-Joon Cho
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Rolf Morhart
- Department of Pediatrics, Garmisch-Partenkichen Medical Center, Garmisch-Partenkirchen, Germany
| | - Clive S. Friedman
- Schulich School of Medicine and Dentistry, Western University, Clinical Skills Building, London, Ontario, Canada
| | - Michael Zasloff
- Surgery and Pediatrics, MedStar Georgetown Transplant Institute, Washington, District of Columbia, USA
| | - Gerard Pals
- Department of Human Genetics, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Jae-Hyuck Shim
- Department of Medicine/Rheumatology, Horae Gene Therapy Center, Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Guangping Gao
- Department of Microbiology and Physiological Systems, Horae Gene Therapy Center, Viral Vector Core, Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Frederick Kaplan
- Department of Orthopaedic Surgery and Medicine, Center for Research in FOP and Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Dimitra Micha
- Department of Human Genetics, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Hrkac S, Novak R, Salai G, Grazio S, Vlahovic T, Grgurevic L. Heterotopic ossification vs. fracture healing: Extracellular vesicle cargo proteins shed new light on bone formation. Bone Rep 2022; 16:101177. [PMID: 35252484 PMCID: PMC8892095 DOI: 10.1016/j.bonr.2022.101177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 02/08/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an extremely rare disease in which bone tissue forms in extraskeletal sites, which is known as heterotopic ossification (HO). Extracellular vesicles (EVs) are small phospholipid-enclosed particles released by various cells which have an emerging, but not completely understood role in various (patho)physiological processes. In order to further study the pathophysiology of FOP we conducted a small observational study comparing the proteomic profiles of EV cargo, derived from pooled plasma of four patient groups: FOP patient (N = 1) during active disease phase (flare-up), FOP patients during remission (N = 2), patients after long bone fracture (N = 20) and healthy controls (N = 10). After isolation of EVs – their protein cargo was determined using liquid chromatography / mass spectrometry, after which a functional gene enrichment analysis was performed. Our results show a sizeable difference of the proteomics profiles in which EVs from the bone fracture group show significant activity of integrin interactions, Wnt, VEGF, IGF-1 and PDGF pathways; conversely, FOP patients' EVs indicate that HO occurs via processes of innate immunity and the Ephrin B signaling pathway. We hypothesize that the Ephrin B signaling (expressed in EVs) contributes to HO by aiding in mesenchymal stem cell recruitment and osteogenic differentiation, as well as by contributing to the inflammatory response, including macrophage chemotaxis and activation. This is, to our knowledge, the first published analysis of EV protein cargo in FOP. Proteomics-based analysis of extracellular vesicles’ protein cargo in FOP patients, bone fracture healing and controls. Marked differences in signaling pathways expressed in extracellular vesicles in FOP vs. patients with bone fractures. Ephrin B signaling pathway expressed in extracellular vesicles identified as a likely cogwheel in heterotopic ossification.
Collapse
|
23
|
Hu Y, Wang Z. Rapamycin prevents heterotopic ossification by inhibiting the mTOR pathway and oxidative stress. Biochem Biophys Res Commun 2021; 573:171-178. [PMID: 34419763 DOI: 10.1016/j.bbrc.2021.07.060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/13/2021] [Accepted: 07/18/2021] [Indexed: 01/02/2023]
Abstract
Rapamycin (RAPA), which was first described as an anti-fungal agent, is a potent immunosuppressant that suppresses tumors and inhibits the mTOR signaling pathway. Heterotopic ossification (HO) is abnormal bone formation outside the skeletal system (e.g., in muscles, tendons, articular capsules and other soft tissues), often due to trauma or injury. There are currently no drugs available to treat traumatic HO, largely due to limited understanding of the disease. In this study, we focused on the role of oxidative stress (OS) in the early stage of traumatic HO, and explored the underlying mechanism of traumatic HO by using RAPA to specifically inhibit the mTOR pathway, which is known to play a role in the pathogenesis of HO. To assess the effects of RAPA in traumatic HO, we used an NSE-BMP4 transgenic mouse model that develops ossification in response to traumatic injury and intramuscular injection of cardiotoxin to initiate injury. These mice were then treated with RAPA or vehicle intraperitoneally every other day for 2 weeks. Our results demonstrate that RAPA can inhibit HO through a number of different mechanisms. We show that OS and a strong inflammatory response contribute to the hypoxia associated with the early stages of HO, and that RAPA inhibits these responses. Furthermore, RAPA reduces the vascularization triggered by mTOR signaling that leads to HO formation. Therefore, we believe that RAPA could be an effective treatment for the early stages of HO.
Collapse
Affiliation(s)
- Yangyang Hu
- Department of Pathology, Anhui Provincial Hospital, The First Affiliated Hospital of the University of Science and Technology of China, 230002, Hefei, Anhui, China.
| | - Zhe Wang
- Department of Pathology, Anhui Provincial Hospital, The First Affiliated Hospital of the University of Science and Technology of China, 230002, Hefei, Anhui, China.
| |
Collapse
|
24
|
Peters J, Köhler HC, Oltmanns K, Besselmann M, Zwaan M, Gutcke A, Rüttermann M. Heterotope Ossifikationen nach Langzeitbeatmung bei Covid-19 Erkrankung. REHABILITATION 2021; 60:231-234. [PMID: 34428803 DOI: 10.1055/a-1339-5365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Jan Peters
- Oberstabsarzt und Assistenzarzt der Klinik für Orthopädie und Unfallchirurgie, Plastische, Rekonstruktive und Handchirurgie Bundeswehrkrankenhaus Westerstede
| | - Hans-Christian Köhler
- Oberfeldarzt und Leiter der Ambulanz der Klinik Orthopädie und Unfallchirurgie, Plastische, Rekonstruktive und Handchirurgie Bundeswehrkrankenhaus Westerstede
| | - Klaas Oltmanns
- Dr. med. Flottillenarzt und Leiter Schwerpunkt Pneumologie der Klinik für Innere Medizin Bundeswehrkrankenhaus Westerstede
| | | | - Martin Zwaan
- Prof. Dr. med. Ehemaliger Direktor des Instituts für Radiologie der Ammerland-Klinik Westerstede
| | - Andre Gutcke
- Oberstarzt und klinischer Direktor der Klinik für Orthopädie und Unfallchirurgie, Plastische, Rekonstruktive und Handchirurgie Bundeswehrkrankenhaus Westerstede
| | - Mike Rüttermann
- Dr. med. Facharzt für Plastische und Ästhetische Chirurgie, Handchirurgie. Fellow of EBOPRAS, Diplomate of the European Board of Hand Surgery (EBHS), Department of Plastic Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. HPC - Oldenburg, Institut für Hand- und Plastische Chirurgie, 26122 Oldenburg, Klinik für Orthopädie und Unfallchirurgie, Plastische, Rekonstruktive und Handchirurgie Bundeswehrkrankenhaus Westerstede
| |
Collapse
|
25
|
Sun Z, Li J, Luo G, Liu W, He Y, Wang F, Qian Y, Fan C. Pharmacological activation of SIRT1 by metformin prevented trauma-induced heterotopic ossification through inhibiting macrophage mediated inflammation. Eur J Pharmacol 2021; 909:174386. [PMID: 34332919 DOI: 10.1016/j.ejphar.2021.174386] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
Trauma-induced heterotopic ossification (HO) is the aberrant extra-skeletal bone formation that severely incapacitates patient's daily life. Inflammation is the first stage of this progression, becoming an appealing target of early therapeutic intervention. Metformin, a widely used antidiabetic drug, also poses the therapeutic potential to modulate various inflammatory-related diseases. Therefore, this study aimed to investigate the preventive effect of metformin on trauma-induced HO progression, and unveil the underlying molecular mechanisms. A murine burn/tenotomy model was established to mimic trauma-induced HO in vivo. The anti-inflammation and anti-ossification effects of metformin were evaluated by histological staining and micro-CT. The inhibitory effects of metformin on macrophages activation in vitro were examined by ELISA and qRT-PCR. The underlying molecular mechanisms were further explored by immunofluorescence staining and western-blotting in vivo. Increased macrophages infiltration and inflammatory responses were found at early stage during HO progression. However, metformin dose-dependently attenuated the macrophage-mediated inflammatory responses both in vivo and vitro, which might account for the inhibitory effect of metformin on chondrogenesis and HO formation after trauma. Furthermore, elevated SIRT1 expression and decreased NF-κB p65 acetylation were found in the beneficial effects of metformin. Moreover, similar preventive effects were also found in SRT1720 HCI, a specific SIRT1 activator, while were remarkably reversed after the administration of EX527 (a specific SIRT1 inhibitor) with metformin. Taken together, our results provide a novel evidence that metformin can effectively attenuate trauma-induced HO by mitigating macrophage inflammatory responses through inhibiting NF-κB signaling via SIRT1-dependent mechanisms, which favors future therapeutic investigations for trauma-related disease.
Collapse
Affiliation(s)
- Ziyang Sun
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Juehong Li
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Gang Luo
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Weixuan Liu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Yunwei He
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Feiyan Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China.
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, PR China; Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 201306, PR China.
| |
Collapse
|
26
|
Tu B, Yu B, Wang W, Li J, Yuan F, Zhu J, Fan C. Inhibition of IL-17 prevents the progression of traumatic heterotopic ossification. J Cell Mol Med 2021; 25:7709-7719. [PMID: 34189826 PMCID: PMC8358870 DOI: 10.1111/jcmm.16617] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/05/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022] Open
Abstract
Traumatic heterotopic ossification (HO) is the abnormal formation of bone in soft tissues as a consequence of injury. However, the pathological mechanisms leading to traumatic HO remain unknown. Here, we report that aberrant expression of IL-17 promotes traumatic HO formation by activating β-catenin signalling in mouse model. We found that elevated IL-17 and β-catenin levels are correlated with a high degree of HO formation in specimens from patients and HO animals. We also show that IL-17 initiates and promotes HO progression in mice. Local injection of an IL-17 neutralizing antibody attenuates ectopic bone formation in a traumatic mouse model. IL-17 enhances the osteoblastic differentiation of mesenchymal stem cells (MSCs) by activating β-catenin signalling. Moreover, inhibition of IL-17R or β-catenin signalling by neutralizing antibodies or drugs prevents the osteogenic differentiation of isolated MSCs and decreases HO formation in mouse models. Together, our study identifies a novel role for active IL-17 as the inducer and promoter of ectopic bone formation and suggests that IL-17 inhibition might be a potential therapeutic target in traumatic HO.
Collapse
Affiliation(s)
- Bing Tu
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bo Yu
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Wang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Feng Yuan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jing Zhu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
27
|
Liu S, Zheng J, Zheng Q. A Case Report of Heterotopic Ossification Caused by High-Voltage Electric Injury. J Burn Care Res 2020; 41:1118-1121. [PMID: 32453811 DOI: 10.1093/jbcr/iraa076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
This case report described the diagnosis and treatment of a patient with ectopic bone injury due to high-pressure electric shock. A 24-year-old male patient suffered from burns that covered 50% of TBSA, including on limbs and torso, after coming into contact with 10-kV high-voltage electricity. A repeated Meek micrografting technique (MEEK) was applied for wound healing; skin grafts were cut into micrografts and expanded at a ratio of 1:4 to cover large areas after burn trauma. After the injury, right elbow redness, fever, pains, and joint movement disorder were reported by the patient, which might be attributed to excessive exercises and acute scarring. However, these symptoms were not treated immediately. Six months after the injury, his right elbow joint showed 90° locking and a restricted 5° movement capacity. X-ray examination revealed a new bone formation at the inner tibia shin. In addition, 3D CT showed the formation of right tibia, the ruler bone, and the skull of the humming bridge. Under general anesthesia, the right elbow joint was released, the medial collateral ligament was repaired, and the hinge external fixator was fixed. A large number of hyperplastic bone masses were found at the right elbow joint during surgery, specifically in the foot bone hawk's beak nest. No complications after surgery were observed. X-ray examination in February showed disappearance of the bone bridge and normal relationship of the right elbow joint. This case study revealed that electric shock injury could lead to ectopic bone formation, and much attention should be paid on any changes that indicate aseptic inflammation, such as redness, swelling, fever, and pain during the treatment process. Finally, identification of the scar constriction phase could indicate surgical treatment in order to promote the limb rehabilitation process.
Collapse
Affiliation(s)
- Shengli Liu
- Department of Burn and Plastic Surgery, Southeast Hospital Affiliated to Xiamen University, Zhangzhou, China
| | - Jiansheng Zheng
- Department of Burn and Plastic Surgery, Southeast Hospital Affiliated to Xiamen University, Zhangzhou, China
| | - Qingyi Zheng
- Department of Burn and Plastic Surgery, Southeast Hospital Affiliated to Xiamen University, Zhangzhou, China
| |
Collapse
|
28
|
Li L, Tuan RS. Mechanism of traumatic heterotopic ossification: In search of injury-induced osteogenic factors. J Cell Mol Med 2020; 24:11046-11055. [PMID: 32853465 PMCID: PMC7576286 DOI: 10.1111/jcmm.15735] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Heterotopic ossification (HO) is a pathological condition of abnormal bone formation in soft tissue. Three factors have been proposed as required to induce HO: (a) osteogenic precursor cells, (b) osteoinductive agents and (c) an osteoconductive environment. Since Urist's landmark discovery of bone induction in skeletal muscle tissue by demineralized bone matrix, it is generally believed that skeletal muscle itself is a conductive environment for osteogenesis and that resident progenitor cells in skeletal muscle are capable of differentiating into osteoblast to form bone. However, little is known about the naturally occurring osteoinductive agents that triggered this osteogenic response in the first place. This article provides a review of the emerging findings regarding distinct types of HO to summarize the current understanding of HO mechanisms, with special attention to the osteogenic factors that are induced following injury. Specifically, we hypothesize that muscle injury‐induced up‐regulation of local bone morphogenetic protein‐7 (BMP‐7) level, combined with glucocorticoid excess‐induced down‐regulation of circulating transforming growth factor‐β1 (TGF‐β1) level, could be an important causative mechanism of traumatic HO formation.
Collapse
Affiliation(s)
- La Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
29
|
Pulik Ł, Mierzejewski B, Ciemerych MA, Brzóska E, Łęgosz P. The Survey of Cells Responsible for Heterotopic Ossification Development in Skeletal Muscles-Human and Mouse Models. Cells 2020; 9:cells9061324. [PMID: 32466405 PMCID: PMC7349686 DOI: 10.3390/cells9061324] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/16/2020] [Accepted: 05/21/2020] [Indexed: 12/18/2022] Open
Abstract
Heterotopic ossification (HO) manifests as bone development in the skeletal muscles and surrounding soft tissues. It can be caused by injury, surgery, or may have a genetic background. In each case, its development might differ, and depending on the age, sex, and patient's conditions, it could lead to a more or a less severe outcome. In the case of the injury or surgery provoked ossification development, it could be, to some extent, prevented by treatments. As far as genetic disorders are concerned, such prevention approaches are highly limited. Many lines of evidence point to the inflammatory process and abnormalities in the bone morphogenetic factor signaling pathway as the molecular and cellular backgrounds for HO development. However, the clear targets allowing the design of treatments preventing or lowering HO have not been identified yet. In this review, we summarize current knowledge on HO types, its symptoms, and possible ways of prevention and treatment. We also describe the molecules and cells in which abnormal function could lead to HO development. We emphasize the studies involving animal models of HO as being of great importance for understanding and future designing of the tools to counteract this pathology.
Collapse
Affiliation(s)
- Łukasz Pulik
- Department of Orthopaedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland;
| | - Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland; (B.M.); (M.A.C.)
| | - Maria A. Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland; (B.M.); (M.A.C.)
| | - Edyta Brzóska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland; (B.M.); (M.A.C.)
- Correspondence: (E.B.); (P.Ł.); Tel.: +48-22-5542-203 (E.B.); +48-22-5021-514 (P.Ł.)
| | - Paweł Łęgosz
- Department of Orthopaedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland;
- Correspondence: (E.B.); (P.Ł.); Tel.: +48-22-5542-203 (E.B.); +48-22-5021-514 (P.Ł.)
| |
Collapse
|
30
|
Pignolo RJ, Kaplan FS. Druggable targets, clinical trial design and proposed pharmacological management in fibrodysplasia ossificans progressiva. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1751122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Robert J. Pignolo
- Departments of Medicine, Physiology & Biomedical Engineering, and the Robert and Arlene Kogod Center on Aging, Alix School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Frederick S. Kaplan
- Departments of Orthopaedic Surgery, Medicine, and the Center for Research in FOP & Related Disorders, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
31
|
Kaji DA, Tan Z, Johnson GL, Huang W, Vasquez K, Lehoczky JA, Levi B, Cheah KS, Huang AH. Cellular Plasticity in Musculoskeletal Development, Regeneration, and Disease. J Orthop Res 2020; 38:708-718. [PMID: 31721278 PMCID: PMC7213644 DOI: 10.1002/jor.24523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/04/2019] [Indexed: 02/04/2023]
Abstract
In this review, we highlight themes from a recent workshop focused on "Plasticity of Cell Fate in Musculoskeletal Tissues" held at the Orthopaedic Research Society's 2019 annual meeting. Experts in the field provided examples of mesenchymal cell plasticity during normal musculoskeletal development, regeneration, and disease. A thorough understanding of the biology underpinning mesenchymal cell plasticity may offer a roadmap for promoting regeneration while attenuating pathologic differentiation. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:708-718, 2020.
Collapse
Affiliation(s)
- Deepak A. Kaji
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, NYC, NY, USA
| | - Zhijia Tan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Gemma L. Johnson
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA, USA,Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Wesley Huang
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kaetlin Vasquez
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jessica A. Lehoczky
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Benjamin Levi
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Alice H. Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, NYC, NY, USA
| |
Collapse
|
32
|
Brady RD, Zhao MZ, Wong KR, Casilla-Espinosa PM, Yamakawa GR, Wortman RC, Sun M, Grills BL, Mychasiuk R, O'Brien TJ, Agoston DV, Lee PVS, McDonald SJ, Robinson DL, Shultz SR. A novel rat model of heterotopic ossification after polytrauma with traumatic brain injury. Bone 2020; 133:115263. [PMID: 32032779 DOI: 10.1016/j.bone.2020.115263] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 01/08/2023]
Abstract
Neurological heterotopic ossification (NHO) is characterized by abnormal bone growth in soft tissue and joints in response to injury to the central nervous system. The ectopic bone frequently causes pain, restricts mobility, and decreases the quality of life for those affected. NHO commonly develops in severe traumatic brain injury (TBI) patients, particularly in the presence of concomitant musculoskeletal injuries (i.e. polytrauma). There are currently no animal models that accurately mimic these combinations of injuries, which has limited our understanding of NHO pathobiology, as well as the development of biomarkers and treatments, in TBI patients. In order to address this shortcoming, here we present a novel rat model that combines TBI, femoral fracture, and muscle crush injury. Young adult male Sprague Dawley rats were randomly assigned into three different injury groups: triple sham-injury, peripheral injury only (i.e., sham-TBI + fracture + muscle injury) or triple injury (i.e., TBI + fracture + muscle injury). Evidence of ectopic bone in the injured hind-limb, as confirmed by micro-computed tomography (μCT), was found at 6-weeks post-injury in 70% of triple injury rats, 20% of peripheral injury rats, and 0% of the sham-injured controls. Furthermore, the triple injury rats had higher ectopic bone severity scores than the sham-injured group. This novel model will provide a platform for future studies to identify underlying mechanisms, biomarkers, and develop evidence based pharmacological treatments to combat this debilitating long-term complication of TBI and polytrauma.
Collapse
Affiliation(s)
- Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
| | - Michael Z Zhao
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Ker R Wong
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Pablo M Casilla-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Ryan C Wortman
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Brian L Grills
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University, Bethesda, MD, USA
| | - Peter V S Lee
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Dale L Robinson
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia; Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
33
|
Huang Y, Wang X, Lin H. The hypoxic microenvironment: a driving force for heterotopic ossification progression. Cell Commun Signal 2020; 18:20. [PMID: 32028956 PMCID: PMC7006203 DOI: 10.1186/s12964-020-0509-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/02/2020] [Indexed: 12/23/2022] Open
Abstract
Heterotopic ossification (HO) refers to the formation of bone tissue outside the normal skeletal system. According to its pathogenesis, HO is divided into hereditary HO and acquired HO. There currently lack effective approaches for HO prevention or treatment. A deep understanding of its pathogenesis will provide promising strategies to prevent and treat HO. Studies have shown that the hypoxia-adaptive microenvironment generated after trauma is a potent stimulus of HO. The hypoxic microenvironment enhances the stability of hypoxia-inducible factor-1α (HIF-1α), which regulates a complex network including bone morphogenetic proteins (BMPs), vascular endothelial growth factor (VEGF), and neuropilin-1 (NRP-1), which are implicated in the formation of ectopic bone. In this review, we summarize the current understanding of the triggering role and underlying molecular mechanisms of the hypoxic microenvironment in the initiation and progression of HO, focusing mainly on HIF-1 and it's influenced genes BMP, VEGF, and NRP-1. A better understanding of the role of hypoxia in HO unveils novel therapeutic targets for HO that reduce the local hypoxic microenvironment and inhibit HIF-1α activity. Video Abstract. (MP4 52403 kb)
Collapse
Affiliation(s)
- Yifei Huang
- First Clinical Medical School, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Xinyi Wang
- First Clinical Medical School, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, 461 BaYi Avenue, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
34
|
Guder C, Gravius S, Burger C, Wirtz DC, Schildberg FA. Osteoimmunology: A Current Update of the Interplay Between Bone and the Immune System. Front Immunol 2020; 11:58. [PMID: 32082321 PMCID: PMC7004969 DOI: 10.3389/fimmu.2020.00058] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Immunology, already a discipline in its own right, has become a major part of many different medical fields. However, its relationship to orthopedics and trauma surgery has unfortunately, and perhaps unjustly, been developing rather slowly. Discoveries in recent years have emphasized the immense breadth of communication and connection between both systems and, importantly, the highly promising therapeutic opportunities. Recent discoveries of factors originally assigned to the immune system have now also been shown to have a significant impact on bone health and disease, which has greatly changed how we approach treatment of bone pathologies. In case of bone fracture, immune cells, especially macrophages, are present throughout the whole healing process, assure defense against pathogens and discharge a complex variety of effectors to regulate bone modeling. In rheumatoid arthritis and osteoporosis, the immune system contributes to the formation of the pathological and chronic conditions. Fascinatingly, prosthesis failure is not at all solely a mechanical problem of improper strain but works in conjunction with an active contribution of the immune system as a reaction to irritant debris from material wear. Unraveling conjoined mechanisms of the immune and osseous systems heralds therapeutic possibilities for ailments of both. Contemplation of the bone as merely an unchanging support pillar is outdated and obsolete. Instead it is mandatory that this highly diverse network be incorporated in our understanding of the immune system and hematopoiesis.
Collapse
Affiliation(s)
- Christian Guder
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Sascha Gravius
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany.,Department of Orthopedics and Trauma Surgery, University Medical Center Mannheim of University Heidelberg, Mannheim, Germany
| | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
35
|
Zhang Y, Zhan Y, Kou Y, Yin X, Wang Y, Zhang D. Identification of biological pathways and genes associated with neurogenic heterotopic ossification by text mining. PeerJ 2020; 8:e8276. [PMID: 31915578 PMCID: PMC6944123 DOI: 10.7717/peerj.8276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
Background Neurogenic heterotopic ossification is a disorder of aberrant bone formation affecting one in five patients sustaining a spinal cord injury or traumatic brain injury (SCI-TBI-HO). However, the underlying mechanisms of SCI-TBI-HO have proven difficult to elucidate. The aim of the present study is to identify the most promising candidate genes and biological pathways for SCI-TBI-HO. Methods In this study, we used text mining to generate potential explanations for SCI-TBI-HO. Moreover, we employed several additional datasets, including gene expression profile data, drug data and tissue-specific gene expression data, to explore promising genes that associated with SCI-TBI-HO. Results We identified four SCI-TBI-HO-associated genes, including GDF15, LDLR, CCL2, and CLU. Finally, using enrichment analysis, we identified several pathways, including integrin signaling, insulin pathway, internalization of ErbB1, urokinase-type plasminogen activator and uPAR-mediated signaling, PDGFR-beta signaling pathway, EGF receptor (ErbB1) signaling pathway, and class I PI3K signaling events, which may be associated with SCI-TBI-HO. Conclusions These results enhance our understanding of the molecular mechanisms of SCI-TBI-HO and offer new leads for researchers and innovative therapeutic strategies.
Collapse
Affiliation(s)
- Yichong Zhang
- Department of Trauma and Orthopaedic Surgery, Peking University People's Hospital, Beijing, China
| | - Yuanbo Zhan
- Department of Periodontology and Oral Mucosa, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yuhui Kou
- Department of Trauma and Orthopaedic Surgery, Peking University People's Hospital, Beijing, China
| | - Xiaofeng Yin
- Department of Trauma and Orthopaedic Surgery, Peking University People's Hospital, Beijing, China
| | - Yanhua Wang
- Department of Trauma and Orthopaedic Surgery, Peking University People's Hospital, Beijing, China
| | - Dianying Zhang
- Department of Trauma and Orthopaedic Surgery, Peking University People's Hospital, Beijing, China
| |
Collapse
|
36
|
Botman E, Treurniet S, Lubbers WD, Schwarte LA, Schober PR, Sabelis L, Peters EJG, van Schie A, de Vries R, Grunwald Z, Smilde BJ, Nieuwenhuijzen JA, Visser M, Micha D, Bravenboer N, Coen Netelenbos J, Teunissen BP, de Graaf P, Raijmakers PGHM, Smit JM, Eekhoff EMW. When Limb Surgery Has Become the Only Life-Saving Therapy in FOP: A Case Report and Systematic Review of the Literature. Front Endocrinol (Lausanne) 2020; 11:570. [PMID: 32973683 PMCID: PMC7472799 DOI: 10.3389/fendo.2020.00570] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 07/13/2020] [Indexed: 11/22/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare disease in which heterotopic ossification (HO) is formed in muscles, tendons and ligaments. Traumatic events, including surgery, are discouraged as this is known to trigger a flare-up with risk of subsequent HO. Anesthetic management for patients with FOP is challenging. Cervical spine fusion, ankylosis of the temporomandibular joints, thoracic insufficiency syndrome, restrictive chest wall disease, and sensitivity to oral trauma complicate airway management and anesthesia and pose life-threatening risks. We report a patient with FOP suffering from life-threatening antibiotic resistant bacterial infected ulcers of the right lower leg and foot. The anesthetic, surgical and postoperative challenges and considerations are discussed. In addition, the literature on limb surgeries of FOP patients is systemically reviewed. The 44 year-old female patient was scheduled for a through-knee amputation. Airway and pulmonary evaluation elicited severe abnormalities, rendering standard general anesthesia a rather complication-prone approach in this patient. Thus, regional anesthesia, supplemented with intravenous analgosedation and N2O-inhalation were performed in this case. The surgery itself was securely planned to avoid any unnecessary tissue damage. Postoperatively the patient was closely monitored for FOP activity by ultrasound and [18F]PET/CT-scan. One year after surgery, a non-significant amount of HO had formed at the operated site. The systematic review revealed seventeen articles in which thirty-two limb surgeries in FOP patients were described. HO reoccurrence was described in 90% of the cases. Clinical improvement due to improved mobility of the operated joint was noted in 16% of the cases. It should be noted, though, that follow-up time was limited and no or inadequate imaging modalities were used to follow-up in the majority of these cases. To conclude, if medically urgent, limb surgery in FOP is possible even when general anesthesia is not preferred. The procedure should be well-planned, alternative techniques or procedures should be tested prior to surgery and special attention should be paid to the correct positioning of the patient. According to the literature recurrent HO should be expected after surgery of a limb, even though it was limited in the case described.
Collapse
Affiliation(s)
- Esmée Botman
- Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sanne Treurniet
- Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Wouter D. Lubbers
- Department of Anesthesiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Lothar A. Schwarte
- Department of Anesthesiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Patrick R. Schober
- Department of Anesthesiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Louise Sabelis
- Department of Rehabilitation Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Edgar J. G. Peters
- Department of Internal Medicine Section of Infectious Diseases, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Annelies van Schie
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ralph de Vries
- Medical Library, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Zvi Grunwald
- Department of Anesthesiology, Jefferson Health System, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bernard J. Smilde
- Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Marieke Visser
- Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Dimitra Micha
- Department of Clinical Genetics, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - J. Coen Netelenbos
- Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bernd P. Teunissen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Pieter G. H. M. Raijmakers
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jan Maerten Smit
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Bone Center, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Elisabeth M. W. Eekhoff
- Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Elisabeth M. W. Eekhoff
| |
Collapse
|
37
|
Mantovani G, Bastepe M, Monk D, de Sanctis L, Thiele S, Ahmed SF, Bufo R, Choplin T, De Filippo G, Devernois G, Eggermann T, Elli FM, Ramirez AG, Germain-Lee EL, Groussin L, Hamdy NA, Hanna P, Hiort O, Jüppner H, Kamenický P, Knight N, Le Norcy E, Lecumberri B, Levine MA, Mäkitie O, Martin R, Martos-Moreno GÁ, Minagawa M, Murray P, Pereda A, Pignolo R, Rejnmark L, Rodado R, Rothenbuhler A, Saraff V, Shoemaker AH, Shore EM, Silve C, Turan S, Woods P, Zillikens MC, de Nanclares GP, Linglart A. Recommendations for Diagnosis and Treatment of Pseudohypoparathyroidism and Related Disorders: An Updated Practical Tool for Physicians and Patients. Horm Res Paediatr 2020; 93:182-196. [PMID: 32756064 PMCID: PMC8140671 DOI: 10.1159/000508985] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Patients affected by pseudohypoparathyroidism (PHP) or related disorders are characterized by physical findings that may include brachydactyly, a short stature, a stocky build, early-onset obesity, ectopic ossifications, and neurodevelopmental deficits, as well as hormonal resistance most prominently to parathyroid hormone (PTH). In addition to these alterations, patients may develop other hormonal resistances, leading to overt or subclinical hypothyroidism, hypogonadism and growth hormone (GH) deficiency, impaired growth without measurable evidence for hormonal abnormalities, type 2 diabetes, and skeletal issues with potentially severe limitation of mobility. PHP and related disorders are primarily clinical diagnoses. Given the variability of the clinical, radiological, and biochemical presentation, establishment of the molecular diagnosis is of critical importance for patients. It facilitates management, including prevention of complications, screening and treatment of endocrine deficits, supportive measures, and appropriate genetic counselling. Based on the first international consensus statement for these disorders, this article provides an updated and ready-to-use tool to help physicians and patients outlining relevant interventions and their timing. A life-long coordinated and multidisciplinary approach is recommended, starting as far as possible in early infancy and continuing throughout adulthood with an appropriate and timely transition from pediatric to adult care.
Collapse
Affiliation(s)
- Giovanna Mantovani
- Endocrinology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Murat Bastepe
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Monk
- Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Luisa de Sanctis
- Pediatric Endocrinology Unit, Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
| | - Susanne Thiele
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - S. Faisal Ahmed
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Roberto Bufo
- Italian Progressive Osseous Heteroplasia Association (IPOHA), Foggia, Italy
| | - Timothée Choplin
- K20, French PHP and Related Disorders Patient Association, Jouars-Pontchartrain, France
| | - Gianpaolo De Filippo
- APHP, Department of Medicine for Adolescents, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicetre, France
| | - Guillemette Devernois
- K20, French PHP and Related Disorders Patient Association, Jouars-Pontchartrain, France
| | - Thomas Eggermann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Francesca M. Elli
- Endocrinology Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Emily L. Germain-Lee
- Albright Center and Center for Rare Bone Disorders, Division of Pediatric Endocrinology and Diabetes, Connecticut Children’s Medical Center, Farmington, CT, USA,Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lionel Groussin
- APHP, Department of Endocrinology, Cochin Hospital (HUPC), Paris, France,University of Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Neveen A.T. Hamdy
- Division of Endocrinology and Centre for Bone Quality, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick Hanna
- INSERM U1185, Bicêtre Paris Sud – Paris Saclay University, Le Kremlin-Bicêtre, France
| | - Olaf Hiort
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Harald Jüppner
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Kamenický
- INSERM U1185, Bicêtre Paris Sud – Paris Saclay University, Le Kremlin-Bicêtre, France,APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Saclay for Rare Diseases and Filière OSCAR, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France,APHP, Department of Endocrinology and Reproductive Diseases, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - Nina Knight
- Acrodysostosis Support and Research patients’ group, London, UK
| | - Elvire Le Norcy
- University of Paris Descartes, Sorbonne Paris Cité, Paris, France,APHP, Department of Odontology, Bretonneau Hospital (PNVS), Paris, France
| | - Beatriz Lecumberri
- Department of Endocrinology and Nutrition, La Paz University Hospital, Madrid, Spain,Department of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Michael A. Levine
- Division of Endocrinology and Diabetes and Center for Bone Health, Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Outi Mäkitie
- Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Regina Martin
- Osteometabolic Disorders Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Hospital das Clínicas HCFMUSP, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Gabriel Ángel Martos-Moreno
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Hospital La Princesa Institute for Health Research (IIS La Princesa), Madrid, Spain,Department of Pediatrics, Autonomous University of Madrid (UAM), Madrid, Spain,CIBERobn, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Philip Murray
- Department of Paediatric Endocrinology, Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Arrate Pereda
- Molecular (Epi)Genetics Laboratory, BioAraba Research Health Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Spain
| | | | - Lars Rejnmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Rebeca Rodado
- AEPHP, Spanish PHP and Related Disorders Patient Association, Almeria, Spain
| | - Anya Rothenbuhler
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Saclay for Rare Diseases and Filière OSCAR, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France,APHP, Endocrinology and Diabetes for Children, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - Vrinda Saraff
- Department of Endocrinology and Diabetes, Birmingham Children’s Hospital, Birmingham, UK
| | - Ashley H. Shoemaker
- Pediatric Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eileen M. Shore
- Departments of Orthopedic Surgery and Genetics, Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Silve
- APHP, Service de Biochimie et Génétique Moléculaires, Hôpital Cochin, Paris, France
| | - Serap Turan
- Department of Pediatrics, Division of Endocrinology and Diabetes, Marmara University, Istanbul, Turkey
| | - Philip Woods
- Acrodysostosis Support and Research patients’ group, London, UK
| | - M. Carola Zillikens
- Department of Internal Medicine, Bone Center Erasmus MC – University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Guiomar Perez de Nanclares
- Molecular (Epi)Genetics Laboratory, BioAraba Research Health Institute, Araba University Hospital-Txagorritxu, Vitoria-Gasteiz, Spain
| | - Agnès Linglart
- INSERM U1185, Bicêtre Paris Sud – Paris Saclay University, Le Kremlin-Bicêtre, France,APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Saclay for Rare Diseases and Filière OSCAR, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France,APHP, Endocrinology and Diabetes for Children, Bicêtre Paris Saclay Hospital (HUPS), Le Kremlin-Bicêtre, France
| |
Collapse
|
38
|
Jodoin M, Rouleau DM, Therrien E, Chauny JM, Sandman E, Larson-Dupuis C, Leduc S, Gosselin N, De Beaumont L. Investigating the incidence and magnitude of heterotopic ossification with and without joints involvement in patients with a limb fracture and mild traumatic brain injury. Bone Rep 2019; 11:100222. [PMID: 31463337 PMCID: PMC6706636 DOI: 10.1016/j.bonr.2019.100222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/25/2019] [Accepted: 08/12/2019] [Indexed: 01/01/2023] Open
Abstract
Objectives This study seeks to evaluate the incidence rate of heterotopic ossification (HO) formation in patients afflicted by an isolated limb fracture (ILF) and a concomitant mild traumatic brain injury (mTBI). Methods The current study is an observational study including ILF patients with or without a concomitant mTBI recruited from an orthopedic clinic of a Level 1 Trauma Hospital. Patients were diagnosed with a mTBI according to the American Congress of Rehabilitation Medicine (ACRM) criteria. Radiographs taken on average 3 months post-trauma were analyzed separately by two distinct specialists for the presence of HO proximally to the fracture site (joints or extra joints). Both raters referred to Brooker's and Della's Valle's classification to establish signs of HO. First, analyses were conducted for the full sample. Secondly, a matched cohort was used in order to control for specific factors, namely age, sex, type of injury, and time elapsed between the accident and the analyzed radiograph. Results The full sample included a total of 183 patients with an ILF (94 females; 47.5 years old), of which 50 had a concomitant mTBI and 133 without. Radiographic evidence of HO was significantly higher in patients with an ILF and a mTBI compared to ILF patients (X2 = 6.50; p = 0.01). The matched cohort consisted of 94 participants (i.e.; 47 patients from the ILF + mTBI group and 47 patients from the ILF group). Again, ILF + mTBI patients presented significantly higher rates of HO signs in comparison to ILF patients (X2 = 3.69; p = 0.04). Presence of HO was associated with prolonged delays to return to work (RTW) only in ILF + mTBI patients (F = 4.055; p = 0.05) but not in ILF patients (F = 0.823; p = 0.37). Conclusions Study findings suggest that rates of HO are significantly higher proximally to fracture sites when ILF patients sustain a concomitant mTBI, even after controlling for factors known to influence HO. Moreover, results show that HO is associated with a prolonged RTW only in ILF patients with a concomitant mTBI but not in ILF-only patients. The impact of mTBI on HO formation warrants further attention to detect early signs of HO, to identify shared physiopathological mechanisms and, ultimately, to design targeted therapies. Rates of HO are significantly higher in patients with a fracture and a mTBI compared to patients with a fracture only. Factors such as sex, age, joint involvement, and surgical procedures were unrelated to the detection of signs of HO Presence of HO negatively impacted RTW delays in patients with a concomitant mTBI
Collapse
Affiliation(s)
- Marianne Jodoin
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Départment de Psychologie, Université de Montréal, Montréal, Québec, Canada
| | - Dominique M Rouleau
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Départment de Chirurgie, Université de Montréal, Montréal, Québec, Canada
| | - Erik Therrien
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Départment de Chirurgie, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Marc Chauny
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - Emilie Sandman
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Départment de Chirurgie, Université de Montréal, Montréal, Québec, Canada
| | - Camille Larson-Dupuis
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Départment de Psychologie, Université de Montréal, Montréal, Québec, Canada
| | - Stephane Leduc
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Départment de Chirurgie, Université de Montréal, Montréal, Québec, Canada
| | - Nadia Gosselin
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Départment de Psychologie, Université de Montréal, Montréal, Québec, Canada
| | - Louis De Beaumont
- Centre de Recherche de l'Hôpital Sacré-Coeur de Montréal, Montréal, Québec, Canada.,Départment de Chirurgie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
39
|
Ware AD, Brewer N, Meyers C, Morris C, McCarthy E, Shore EM, James AW. Differential Vascularity in Genetic and Nonhereditary Heterotopic Ossification. Int J Surg Pathol 2019; 27:859-867. [PMID: 31250694 DOI: 10.1177/1066896919857135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction. Nonhereditary heterotopic ossification (NHO) is a common complication of trauma. Progressive osseous heteroplasia (POH) and fibrodysplasia ossificans progressiva (FOP) are rare genetic causes of heterotopic bone. In this article, we detail the vascular patterning associated with genetic versus NHO. Methods. Vascular histomorphometric analysis was performed on patient samples from POH, FOP, and NHO. Endpoints for analysis included blood vessel (BV) number, area, density, size, and wall thickness. Results. Results demonstrated conserved temporal dynamic changes in vascularity across all heterotopic ossification lesions. Immature areas had the highest BV number, while the more mature foci had the highest BV area. Most vascular parameters were significantly increased in genetic as compared with NHO. Discussion. In sum, both genetic and NHO show temporospatial variation in vascularity. These findings suggest that angiogenic pathways are potential therapeutic targets in both genetic and nonhereditary forms of heterotopic ossification.
Collapse
|
40
|
Meyers C, Lisiecki J, Miller S, Levin A, Fayad L, Ding C, Sono T, McCarthy E, Levi B, James AW. Heterotopic Ossification: A Comprehensive Review. JBMR Plus 2019; 3:e10172. [PMID: 31044187 PMCID: PMC6478587 DOI: 10.1002/jbm4.10172] [Citation(s) in RCA: 292] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/31/2018] [Accepted: 01/13/2019] [Indexed: 12/17/2022] Open
Abstract
Heterotopic ossification (HO) is a diverse pathologic process, defined as the formation of extraskeletal bone in muscle and soft tissues. HO can be conceptualized as a tissue repair process gone awry and is a common complication of trauma and surgery. This comprehensive review seeks to synthesize the clinical, pathoetiologic, and basic biologic features of HO, including nongenetic and genetic forms. First, the clinical features, radiographic appearance, histopathologic diagnosis, and current methods of treatment are discussed. Next, current concepts regarding the mechanistic bases for HO are discussed, including the putative cell types responsible for HO formation, the inflammatory milieu and other prerequisite “niche” factors for HO initiation and propagation, and currently available animal models for the study of HO of this common and potentially devastating condition. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Carolyn Meyers
- Department of Pathology Johns Hopkins University Baltimore MD USA
| | | | - Sarah Miller
- Department of Pathology Johns Hopkins University Baltimore MD USA
| | - Adam Levin
- Department of Orthopaedic Surgery Johns Hopkins University Baltimore MD USA
| | - Laura Fayad
- Department of Radiology Johns Hopkins University Baltimore MD USA
| | - Catherine Ding
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center Los Angeles CA USA
| | - Takashi Sono
- Department of Pathology Johns Hopkins University Baltimore MD USA
| | - Edward McCarthy
- Department of Pathology Johns Hopkins University Baltimore MD USA
| | - Benjamin Levi
- Department of Surgery University of Michigan Ann Arbor MI USA
| | - Aaron W James
- Department of Pathology Johns Hopkins University Baltimore MD USA.,UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center Los Angeles CA USA
| |
Collapse
|
41
|
Pignolo RJ, Wang H, Kaplan FS. Fibrodysplasia Ossificans Progressiva (FOP): A Segmental Progeroid Syndrome. Front Endocrinol (Lausanne) 2019; 10:908. [PMID: 31998237 PMCID: PMC6966325 DOI: 10.3389/fendo.2019.00908] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/12/2019] [Indexed: 12/16/2022] Open
Abstract
Segmental progeroid syndromes are commonly represented by genetic conditions which recapitulate aspects of physiological aging by similar, disparate, or unknown mechanisms. Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease caused by mutations in the gene for ACVR1/ALK2 encoding Activin A receptor type I/Activin-like kinase 2, a bone morphogenetic protein (BMP) type I receptor, and results in the formation of extra-skeletal ossification and a constellation of others features, many of which resemble accelerated aging. The median estimated lifespan of individuals with FOP is approximately 56 years of age. Characteristics of precocious aging in FOP include both those that are related to dysregulated BMP signaling as well as those secondary to early immobilization. Progeroid features that may primarily be associated with mutations in ACVR1 include osteoarthritis, hearing loss, alopecia, subcutaneous lipodystrophy, myelination defects, heightened inflammation, menstrual abnormalities, and perhaps nephrolithiasis. Progeroid features that may secondarily be related to immobilization from progressive heterotopic ossification include decreased vital capacity, osteoporosis, fractures, sarcopenia, and predisposition to respiratory infections. Some manifestations of precocious aging may be attributed to both primary and secondary effects of FOP. At the level of lesion formation in FOP, soft tissue injury resulting in hypoxia, cell damage, and inflammation may lead to the accumulation of senescent cells as in aged tissue. Production of Activin A, platelet-derived growth factor, metalloproteinases, interleukin 6, and other inflammatory cytokines as part of the senescence-associated secretory phenotype could conceivably mediate the initial signaling cascade that results in the intense fibroproliferative response as well as the tissue-resident stem cell reprogramming leading up to ectopic endochondral bone formation. Consideration of FOP as a segmental progeroid syndrome offers a unique perspective into potential mechanisms of normal aging and may also provide insight for identification of new targets for therapeutic interventions in FOP.
Collapse
Affiliation(s)
- Robert J. Pignolo
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, United States
- Department of Physiology-Biomedical Engineering, Mayo Clinic Alix School of Medicine, Rochester, MN, United States
- Kogod Center on Aging, Mayo Clinic Alix School of Medicine, Rochester, MN, United States
- *Correspondence: Robert J. Pignolo
| | - Haitao Wang
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN, United States
- Department of Physiology-Biomedical Engineering, Mayo Clinic Alix School of Medicine, Rochester, MN, United States
- Kogod Center on Aging, Mayo Clinic Alix School of Medicine, Rochester, MN, United States
| | - Frederick S. Kaplan
- Department of Orthopaedic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Center for Research in FOP and Related Disorders, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
42
|
Barruet E, Morales BM, Cain CJ, Ton AN, Wentworth KL, Chan TV, Moody TA, Haks MC, Ottenhoff TH, Hellman J, Nakamura MC, Hsiao EC. NF-κB/MAPK activation underlies ACVR1-mediated inflammation in human heterotopic ossification. JCI Insight 2018; 3:122958. [PMID: 30429363 DOI: 10.1172/jci.insight.122958] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/11/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Inflammation helps regulate normal growth and tissue repair. Although bone morphogenetic proteins (BMPs) and inflammation are known contributors to abnormal bone formation, how these pathways interact in ossification remains unclear. METHODS We examined this potential link in patients with fibrodysplasia ossificans progressiva (FOP), a genetic condition of progressive heterotopic ossification caused by activating mutations in the Activin A type I receptor (ACVR1/ALK2). FOP patients show exquisite sensitivity to trauma, suggesting that BMP pathway activation may alter immune responses. We studied primary blood, monocyte, and macrophage samples from control and FOP subjects using multiplex cytokine, gene expression, and protein analyses; examined CD14+ primary monocyte and macrophage responses to TLR ligands; and assayed BMP, TGF-β activated kinase 1 (TAK1), and NF-κB pathways. RESULTS FOP subjects at baseline without clinically evident heterotopic ossification showed increased serum IL-3, IL-7, IL-8, and IL-10. CD14+ primary monocytes treated with the TLR4 activator LPS showed increased CCL5, CCR7, and CXCL10; abnormal cytokine/chemokine secretion; and prolonged activation of the NF-κB pathway. FOP macrophages derived from primary monocytes also showed abnormal cytokine/chemokine secretion, increased TGF-β production, and p38MAPK activation. Surprisingly, SMAD phosphorylation was not significantly changed in the FOP monocytes/macrophages. CONCLUSIONS Abnormal ACVR1 activity causes a proinflammatory state via increased NF-κB and p38MAPK activity. Similar changes may contribute to other types of heterotopic ossification, such as in scleroderma and dermatomyositis; after trauma; or with recombinant BMP-induced bone fusion. Our findings suggest that chronic antiinflammatory treatment may be useful for heterotopic ossification.
Collapse
Affiliation(s)
- Emilie Barruet
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, UCSF, San Francisco, California, USA
| | - Blanca M Morales
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, UCSF, San Francisco, California, USA
| | - Corey J Cain
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, UCSF, San Francisco, California, USA
| | - Amy N Ton
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, UCSF, San Francisco, California, USA
| | - Kelly L Wentworth
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, UCSF, San Francisco, California, USA
| | - Tea V Chan
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, UCSF, San Francisco, California, USA
| | - Tania A Moody
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, UCSF, San Francisco, California, USA
| | - Mariëlle C Haks
- Leiden University Medical Center, Department of Infectious Diseases, Leiden, Netherlands
| | - Tom Hm Ottenhoff
- Leiden University Medical Center, Department of Infectious Diseases, Leiden, Netherlands
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, UCSF, San Francisco, California, USA
| | - Mary C Nakamura
- Division of Rheumatology, Department of Medicine, San Francisco VA Health Care System, UCSF, San Francisco, California, USA
| | - Edward C Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, and the Institute for Human Genetics, UCSF, San Francisco, California, USA
| |
Collapse
|
43
|
Immunological and morphological analysis of heterotopic ossification differs to healthy controls. BMC Musculoskelet Disord 2018; 19:327. [PMID: 30205831 PMCID: PMC6134767 DOI: 10.1186/s12891-018-2246-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Background Formation of lamellar bone in non-osseus tissue is a pathological process called heterotopic ossification. It is the aim of this study to analyse the morphology and immunological status of patients with heterotopic ossification compared to individual healthy persons. Methods Human bone marrow and blood samples were obtained from 6 systemically healthy individuals and 4 patients during resection of heterotopic ossification from bone at hip arthroplasty. Bone was fragmented and treated with purified collagenase. Immunofluorescence surface staining was performed and analyzed with flow cytometry. Microcomputed tomography scanning was done performed at a resolution of 11 and 35 μm isometric voxel size respectively using a two different cone beam X-computer tomography systems and a microfocus X-ray tube. Subsequently the volume data was morphometrically analysed. Results The monocytes, stem cells, stroma cells and granulocytes progenitor cells were strongly reduced in the heterotopic ossification patient. Additionally a significant reduction of stromal stem cells cells and CD34 positive stem cells was observed. The frequency of NK-cells, B cells and T cells were not altered in the patients with heterotopic ossification compared to a healthy person. Micromorphometric parameters showed a lower content of mineralized bone tissue compared to normal bone. Mean trabecular thickness showed a high standard deviation, indicating a high variation in trabecular thickness, anisotropy and reducing bone strength. Conclusions This work shows altered immunological distribution that is accompanied by a low decrease in bone volume fraction and tissue mineral density in the heterotopic ossification sample compared to normal bone. Compared to healthy subjects, this might reflect an immunological participation in the development of this entity.
Collapse
|
44
|
Mantovani G, Bastepe M, Monk D, de Sanctis L, Thiele S, Usardi A, Ahmed SF, Bufo R, Choplin T, De Filippo G, Devernois G, Eggermann T, Elli FM, Freson K, García Ramirez A, Germain-Lee EL, Groussin L, Hamdy N, Hanna P, Hiort O, Jüppner H, Kamenický P, Knight N, Kottler ML, Le Norcy E, Lecumberri B, Levine MA, Mäkitie O, Martin R, Martos-Moreno GÁ, Minagawa M, Murray P, Pereda A, Pignolo R, Rejnmark L, Rodado R, Rothenbuhler A, Saraff V, Shoemaker AH, Shore EM, Silve C, Turan S, Woods P, Zillikens MC, Perez de Nanclares G, Linglart A. Diagnosis and management of pseudohypoparathyroidism and related disorders: first international Consensus Statement. Nat Rev Endocrinol 2018; 14:476-500. [PMID: 29959430 PMCID: PMC6541219 DOI: 10.1038/s41574-018-0042-0] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This Consensus Statement covers recommendations for the diagnosis and management of patients with pseudohypoparathyroidism (PHP) and related disorders, which comprise metabolic disorders characterized by physical findings that variably include short bones, short stature, a stocky build, early-onset obesity and ectopic ossifications, as well as endocrine defects that often include resistance to parathyroid hormone (PTH) and TSH. The presentation and severity of PHP and its related disorders vary between affected individuals with considerable clinical and molecular overlap between the different types. A specific diagnosis is often delayed owing to lack of recognition of the syndrome and associated features. The participants in this Consensus Statement agreed that the diagnosis of PHP should be based on major criteria, including resistance to PTH, ectopic ossifications, brachydactyly and early-onset obesity. The clinical and laboratory diagnosis should be confirmed by a molecular genetic analysis. Patients should be screened at diagnosis and during follow-up for specific features, such as PTH resistance, TSH resistance, growth hormone deficiency, hypogonadism, skeletal deformities, oral health, weight gain, glucose intolerance or type 2 diabetes mellitus, and hypertension, as well as subcutaneous and/or deeper ectopic ossifications and neurocognitive impairment. Overall, a coordinated and multidisciplinary approach from infancy through adulthood, including a transition programme, should help us to improve the care of patients affected by these disorders.
Collapse
Affiliation(s)
- Giovanna Mantovani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Murat Bastepe
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - David Monk
- Imprinting and Cancer Group, Cancer Epigenetic and Biology Program (PEBC), Institut d'Investigació Biomedica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Luisa de Sanctis
- Pediatric Endocrinology Unit, Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| | - Susanne Thiele
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Alessia Usardi
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Sud for Rare Diseases and Filière OSCAR, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
- APHP, Endocrinology and diabetes for children, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - S Faisal Ahmed
- Developmental Endocrinology Research Group, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Roberto Bufo
- IPOHA, Italian Progressive Osseous Heteroplasia Association, Cerignola, Foggia, Italy
| | - Timothée Choplin
- K20, French PHP and related disorders patient association, Jouars Pontchartrain, France
| | - Gianpaolo De Filippo
- APHP, Department of medicine for adolescents, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - Guillemette Devernois
- K20, French PHP and related disorders patient association, Jouars Pontchartrain, France
| | - Thomas Eggermann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Francesca M Elli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Endocrinology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Gasthuisberg, University of Leuven, Leuven, Belgium
| | - Aurora García Ramirez
- AEPHP, Spanish PHP and related disorders patient association, Huércal-Overa, Almería, Spain
| | - Emily L Germain-Lee
- Albright Center & Center for Rare Bone Disorders, Division of Pediatric Endocrinology & Diabetes, Connecticut Children's Medical Center, Farmington, CT, USA
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lionel Groussin
- APHP, Department of Endocrinology, Cochin Hospital (HUPC), Paris, France
- University of Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Neveen Hamdy
- Department of Medicine, Division of Endocrinology and Centre for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
| | - Patrick Hanna
- INSERM U1169, Bicêtre Paris Sud, Paris Sud - Paris Saclay University, Le Kremlin-Bicêtre, France
| | - Olaf Hiort
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Harald Jüppner
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Kamenický
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Sud for Rare Diseases and Filière OSCAR, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
- APHP, Department of Endocrinology and Reproductive Diseases, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
- INSERM U1185, Paris Sud - Paris Saclay University, Le Kremlin-Bicêtre, France
| | - Nina Knight
- UK acrodysostosis patients' group, London, UK
| | - Marie-Laure Kottler
- Department of Genetics, Reference Centre for Rare Disorders of Calcium and Phosphate Metabolism, Caen University Hospital, Caen, France
- BIOTARGEN, UNICAEN, Normandie University, Caen, France
| | - Elvire Le Norcy
- University of Paris Descartes, Sorbonne Paris Cité, Paris, France
- APHP, Department of Odontology, Bretonneau Hospital (PNVS), Paris, France
| | - Beatriz Lecumberri
- Department of Endocrinology and Nutrition, La Paz University Hospital, Madrid, Spain
- Department of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain
- Endocrine Diseases Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Michael A Levine
- Division of Endocrinology and Diabetes and Center for Bone Health, Children's Hospital of Philadelphia and Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Outi Mäkitie
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Regina Martin
- Osteometabolic Disorders Unit, Hormone and Molecular Genetics Laboratory (LIM/42), Endocrinology Division, Hospital das Clínicas HCFMUSP, Faculty of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Gabriel Ángel Martos-Moreno
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, CIBERobn, ISCIII, Madrid, Spain
- Department of Pediatrics, Autonomous University of Madrid (UAM), Madrid, Spain
- Endocrine Diseases Research Group, Hospital La Princesa Institute for Health Research (IIS La Princesa), Madrid, Spain
| | | | - Philip Murray
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Arrate Pereda
- Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, Hospital Universitario Araba-Txagorritxu, Vitoria-Gasteiz, Alava, Spain
| | | | - Lars Rejnmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Rebecca Rodado
- AEPHP, Spanish PHP and related disorders patient association, Huércal-Overa, Almería, Spain
| | - Anya Rothenbuhler
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Sud for Rare Diseases and Filière OSCAR, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
- APHP, Endocrinology and diabetes for children, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France
| | - Vrinda Saraff
- Department of Endocrinology and Diabetes, Birmingham Children's Hospital, Birmingham, UK
| | - Ashley H Shoemaker
- Pediatric Endocrinology and Diabetes, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eileen M Shore
- Departments of Orthopaedic Surgery and Genetics, Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Silve
- APHP, Service de Biochimie et Génétique Moléculaires, Hôpital Cochin, Paris, France
| | - Serap Turan
- Department of Pediatrics, Division of Endocrinology and Diabetes, Marmara University, Istanbul, Turkey
| | | | - M Carola Zillikens
- Department of Internal Medicine, Bone Center Erasmus MC - University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Guiomar Perez de Nanclares
- Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, Hospital Universitario Araba-Txagorritxu, Vitoria-Gasteiz, Alava, Spain.
| | - Agnès Linglart
- APHP, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Platform of Expertise Paris-Sud for Rare Diseases and Filière OSCAR, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France.
- APHP, Endocrinology and diabetes for children, Bicêtre Paris Sud Hospital (HUPS), Le Kremlin-Bicêtre, France.
- INSERM U1169, Bicêtre Paris Sud, Paris Sud - Paris Saclay University, Le Kremlin-Bicêtre, France.
| |
Collapse
|
45
|
Tuzmen C, Verdelis K, Weiss L, Campbell P. Crosstalk between substance P and calcitonin gene-related peptide during heterotopic ossification in murine Achilles tendon. J Orthop Res 2018; 36:1444-1455. [PMID: 29227562 PMCID: PMC6449576 DOI: 10.1002/jor.23833] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 12/04/2017] [Indexed: 02/04/2023]
Abstract
Heterotopic ossification (HO) is abnormal bone formation within soft tissue, usually predisposed by neurogenic or musculoskeletal trauma. Inflammation resulting from trauma is considered to be the main trigger for HO by eliciting changes within the injury site, including elevation of bone morphogenetic proteins (BMPs). Recent research, however, has also associated changes in sensory neuropeptide expression with HO. Substance P (SP) and calcitonin gene-related peptide (CGRP) are two of those neuropeptides that have been implicated with various aspects of HO, including regulation of inflammation and BMP signaling. Despite discoveries associating SP and CGRP with soft tissue HO, it remains unclear whether SP and CGRP have a direct role in the induction of HO. Here, we investigated the effect of SP and CGRP in vivo with the aid of inkjet-based biopatterning technology to controllably deliver these neuropeptides onto a murine Achilles tendon. While we did not observe any significant effect with CGRP, SP alone promoted HO in vivo with increased expression of BMP2. Remarkably, when SP and CGRP were delivered together, CGRP counteracted the effect of SP and essentially blocked SP-induced HO. This report contributes to the understanding of the complex problem of HO pathophysiology and warrants more study to better elucidate the interplay between SP and CGRP in the induction of HO. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1444-1455, 2018.
Collapse
Affiliation(s)
- Ceren Tuzmen
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Kostas Verdelis
- Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lee Weiss
- Robotics Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Phil Campbell
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania,Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania,Engineering Research Accelerator, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
46
|
Abstract
BACKGROUND Fibrodysplasia ossificans progressiva (FOP) causes progressive disability due to heterotopic ossification from episodic flare-ups. Using data from 500 FOP patients (representing 63% of all known patients world-wide), age- and joint-specific risks of new joint involvement were estimated using parametric and nonparametric statistical methods. RESULTS Compared to data from a 1994 survey of 44 individuals with FOP, our current estimates of age- and joint-specific risks of new joint involvement are more accurate (narrower confidence limits), based on a wider range of ages, and have less bias due to its greater comprehensiveness (captures over three-fifths of the known FOP patients worldwide). For the neck, chest, abdomen, and upper back, the estimated hazard decreases over time. For the jaw, lower back, shoulder, elbow, wrist, fingers, hip, knee, ankle, and foot, the estimated hazard increases initially then either plateaus or decreases. At any given time and for any anatomic site, the data indicate which joints are at risk. CONCLUSIONS This study of approximately 63% of the world's known population of FOP patients provides a refined estimate of risk for new involvement at any joint at any age, as well as the proportion of patients with uninvolved joints at any age. Importantly, these joint-specific survival curves can be used to facilitate clinical trial design and to determine if potential treatments can modify the predicted trajectory of progressive joint dysfunction.
Collapse
Affiliation(s)
- Robert J Pignolo
- Department of Medicine, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, United States.
| | - Blythe P Durbin-Johnson
- Department of Public Health Sciences, School of Medicine, Department of Biomedical Engineering, University of California, Davis, CA, United States.
| | - David M Rocke
- Department of Public Health Sciences, School of Medicine, Department of Biomedical Engineering, University of California, Davis, CA, United States.
| | - Frederick S Kaplan
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; The Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
47
|
Han HJ, Jain P, Resnick AC. Shared ACVR1 mutations in FOP and DIPG: Opportunities and challenges in extending biological and clinical implications across rare diseases. Bone 2018; 109:91-100. [PMID: 28780023 PMCID: PMC7888549 DOI: 10.1016/j.bone.2017.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Gain-of-function mutations in the Type I Bone Morphogenic Protein (BMP) receptor ACVR1 have been identified in two diseases: Fibrodysplasia Ossificans Progressiva (FOP), a rare autosomal dominant disorder characterized by genetically driven heterotopic ossification, and in 20-25% of Diffuse Intrinsic Pontine Gliomas (DIPGs), a pediatric brain tumor with no effective therapies and dismal median survival. While the ACVR1 mutation is causal for FOP, its role in DIPG tumor biology remains under active investigation. Here, we discuss cross-fertilization between the FOP and DIPG fields, focusing on the biological mechanisms and principles gleaned from FOP that can be applied to DIPG biology. We highlight our current knowledge of ACVR1 in both diseases, and then describe the growing opportunities and barriers to effectively investigate ACVR1 in DIPG. Importantly, learning from other seemingly unrelated diseases harboring similar mutations may uncover novel mechanisms or processes for future investigation.
Collapse
Affiliation(s)
- Harry J Han
- Division of Neurosurgery, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Room 4052, Philadelphia 19104, PA, United States
| | - Payal Jain
- Division of Neurosurgery, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Room 4052, Philadelphia 19104, PA, United States
| | - Adam C Resnick
- Division of Neurosurgery, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Colket Translational Research Building Room 4052, 3501 Civic Center Blvd, Philadelphia 19104, PA, United States; Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Room 4052, Philadelphia 19104, PA, United States.
| |
Collapse
|
48
|
Brady RD, Shultz SR, McDonald SJ, O'Brien TJ. Neurological heterotopic ossification: Current understanding and future directions. Bone 2018; 109:35-42. [PMID: 28526267 DOI: 10.1016/j.bone.2017.05.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 12/30/2022]
Abstract
Neurological heterotopic ossification (NHO) involves the formation of bone in soft tissue following a neurological condition, of which the most common are brain and spinal cord injuries. NHO often forms around the hip, knee and shoulder joints, causing severe pain and joint deformation which is associated with significant morbidity and reduced quality of life. The cellular and molecular events that initiate NHO have been the focus of an increasing number of human and animal studies over the past decade, with this work largely driven by the need to unearth potential therapeutic interventions to prevent the formation of NHO. This review provides an overview of the present understanding of NHO pathogenesis and pathobiology, current treatments, novel therapeutic targets, potential biomarkers and future directions.
Collapse
Affiliation(s)
- Rhys D Brady
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC, 3010, Australia.
| | - Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC, 3010, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, VIC, 3086, Australia
| | - Terence J O'Brien
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC, 3010, Australia
| |
Collapse
|
49
|
Wang H, Behrens EM, Pignolo RJ, Kaplan FS. ECSIT links TLR and BMP signaling in FOP connective tissue progenitor cells. Bone 2018; 109:201-209. [PMID: 29288875 DOI: 10.1016/j.bone.2017.12.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 12/26/2017] [Indexed: 01/12/2023]
Abstract
Clinical and laboratory observations strongly suggest that the innate immune system induces flare-ups in the setting of dysregulated bone morphogenetic protein (BMP) signaling in fibrodysplasia ossificans progressiva (FOP). In order to investigate the signaling substrates of this hypothesis, we examined toll-like receptor (TLR) activation and bone morphogenetic protein (BMP) signaling in connective tissue progenitor cells (CTPCs) from FOP patients and unaffected individuals. We found that inflammatory stimuli broadly activate TLR expression in FOP CTPCs and that TLR3/TLR4 signaling amplifies BMP pathway signaling through both ligand dependent and independent mechanisms. Importantly, Evolutionarily Conserved Signaling Intermediate in the Toll Pathway (ECSIT) integrates TLR injury signaling with dysregulated BMP pathway signaling in FOP CTPCs. These findings provide novel insight into the cell autonomous integration of injury signals from the innate immune system with dysregulated response signals from the BMP signaling pathway and provide new exploratory targets for therapeutic approaches to blocking the induction and amplification of FOP lesions.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Physiology and Biomedical Research, The Mayo Clinic, Rochester, MN, United States
| | - Edward M Behrens
- Department of Pediatrics, Division of Rheumatology, The Perelman School of Medicine of The University of Pennsylvania, Philadelphia, PA, United States
| | - Robert J Pignolo
- Department of Medicine, The Mayo Clinic, Rochester, MN, United States.
| | - Frederick S Kaplan
- Department of Orthopaedic Surgery, The Perelman School of Medicine of The University of Pennsylvania, Philadelphia, PA, United States; Department of Medicine, The Perelman School of Medicine of The University of Pennsylvania, Philadelphia, PA, United States; The Center for Research in FOP & Related Disorders, The Perelman School of Medicine of The University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
50
|
Le V, Anderson E, Akiyama T, Wharton KA. Drosophila models of FOP provide mechanistic insight. Bone 2018; 109:192-200. [PMID: 29128351 PMCID: PMC11957552 DOI: 10.1016/j.bone.2017.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 01/04/2023]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare bone disease characterized by episodic events of heterotopic ossification (HO). All cases of FOP have been attributed to mutations in the ACVR1 gene that render the encoded BMP type I ALK2 receptor hypersensitive, resulting in the activation of BMP signaling, at inappropriate times in inappropriate locations. The episodic or sporadic nature of HO associated with FOP rests with the occurrence of specific 'triggers' that push the hypersensitive ALK2-FOP receptor into full signaling mode. Identification of these triggers and their mechanism of action is critical for preventing HO and its devastating consequences in FOP patients. Models of FOP, generated in Drosophila, are shown to activate the highly conserved BMP signaling pathway in both Drosophila cell culture and in developing tissues in vivo. The most common FOP mutation, R206H, in ALK2 and its synonymous mutation, K262H, in the orthologous Drosophila receptor Sax, abolish the ability of wild type receptors to inhibit BMP ligand-induced signaling and lead to ubiquitous pathway activation in both cases but with important differences. When expressed in Drosophila, human ALK2R206H exhibits constitutive signaling. SaxK262H on the other hand can elicit excessive signaling similar to that observed for ALK2R206H in mammalian systems in vivo. For example, hyperactive signaling mediated by SaxK262H is triggered by an increase in ligand or in type II receptors. Interestingly, while the constitutive nature of ALK2R2026H in Drosophila requires activation by the type II receptor, it does not require its ligand binding domain. The differences exhibited by the two Drosophila FOP models enable a valuable comparative analysis poised to reveal critical regulatory mechanisms governing signaling output from these mutated receptors. Modifier screens using these Drosophila FOP models will be extremely valuable in identifying genes or compounds that reduce or prevent the hyperactive BMP signaling that initiates HO associated with FOP.
Collapse
Affiliation(s)
- Viet Le
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA; Program in Molecular Medicine, Boston Children's Hospital and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Edward Anderson
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
| | - Takuya Akiyama
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA; Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Kristi A Wharton
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|