1
|
Collins MD, Scott WJ. Thalidomide-induced limb malformations: an update and reevaluation. Arch Toxicol 2025; 99:1643-1747. [PMID: 40198353 DOI: 10.1007/s00204-024-03930-z] [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: 11/18/2024] [Accepted: 12/05/2024] [Indexed: 04/10/2025]
Abstract
Historically, thalidomide-induced congenital malformations have served as an important example of the enhanced susceptibility of developing embryos to chemical perturbation. The compound produced a wide variety of congenital malformations in humans, which were initially detected by an association with a relatively rare limb defect labeled phocomelia. Although true phocomelia in the most severe form is a transverse defect with intercalary absence of limb regions, it is proposed that thalidomide produces a longitudinal limb phenotype in humans under usual circumstances that can become transverse in severe cases with a preferential sensitivity of forelimb over hindlimb, preaxial over postaxial, and left more impacted than the corresponding non-autopod limb bones on the right. The thalidomide-induced limb phenotype in humans is described and followed by a hierarchical comparison with various laboratory animal species. Mechanistic studies have been hampered by the fact that only non-human primates and rabbits have malformations that are anatomically similar to humans. Included in this review are unpublished data on limb malformations produced by thalidomide in rhesus monkeys from experiments performed more than 50 years ago. The critical period in gestation for the induction of phocomelia may initiate prior to the development of the embryonic limb bud, which contrasts with other chemical and physical agents that are known to produce this phenotype. The importance of toxicokinetic parameters is reviewed including dose, enantiomers, absorption, distribution, and both non-enzymatic and enzymatic biotransformations. The limb embryopathy mechanism that provides a partial explanation of the limb phenotype is that cereblon binds to thalidomide creating a protein complex that ubiquitinates protein substrates (CRL4CRBN) that are not targets for the complex in the absence of the thalidomide. One of these neosubstrates is SALL4 which when mutated causes a syndrome that phenocopies aspects of thalidomide embryopathy. Other candidate neosubstrates for the complex that have been found in non-human species may contribute to an understanding of the limb defect including PLZF, p63, and various zinc finger transcription factors. It is proposed that it is important to consider the species-specificity of the compound when considering potential mechanistic pathways and that some of the more traditional mechanisms for explaining the embryopathy, such as anti-angiogenesis and redox perturbation, may contribute to a full understanding of this teratogen.
Collapse
Affiliation(s)
- Michael D Collins
- Department of Environmental Health Sciences and Molecular Toxicology Interdisciplinary Program, UCLA School of Public Health, CHS 46-078, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.
| | - William J Scott
- Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, OH, 45229, USA
| |
Collapse
|
2
|
Mee JA, Ly C, Pigott GC. Same trait, different genes: pelvic spine loss in three brook stickleback populations in Alberta, Canada. Evol Lett 2025; 9:115-124. [PMID: 39906589 PMCID: PMC11790222 DOI: 10.1093/evlett/qrae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/07/2024] [Accepted: 09/23/2024] [Indexed: 02/06/2025] Open
Abstract
The genetic basis of phenotypic or adaptive parallelism can reveal much about constraints on evolution. This study investigated the genetic basis of a canonically parallel trait: pelvic spine reduction in sticklebacks. Pelvic reduction has a highly parallel genetic basis in threespine stickleback in populations around the world, always involving a deletion of the pel1 enhancer of Pitx1. We conducted a genome-wide association study to investigate the genetic basis of pelvic spine reduction in 3 populations of brook stickleback in Alberta, Canada. Pelvic reduction did not involve Pitx1 in any of the 3 populations. Instead, pelvic reduction in 1 population involved a mutation in an exon of Tbx4, and it involved a mutation in an intron of Lmbr1 in the other two populations. Hence, the parallel phenotypic evolution of pelvic spine reduction across stickleback genera, and among brook stickleback populations, has a nonparallel genetic basis. This suggests that there may be redundancy in the genetic basis of this adaptive polymorphism, but it is not clear whether a lack of parallelism indicates a lack of constraint on the evolution of this adaptive trait. Whether different pleiotropic effects of different mutations have different fitness consequences or whether certain pelvic reduction mutations confer specific benefits in certain environments remains to be determined.
Collapse
Affiliation(s)
- Jonathan A Mee
- Department of Biology, Mount Royal University, Calgary, Alberta, Canada
| | - Carolyn Ly
- Department of Biology, Mount Royal University, Calgary, Alberta, Canada
| | - Grace C Pigott
- Department of Biology, Mount Royal University, Calgary, Alberta, Canada
| |
Collapse
|
3
|
Adhiyaman A, Tracey OC, Umesh A, Nian PP, Silverstein MK, Doyle SM, Scher DM. Prenatal Counseling for Congenital Clubfoot. JOURNAL OF THE PEDIATRIC ORTHOPAEDIC SOCIETY OF NORTH AMERICA 2024; 9:100130. [PMID: 40432674 PMCID: PMC12088293 DOI: 10.1016/j.jposna.2024.100130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/17/2024] [Accepted: 09/22/2024] [Indexed: 05/29/2025]
Abstract
Congenital clubfoot is a common deformity that affects 1 in 1000 newborns and is frequently detected prenatally during routine prenatal care. A wide variety of detection methods and testing are used to identify clubfoot and other congenital anomalies in the fetus, including complete ultrasonography, amniocentesis, chorionic villus sampling, or cell-free DNA. Newer studies have associated certain genome sequences to clubfoot specifically. It is important for orthopaedic surgeons to understand the implications of the various tests to provide the appropriate prenatal counseling. Early prenatal detection of clubfoot can help parents prepare for the demands of caring for a child with clubfoot and build trust between families and clinicians, ultimately leading to better, patient-centered care for their children. Key Concepts (1)Prompt recognition and treatment of congenital clubfoot is imperative to facilitate optimal treatment.(2)Prenatal diagnosis usually consists of sonography of the plantar surface of both feet at 13-16 weeks' gestation.(3)Additional post-natal testing may be indicated in patients with neurological impairment or syndromic features.(4)Genetic markers such as PITX1, RBM10, HOX, and CASP (among others) have been identified as involved in clubfoot development and have implications on prenatal testing and counseling.
Collapse
Affiliation(s)
| | | | - Amith Umesh
- Hospital for Special Surgery, New York, NY, USA
| | | | | | | | | |
Collapse
|
4
|
Doubková M, Knitlová J, Vondrášek D, Eckhardt A, Novotný T, Ošt’ádal M, Filová E, Bačáková L. Harnessing the Biomimetic Effect of Macromolecular Crowding in the Cell-Derived Model of Clubfoot Fibrosis. Biomacromolecules 2024; 25:6485-6502. [PMID: 39214607 PMCID: PMC11480992 DOI: 10.1021/acs.biomac.4c00653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Fibrotic changes in pediatric clubfoot provide an opportunity to improve corrective therapy and prevent relapses with targeted drugs. This study defines the parameters of clubfoot fibrosis and presents a unique analysis of a simple pseudo-3D in vitro model for disease-specific high-throughput drug screening experiments. The model combines clubfoot-derived fibroblasts with a biomimetic cultivation environment induced by the water-soluble polymers Ficoll and Polyvinylpyrrolidone, utilizing the principle of macromolecular crowding. We achieved higher conversion of soluble collagen into insoluble collagen, accelerated formation of the extracellular matrix layer and upregulated fibrosis-related genes in the mixed Ficoll environment. To test the model, we evaluated the effect of a potential antifibrotic drug, minoxidil, emphasizing collagen content and cross-linking. While the model amplified overall collagen deposition, minoxidil effectively blocked the expression of lysyl hydroxylases, which are responsible for the increased occurrence of specific collagen cross-linking in various fibrotic tissues. This limited the formation of collagen cross-link in both the model and control environments. Our findings provide a tool for expanding preclinical research for clubfoot and similar fibroproliferative conditions.
Collapse
Affiliation(s)
- Martina Doubková
- Laboratory
of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague 4, Czech Republic
- Second
Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic
| | - Jarmila Knitlová
- Laboratory
of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague 4, Czech Republic
- Faculty
of Science, Charles University, Albertov 6, 128 00 Prague 2, Czech Republic
| | - David Vondrášek
- Laboratory
of Biomathematics, Institute of Physiology
of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague 4, Czech Republic
| | - Adam Eckhardt
- Laboratory
of Translational Metabolism, Institute of
Physiology of the Czech Academy of Sciences, Videnska 1083, 142
00 Prague 4, Czech
Republic
| | - Tomáš Novotný
- Department
of Orthopaedics, Masaryk Hospital, Socialni Pece 3316/12A, 401 13 Usti nad Labem, Czech Republic
- Department
of Histology and Embryology, Second Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic
| | - Martin Ošt’ádal
- Department
of Orthopaedics, University Hospital Bulovka,
Charles University, Budinova
67/2, 180 81 Prague
8, Czech Republic
| | - Elena Filová
- Laboratory
of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague 4, Czech Republic
| | - Lucie Bačáková
- Laboratory
of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 00 Prague 4, Czech Republic
| |
Collapse
|
5
|
Jiratummarat P, Osateerakun P, Tooptakong T, Limpaphayom N. Comparison of kinematics and pedobarography findings between the unaffected foot of patients with unilateral clubfoot and controls. INTERNATIONAL ORTHOPAEDICS 2024; 48:2465-2473. [PMID: 38758492 DOI: 10.1007/s00264-024-06219-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
PURPOSE Idiopathic clubfoot (ICF) involves structural abnormalities in the lower extremities. Approximately half of patients have unilateral ICF, which could be due to differences in limb formation. The contralateral unaffected foot may have subclinical ICF. The objectives were to compare ankle and foot kinematics and pedobarography findings between the unaffected foot of patients with unilateral ICF and controls. METHODS Eleven children with unilateral ICF (11 unaffected feet, 11.7 ± 3.8 years) and 15 age-matched controls (30 control feet, 11.1 ± 3.0 years) were enroled. Five complete gait trials were performed. Data were collected using ten cameras and a two m long Footscan system and compared between groups using the Wilcoxon rank sum test. RESULTS All children with ICF underwent the selective soft tissue release procedure. The unaffected feet showed limited ranges in inversion-eversion and dorsiflexion-plantar flexion on kinematic analysis. There was a delay in landing time in all regions of the foot during heel rise and propulsion. The peak time was achieved significantly later in the unaffected feet compared to the controls. Although plantar pressure parameters were comparable, unaffected feet showed a larger contact area in the midfoot region. CONCLUSIONS Gait data on the unaffected foot in unilateral ICF patients do not correspond to those of controls despite an indistinguishable clinical appearance. This could be due to the effect of treatment, the process of foot development, or a combination. The relationship between genetics and gait deviation in patients with unilateral ICF needs further evaluation. The unaffected foot should receive similar attention during follow-up.
Collapse
Affiliation(s)
- Prajak Jiratummarat
- Department of Orthopaedics, Panyananthaphikkhu Chonprathan Medical Center, Srinakarinwirot University, Nonthaburi, 11120, Thailand
| | - Phatcharapa Osateerakun
- Department of Orthopaedics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Excellence Center for Gait and Motion, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Tanteera Tooptakong
- Bangbuathong Hospital, Ministry of Public Health, Nonthaburi, 11110, Thailand
| | - Noppachart Limpaphayom
- Department of Orthopaedics, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- Excellence Center for Gait and Motion, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, 10330, Thailand.
| |
Collapse
|
6
|
Hung TC, Kingsley DM, Boettiger AN. Boundary stacking interactions enable cross-TAD enhancer-promoter communication during limb development. Nat Genet 2024; 56:306-314. [PMID: 38238628 DOI: 10.1038/s41588-023-01641-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 12/06/2023] [Indexed: 02/15/2024]
Abstract
Although promoters and their enhancers are frequently contained within a topologically associating domain (TAD), some developmentally important genes have their promoter and enhancers within different TADs. Hypotheses about molecular mechanisms enabling cross-TAD interactions remain to be assessed. To test these hypotheses, we used optical reconstruction of chromatin architecture to characterize the conformations of the Pitx1 locus on single chromosomes in developing mouse limbs. Our data support a model in which neighboring boundaries are stacked as a result of loop extrusion, bringing boundary-proximal cis-elements into contact. This stacking interaction also contributes to the appearance of architectural stripes in the population average maps. Through molecular dynamics simulations, we found that increasing boundary strengths facilitates the formation of the stacked boundary conformation, counter-intuitively facilitating border bypass. This work provides a revised view of the TAD borders' function, both facilitating and preventing cis-regulatory interactions, and introduces a framework to distinguish border-crossing from border-respecting enhancer-promoter pairs.
Collapse
Affiliation(s)
- Tzu-Chiao Hung
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - David M Kingsley
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Alistair N Boettiger
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
7
|
Panza R, Albano F, Casto A, Del Vecchio C, Laforgia N, Dibello D. Incidence and prevalence of congenital clubfoot in Apulia: a regional model for future prospective national studies. Ital J Pediatr 2023; 49:151. [PMID: 37964341 PMCID: PMC10648723 DOI: 10.1186/s13052-023-01559-9] [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: 05/29/2023] [Accepted: 11/05/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Congenital clubfoot is a fairly common and severe congenital malformation, most often of idiopathic origin. A smaller percentage of cases is related to chromosomal abnormalities and genetic syndromes. It is estimated that 0.5/1000 newborns are affected worldwide, with a male to female ratio of 2:1 and greater distribution in developing countries (80%). The "European Surveillance of Congenital Anomalies (EUROCAT)" reported clubfoot prevalence in European newborns, but data regarding Italy are missing or poor. We aim to provide detailed data on clubfoot incidence according to the Apulian Regional Registry on Congenital Malformations and to report current knowledge on clubfoot genetic factors. METHODS We extrapolated data from the Regional Registry of Congenital Malformations to evaluate incidence and prevalence of congenital clubfoot in Apulia, Italy over a period of four years (2015-2018). We also performed a narrative review focusing on genetic mutations leading to congenital clubfoot. RESULTS Over the period from 2015 to 2018 in Apulia, Italy, 124,017 births were recorded and 209 cases of clubfoot were found, accounting for an incidence rate of 1.7/1,000 and a prevalence rate of 1.6/1,000. Six families of genes have been reported to have an etiopathogenetic role on congenital clubfoot. CONCLUSIONS Incidence and prevalence of congenital clubfoot in Apulia, Italy, are comparable with those reported in the other Italian regions but higher than those reported in previous studies from Europe. Genetic studies to better classify congenital clubfoot in either syndromic or isolated forms are desirable.
Collapse
Affiliation(s)
- Raffaella Panza
- Neonatology and Neonatal Intensive Care Unit (NICU), University of Bari Aldo Moro, Bari, Italy
| | - Federica Albano
- Orthopaedics Unit, Department of Basic Medical Science, Neuroscience and Sensory Organs, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Alberto Casto
- Orthopaedics Unit, Department of Basic Medical Science, Neuroscience and Sensory Organs, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Cosimo Del Vecchio
- Orthopaedics Unit, Department of Basic Medical Science, Neuroscience and Sensory Organs, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Nicola Laforgia
- Neonatology and Neonatal Intensive Care Unit (NICU), University of Bari Aldo Moro, Bari, Italy.
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy.
| | - Daniela Dibello
- Unit of Pediatric Orthopaedics and Traumatology, Giovanni XXIII Children's Hospital, Via Giovanni Amendola, Bari, 70126, Italy
| |
Collapse
|
8
|
Zhao J, Xu Y. PITX1 plays essential functions in cancer. Front Oncol 2023; 13:1253238. [PMID: 37841446 PMCID: PMC10570508 DOI: 10.3389/fonc.2023.1253238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
PITX1, also known as the pituitary homeobox 1 gene, has emerged as a key regulator in animal growth and development, attracting significant research attention. Recent investigations have revealed the implication of dysregulated PITX1 expression in tumorigenesis, highlighting its involvement in cancer development. Notably, PITX1 interacts with p53 and exerts control over crucial cellular processes including cell cycle progression, apoptosis, and chemotherapy resistance. Its influence extends to various tumors, such as esophageal, colorectal, gastric, and liver cancer, contributing to tumor progression and metastasis. Despite its significance, a comprehensive review examining PITX1's role in oncology remains lacking. This review aims to address this gap by providing a comprehensive overview of PITX1 in different cancer types, with a particular focus on its clinicopathological significance.
Collapse
Affiliation(s)
- Jingpu Zhao
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Yongfeng Xu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Sun D, Ding Z, Hai Y, Cheng Y. Advances in epigenetic research of adolescent idiopathic scoliosis and congenital scoliosis. Front Genet 2023; 14:1211376. [PMID: 37564871 PMCID: PMC10411889 DOI: 10.3389/fgene.2023.1211376] [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: 04/24/2023] [Accepted: 06/30/2023] [Indexed: 08/12/2023] Open
Abstract
Scoliosis is a three-dimensional structural deformity of the spine; more than 80% of scoliosis has no specific pathogenesis but is understood to be closely related to genetic, hormonal, and environmental factors. In recent years, the epigenetic alterations observed in scoliosis have been analyzed in numerous studies to determine the pathogenesis and progression of this condition, however, there is currently no comprehensive review of the epigenetic factors to date. We searched PubMed, Embase, and Web of Science databases for relative studies without language and date restrictions in March 2023. Twenty-five studies were included in this review and analyzed from the four main aspects of epigenetic alteration: DNA methylation, non-coding RNAs, histone modifications, and chromatin remodeling. The relationship between DNA methylation, non-coding RNAs, and scoliosis was considerably reported in the literature, and the corresponding related signaling pathways and novel biomarkers observed in scoliosis provide insights into innovative prevention and treatment strategies. However, the role of histone modifications is rarely reported in scoliosis, and few studies have investigated the relationship between scoliosis and chromatin remodeling. Therefore, these related fields need to be further explored to elucidate the overall effects of epigenetics in scoliosis.
Collapse
Affiliation(s)
| | | | - Yong Hai
- Department of Orthopedic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | | |
Collapse
|
10
|
Dap M, Harter H, Lambert L, Perdriolle-Galet E, Bonnet C, Morel O. Genetic studies in isolated bilateral clubfoot detected by prenatal ultrasound. J Matern Fetal Neonatal Med 2022; 35:10384-10387. [PMID: 36167341 DOI: 10.1080/14767058.2022.2128654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To evaluate the contribution of genetic investigations in case of isolated bilateral clubfoot detected by routine prenatal ultrasound. Pathogenic Copy Number Variations is about 3.9% in fetuses with isolated clubfoot (uni- or bilateral). We hypothesize that this rate could be higher in a homogenous group of fetuses with bilateral clubfoot. METHODS This retrospective single-center study included all women referred to our fetal-medicine center between 2013 and 2020 after ultrasound detection of isolated bilateral clubfoot. Genetic counseling was offered in which the woman was offered an amniocentesis for CMA and targeted investigation for Prader-Willi Syndrome (PWS), Steinert's disease and Spinal Muscular Atrophy (SMA). RESULTS 34 women were referred, 18 of them consented to undergo genetic studies by amniocentesis (18/34; 52.9%). Pathogenic copy number variations (CNVs) were found in 2/18 (11.1%) of cases. One of these CNVs was directly linked to the clubfoot pathology (a deletion in 5q31.1 containing PITX1 gene). Four fetuses (4/18, 22.2%) had variants of unknown significance (VUS). No PWS, SMA or Steinert's disease was found. No case diagnosed with isolated clubfoot prenatally had additional anomalies postnatally. CONCLUSIONS In the case of bilateral isolated clubfoot detected at the antenatal ultrasound, invasive prenatal testing should be offered, and if accepted, a CMA should be done, as pathogenic variations were observed in up to 11.1% of women who got amniocentesis. The findings of this study do not support the systematic recommendation of molecular studies for PWS, SMA, Steinert's disease.
Collapse
Affiliation(s)
- Matthieu Dap
- Obstetrics and Fetal medicine Unit, CHRU of Nancy, Nancy, France.,Department of Fetopathology and Placental Pathology, CHRU of Nancy, Nancy, France
| | - Hélène Harter
- Obstetrics and Fetal medicine Unit, CHRU of Nancy, Nancy, France
| | | | | | - Céline Bonnet
- Department of Genetics, CHRU de Nancy, Vandoeuvre-lès-Nancy, Nancy, France
| | - Olivier Morel
- Obstetrics and Fetal medicine Unit, CHRU of Nancy, Nancy, France.,INSERM, Diagnostic and Interventional Adaptive Imaging, University of Lorraine, Nancy, France
| |
Collapse
|
11
|
Bianco AM, Ragusa G, Di Carlo V, Faletra F, Di Stazio M, Racano C, Trisolino G, Cappellani S, De Pellegrin M, d’Addetta I, Carluccio G, Monforte S, Andreacchio A, Dibello D, d’Adamo AP. What Is the Exact Contribution of PITX1 and TBX4 Genes in Clubfoot Development? An Italian Study. Genes (Basel) 2022; 13:1958. [PMID: 36360195 PMCID: PMC9690101 DOI: 10.3390/genes13111958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 09/13/2023] Open
Abstract
Congenital clubfoot is a common pediatric malformation that affects approximately 0.1% of all births. 80% of the cases appear isolated, while 20% can be secondary or associated with complex syndromes. To date, two genes that appear to play an important role are PTIX1 and TBX4, but their actual impact is still unclear. Our study aimed to evaluate the prevalence of pathogenic variants in PITX1 and TBX4 in Italian patients with idiopathic clubfoot. PITX1 and TBX4 genes were analyzed by sequence and SNP array in 162 patients. We detected only four nucleotide variants in TBX4, predicted to be benign or likely benign. CNV analysis did not reveal duplications or deletions involving both genes and intragenic structural variants. Our data proved that the idiopathic form of congenital clubfoot was rarely associated with mutations and CNVs on PITX1 and TBX4. Although in some patients, the disease was caused by mutations in both genes; they were responsible for only a tiny minority of cases, at least in the Italian population. It was not excluded that other genes belonging to the same TBX4-PITX1 axis were involved, even if genetic complexity at the origin of clubfoot required the involvement of other factors.
Collapse
Affiliation(s)
- Anna Monica Bianco
- Genetics Unit, Institute for Maternal and Child Health, IRCCS “Burlo Garofolo”, 34137 Trieste, Italy
| | - Giulia Ragusa
- Genetics Unit, Institute for Maternal and Child Health, IRCCS “Burlo Garofolo”, 34137 Trieste, Italy
| | - Valentina Di Carlo
- Unit of Paediatric Orthopaedic and Traumatology, Institute for Maternal and Child Health, IRCCS “Burlo Garofolo”, 34147 Trieste, Italy
| | - Flavio Faletra
- Genetics Unit, Institute for Maternal and Child Health, IRCCS “Burlo Garofolo”, 34137 Trieste, Italy
| | - Mariateresa Di Stazio
- Genetics Unit, Institute for Maternal and Child Health, IRCCS “Burlo Garofolo”, 34137 Trieste, Italy
| | - Costantina Racano
- Unit of Pediatric Orthopaedics and Traumatology, Istituto Ortopedico Rizzoli (IRCCS), 40136 Bologna, Italy
| | - Giovanni Trisolino
- Unit of Pediatric Orthopaedics and Traumatology, Istituto Ortopedico Rizzoli (IRCCS), 40136 Bologna, Italy
| | - Stefania Cappellani
- Genetics Unit, Institute for Maternal and Child Health, IRCCS “Burlo Garofolo”, 34137 Trieste, Italy
| | | | - Ignazio d’Addetta
- Unit of Pediatric Orthopaedics and Traumatology Giovanni XXIII Children’s Hospital, Via Giovanni Amendola, 70126 Bari, Italy
| | - Giuseppe Carluccio
- Unit of Pediatric Orthopaedics and Traumatology Giovanni XXIII Children’s Hospital, Via Giovanni Amendola, 70126 Bari, Italy
| | - Sergio Monforte
- Pediatric Orthopedic of Buzzi Children Hospital of Milano, 20154 Milan, Italy
| | - Antonio Andreacchio
- Pediatric Orthopedic of Buzzi Children Hospital of Milano, 20154 Milan, Italy
| | - Daniela Dibello
- Unit of Pediatric Orthopaedics and Traumatology Giovanni XXIII Children’s Hospital, Via Giovanni Amendola, 70126 Bari, Italy
| | - Adamo P. d’Adamo
- Genetics Unit, Institute for Maternal and Child Health, IRCCS “Burlo Garofolo”, 34137 Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy
| |
Collapse
|
12
|
Muhammad H, Haryana SM, Magetsari R, Karsten S, Saraswati PA. Genes on syndromic and idiopathic CTEV: A systematic review. INTERNATIONAL JOURNAL OF SURGERY OPEN 2022. [DOI: 10.1016/j.ijso.2022.100547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
13
|
The molecular genetics of human appendicular skeleton. Mol Genet Genomics 2022; 297:1195-1214. [PMID: 35907958 DOI: 10.1007/s00438-022-01930-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/09/2022] [Indexed: 10/16/2022]
Abstract
Disorders that result from de-arrangement of growth, development and/or differentiation of the appendages (limbs and digit) are collectively called as inherited abnormalities of human appendicular skeleton. The bones of appendicular skeleton have central role in locomotion and movement. The different types of appendicular skeletal abnormalities are well described in the report of "Nosology and Classification of Genetic skeletal disorders: 2019 Revision". In the current article, we intend to present the embryology, developmental pathways, disorders and the molecular genetics of the appendicular skeletal malformations. We mainly focused on the polydactyly, syndactyly, brachydactyly, split-hand-foot malformation and clubfoot disorders. To our knowledge, only nine genes of polydactyly, five genes of split-hand-foot malformation, nine genes for syndactyly, eight genes for brachydactyly and only single gene for clubfoot have been identified to be involved in disease pathophysiology. The current molecular genetic data will help life sciences researchers working on the rare skeletal disorders. Moreover, the aim of present systematic review is to gather the published knowledge on molecular genetics of appendicular skeleton, which would help in genetic counseling and molecular diagnosis.
Collapse
|
14
|
Quiggle A, Charng WL, Antunes L, Nikolov M, Bledsoe X, Hecht JT, Dobbs MB, Gurnett CA. Whole Exome Sequencing in Individuals with Idiopathic Clubfoot Reveals a Recurrent Filamin B (FLNB) Deletion. Clin Orthop Relat Res 2022; 480:421-430. [PMID: 34491919 PMCID: PMC8747482 DOI: 10.1097/corr.0000000000001957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/11/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Clubfoot, a congenital deformity that presents as a rigid, inward turning of the foot, affects approximately 1 in 1000 infants and occurs as an isolated birth defect in 80% of patients. Despite its high level of heritability, few causative genes have been identified, and mutations in known genes are only responsible for a small portion of clubfoot heritability. QUESTIONS/PURPOSES (1) Are any rare gene variants enriched (that is, shared) in unrelated patients with isolated clubfoot? (2) Are there other rare variants in the identified gene (Filamin B) in these patients with clubfoot? METHODS Whole-exome sequence data were generated from a discovery cohort of 183 unrelated probands with clubfoot and 2492 controls. Variants were filtered with minor allele frequency < 0.02 to identify rare variants as well as small insertions and deletions (indels) resulting in missense variants, nonsense or premature truncation, or in-frame deletions. A candidate deletion was then genotyped in another cohort of 974 unrelated patients with clubfoot (a replication cohort). Other rare variants in the candidate gene were also investigated. A segregation analysis was performed in multigenerational families of individuals with clubfoot to see if the genotypes segregate with phenotypes. Single-variant association analysis was performed using the Fisher two-tailed exact test (exact p values are presented to give an indication of the magnitude of the association). RESULTS There were no recurrent variants in the known genes causing clubfoot in this study. A three-base pair in-frame codon deletion of Filamin B (FLNB) (p.E1792del, rs1470699812) was identified in 1.6% (3 of 183) of probands with clubfoot in the discovery cohort compared with 0% of controls (0 of 2492) (odds ratio infinity (inf) [95% CI 5.64 to inf]; p = 3.18 x 10-5) and 0.0016% of gnomAD controls (2 of 125,709) (OR 1.01 x 103 [95% CI 117.42 to 1.64 x 104]; p = 3.13 x 10-8). By screening a replication cohort (n = 974 patients), we found two probands with the identical FLNB deletion. In total, the deletion was identified in 0.43% (5 of 1157) of probands with clubfoot compared with 0% of controls and 0.0016% of gnomAD controls (OR 268.5 [95% CI 43.68 to 2.88 x 103]; p = 1.43 x 10-9). The recurrent FLNB p.E1792del variant segregated with clubfoot, with incomplete penetrance in two families. Affected individuals were more likely to be male and have bilateral clubfoot. Although most patients had isolated clubfoot, features consistent with Larsen syndrome, including upper extremity abnormalities such as elbow and thumb hypermobility and wide, flat thumbs, were noted in affected members of one family. We identified 19 additional rare FLNB missense variants located throughout the gene in patients with clubfoot. One of these missense variants, FLNB p.G2397D, exhibited incomplete penetrance in one family. CONCLUSION A recurrent FLNB E1792 deletion was identified in 0.43% of 1157 isolated patients with clubfoot. Given the absence of any recurrent variants in our discovery phase (n = 183) for any of the known genes causing clubfoot, our findings support that novel and rare missense variants in FLNB in patients with clubfoot, although rare, may be among the most commonly known genetic causes of clubfoot. Patients with FLNB variants often have isolated clubfoot, but they and their family members may be at an increased risk of having additional clinical features consistent with Larsen syndrome. CLINICAL RELEVANCE Identification of FLNB variants may be useful for determining clubfoot recurrence risk and comorbidities.
Collapse
Affiliation(s)
- Ashley Quiggle
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Wu-Lin Charng
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Lilian Antunes
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Momchil Nikolov
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Xavier Bledsoe
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jacqueline T. Hecht
- Department of Pediatrics, McGovern Medical School and School of Dentistry, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | | |
Collapse
|
15
|
Wang N, Zhang J, Lv H, Liu Z. Regulation of COL1A2, AKT3 genes, and related signaling pathway in the pathology of congenital talipes equinovarus. Front Pediatr 2022; 10:890109. [PMID: 35935376 PMCID: PMC9355787 DOI: 10.3389/fped.2022.890109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital talipes equinovarus (CTEV) is one of the most common congenital limb defects in children, which is a multifactorial and complex disease that associates with many unknown genetic, social-demographic, and environmental risk factors. Emerging evidence proved that gene expression or mutation might play an important role in the occurrence and development of CTEV. However, the underlying reasons and involved mechanisms are still not clear. Herein, to probe the potential genes and related signaling pathways involved in CTEV, we first identified the differentially expressed genes (DEGs) by mRNA sequencing in pediatric patients with CTEV compared with normal children. The gene of COL1A2 was upregulated, and AKT3 was downregulated at the transcriptional level. Western blot and quantitative polymerase chain reaction (qRT-PCR) results also showed that the expression of COL1A2 in CTEV was enhanced, and the AKT3 was decreased. Furthermore, the COL1A2 Knock-in (+COL1A2) and AKT3 Knock-out (-AKT3) transgenic mice were used to verify the effects of these two genes in the CTEV, and the results of which showed that both COL1A2 and AKT3 were closely related to the CTEV. We also investigated the effect of the PI3K-AKT3 signaling pathway in CTEV by measuring the relative expression of several key genes using Western blot and qRT-PCR. In line with the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis data, the PI3K-AKT3 signaling pathway might play a potentially important role in the regulation of pathological changes of CTEV. This study will provide new ideas for the mechanism investigation and prenatal diagnosis of CTEV.
Collapse
Affiliation(s)
- Ningqing Wang
- Department of Orthopedics, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Jiangchao Zhang
- Department of Orthopedics, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Haixiang Lv
- Department of Orthopedics, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Zhenjiang Liu
- Department of Orthopedics, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
16
|
Cell-specific alterations in Pitx1 regulatory landscape activation caused by the loss of a single enhancer. Nat Commun 2021; 12:7235. [PMID: 34903763 PMCID: PMC8668926 DOI: 10.1038/s41467-021-27492-1] [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: 03/25/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
Developmental genes are frequently controlled by multiple enhancers sharing similar specificities. As a result, deletions of such regulatory elements have often failed to reveal their full function. Here, we use the Pitx1 testbed locus to characterize in detail the regulatory and cellular identity alterations following the deletion of one of its enhancers (Pen). By combining single cell transcriptomics and an in-embryo cell tracing approach, we observe an increased fraction of Pitx1 non/low-expressing cells and a decreased fraction of Pitx1 high-expressing cells. We find that the over-representation of Pitx1 non/low-expressing cells originates from a failure of the Pitx1 locus to coordinate enhancer activities and 3D chromatin changes. This locus mis-activation induces a localized heterochrony and a concurrent loss of irregular connective tissue, eventually leading to a clubfoot phenotype. This data suggests that, in some cases, redundant enhancers may be used to locally enforce a robust activation of their host regulatory landscapes.
Collapse
|
17
|
Upregulated Transcription Factor PITX1 Predicts Poor Prognosis in Kidney Renal Clear Cell Carcinoma-Based Bioinformatic Analysis and Experimental Verification. DISEASE MARKERS 2021; 2021:7694239. [PMID: 34868397 PMCID: PMC8633854 DOI: 10.1155/2021/7694239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/09/2021] [Indexed: 12/02/2022]
Abstract
Paired-like homeodomain transcription factor 1 (PITX1) is involved in numerous biological processes, including cell growth, progression, and invasion in various malignant tumors. Nevertheless, the relationship between PITX1 and kidney renal clear cell carcinoma (KIRC) remains unclear. The clinical role and functions of PITX1 were analyzed by integrating multiple open-access online datasets. Further experimental verification was performed via quantitative real-time PCR (qRT-PCR) to detect the expression of PITX1 in 10 pairs of KIRC tissues. Our results revealed that PITX1 mRNA was overexpressed in tumor tissues compared with normal tissues in the TCGA-KIRC database (p < 0.001) and numerous independent cohorts (p < 0.05). Further, high expression of PITX1 mRNA was detected in KIRC tissues compared with adjacent normal tissues in our center by qRT-PCR (N = 10, p < 0.05). Logistic regression analysis demonstrated that the PITX1 level was positively associated with KIRC patients, T and M stages, histologic grade, and pathologic stage (all p < 0.05). Survival analysis showed that upregulation of PITX1 mRNA was associated with poor overall survival (OS), disease-free survival (DFS), and disease-specific survival (DSS) (all p < 0.05). Univariate/multivariate Cox hazard regression analysis revealed that PITX1 was an independent risk factor for OS in patients with KIRC (HR = 1.998, p = 0.003). Accordingly, the time-independent receiver operating characteristic (ROC) curve confirmed that PITX1 had good predictive efficacy for OS and DSS. Meanwhile, a prediction model constructed by nomogram was used to predict the OS of KIRC patients, and the calibration plot indicated this model shows high accuracy. We also revealed some downstream target genes of PITX1-related signaling pathways. Our finding suggested that high PITX1 mRNA expression may act as an independent predictive factor of poor prognosis in patients with KIRC. The prognostic model based on the nomogram would be instrumental in evaluating the survival rate in KIRC patients.
Collapse
|
18
|
Hordyjewska-Kowalczyk E, Nowosad K, Jamsheer A, Tylzanowski P. Genotype-phenotype correlation in clubfoot (talipes equinovarus). J Med Genet 2021; 59:209-219. [PMID: 34782442 DOI: 10.1136/jmedgenet-2021-108040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022]
Abstract
Clubfoot (talipes equinovarus) is a congenital malformation affecting muscles, bones, connective tissue and vascular or neurological structures in limbs. It has a complex aetiology, both genetic and environmental. To date, the most important findings in clubfoot genetics involve PITX1 variants, which were linked to clubfoot phenotype in mice and humans. Additionally, copy number variations encompassing TBX4 or single nucleotide variants in HOXC11, the molecular targets of the PITX1 transcription factor, were linked to the clubfoot phenotype. In general, genes of cytoskeleton and muscle contractile apparatus, as well as components of the extracellular matrix and connective tissue, are frequently linked with clubfoot aetiology. Last but not least, an equally important element, that brings us closer to a better understanding of the clubfoot genotype/phenotype correlation, are studies on the two known animal models of clubfoot-the pma or EphA4 mice. This review will summarise the current state of knowledge of the molecular basis of this congenital malformation.
Collapse
Affiliation(s)
- Ewa Hordyjewska-Kowalczyk
- Department of Biomedical Sciences, Laboratory of Molecular Genetics, Medical University of Lublin, Lublin, Lubelskie, Poland
| | - Karol Nowosad
- Department of Biomedical Sciences, Laboratory of Molecular Genetics, Medical University of Lublin, Lublin, Lubelskie, Poland.,The Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Wielkopolskie, Poland
| | - Przemko Tylzanowski
- Department of Biomedical Sciences, Laboratory of Molecular Genetics, Medical University of Lublin, Lublin, Lubelskie, Poland .,Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, KU Leuven, Leuven, Flanders, Belgium
| |
Collapse
|
19
|
Swank S, Sanger TJ, Stuart YE. (Non)Parallel developmental mechanisms in vertebrate appendage reduction and loss. Ecol Evol 2021; 11:15484-15497. [PMID: 34824770 PMCID: PMC8601893 DOI: 10.1002/ece3.8226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/31/2021] [Accepted: 09/21/2021] [Indexed: 01/16/2023] Open
Abstract
Appendages have been reduced or lost hundreds of times during vertebrate evolution. This phenotypic convergence may be underlain by shared or different molecular mechanisms in distantly related vertebrate clades. To investigate, we reviewed the developmental and evolutionary literature of appendage reduction and loss in more than a dozen vertebrate genera from fish to mammals. We found that appendage reduction and loss was nearly always driven by modified gene expression as opposed to changes in coding sequences. Moreover, expression of the same genes was repeatedly modified across vertebrate taxa. However, the specific mechanisms by which expression was modified were rarely shared. The multiple routes to appendage reduction and loss suggest that adaptive loss of function phenotypes might arise routinely through changes in expression of key developmental genes.
Collapse
Affiliation(s)
- Samantha Swank
- Department of BiologyLoyola University ChicagoChicagoIllinoisUSA
| | - Thomas J. Sanger
- Department of BiologyLoyola University ChicagoChicagoIllinoisUSA
| | - Yoel E. Stuart
- Department of BiologyLoyola University ChicagoChicagoIllinoisUSA
| |
Collapse
|
20
|
Duboc V, Sulaiman FA, Feneck E, Kucharska A, Bell D, Holder-Espinasse M, Logan MPO. Tbx4 function during hindlimb development reveals a mechanism that explains the origins of proximal limb defects. Development 2021; 148:271903. [PMID: 34423345 PMCID: PMC8497778 DOI: 10.1242/dev.199580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/02/2021] [Indexed: 11/20/2022]
Abstract
We dissect genetically a gene regulatory network that involves the transcription factors Tbx4, Pitx1 and Isl1 acting cooperatively to establish the hindlimb bud, and identify key differences in the pathways that initiate formation of the hindlimb and forelimb. Using live image analysis of murine limb mesenchyme cells undergoing chondrogenesis in micromass culture, we distinguish a series of changes in cellular behaviours and cohesiveness that are required for chondrogenic precursors to undergo differentiation. Furthermore, we provide evidence that the proximal hindlimb defects observed in Tbx4 mutant mice result from a failure in the early differentiation step of chondroprogenitors into chondrocytes, providing an explanation for the origins of proximally biased limb defects.
Collapse
Affiliation(s)
- Veronique Duboc
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Fatima A Sulaiman
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Eleanor Feneck
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Anna Kucharska
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Donald Bell
- Light Microscopy, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | | - Malcolm P O Logan
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
21
|
Knitlova J, Doubkova M, Plencner M, Vondrasek D, Eckhardt A, Ostadal M, Musilkova J, Bacakova L, Novotny T. Minoxidil decreases collagen I deposition and tissue-like contraction in clubfoot-derived cells: a way to improve conservative treatment of relapsed clubfoot? Connect Tissue Res 2021; 62:554-569. [PMID: 32951485 DOI: 10.1080/03008207.2020.1816992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIM Clubfoot is a congenital deformity affecting the musculoskeletal system, resulting in contracted and stiff tissue in the medial part of the foot. Minoxidil (MXD) has an inhibitory effect on lysyl hydroxylase, which influences the quality of extracellular matrix crosslinking, and could therefore be used to reduce the stiffness and to improve the flexibility of the tissue. We assessed the in vitro antifibrotic effects of minoxidil on clubfoot-derived cells. METHODS Cell viability and proliferation were quantified by xCELLigence, MTS, and LIVE/DEAD assays. The amount of collagen I deposited into the extracellular matrix was quantified using immunofluorescence with subsequent image segmentation analysis, hydroxyproline assay, and Second Harmonic Generation imaging. Extracellular matrix contraction was studied in a 3D model of cell-populated collagen gel lattices. RESULTS MXD concentrations of 0.25, 0.5, and 0.75 mM inhibited the cell proliferation in a concentration-dependent manner without causing a cytotoxic effect. Exposure to ≥0.5 mM MXD resulted in a decrease in collagen type I accumulation after 8 and 21 days in culture. Changes in collagen fiber assembly were observed by immunofluorescence microscopy and nonlinear optical microscopy (second harmonic generation). MXD also inhibited the contraction of cell-populated collagen lattices (0.5 mM by 22%; 0.75 mM by 28%). CONCLUSIONS Minoxidil exerts an in vitro inhibitory effect on the cell proliferation, collagen accumulation, and extracellular matrix contraction processes that are associated with clubfoot fibrosis. This study provides important preliminary results demonstrating the potential relevance of MXD for adjuvant pharmacological therapy in standard treatment of relapsed clubfoot.
Collapse
Affiliation(s)
- Jarmila Knitlova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Doubkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Plencner
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - David Vondrasek
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Physical Education and Sport, Charles University, Prague, Czech Republic
| | - Adam Eckhardt
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Ostadal
- Department of Orthopaedics, First Faculty of Medicine, Charles University and Na Bulovce Hospital, Prague, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Novotny
- Second Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Orthopaedics, Masaryk Hospital, Usti Nad Labem, Czech Republic
| |
Collapse
|
22
|
Latypova X, Creadore SG, Dahan-Oliel N, Gustafson AG, Wei-Hung Hwang S, Bedard T, Shazand K, van Bosse HJP, Giampietro PF, Dieterich K. A Genomic Approach to Delineating the Occurrence of Scoliosis in Arthrogryposis Multiplex Congenita. Genes (Basel) 2021; 12:genes12071052. [PMID: 34356068 PMCID: PMC8305424 DOI: 10.3390/genes12071052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Arthrogryposis multiplex congenita (AMC) describes a group of conditions characterized by the presence of non-progressive congenital contractures in multiple body areas. Scoliosis, defined as a coronal plane spine curvature of ≥10 degrees as measured radiographically, has been reported to occur in approximately 20% of children with AMC. To identify genes that are associated with both scoliosis as a clinical outcome and AMC, we first queried the DECIPHER database for copy number variations (CNVs). Upon query, we identified only two patients with both AMC and scoliosis (AMC-SC). The first patient contained CNVs in three genes (FBN2, MGF10, and PITX1), while the second case had a CNV in ZC4H2. Looking into small variants, using a combination of Human Phenotype Ontogeny and literature searching, 908 genes linked with scoliosis and 444 genes linked with AMC were identified. From these lists, 227 genes were associated with AMC-SC. Ingenuity Pathway Analysis (IPA) was performed on the final gene list to gain insight into the functional interactions of genes and various categories. To summarize, this group of genes encompasses a diverse group of cellular functions including transcription regulation, transmembrane receptor, growth factor, and ion channels. These results provide a focal point for further research using genomics and animal models to facilitate the identification of prognostic factors and therapeutic targets for AMC.
Collapse
Affiliation(s)
- Xenia Latypova
- Grenoble Institut Neurosciences, Université Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, 38000 Grenoble, France;
| | | | - Noémi Dahan-Oliel
- Shriners Hospitals for Children, Montreal, QC H4A 0A9, Canada;
- School of Physical & Occupational Therapy, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada
| | | | - Steven Wei-Hung Hwang
- Shriners Hospitals for Children, Philadelphia, PA 19140, USA; (S.W.-H.H.); (H.J.P.v.B.)
| | - Tanya Bedard
- Alberta Congenital Anomalies Surveillance System, Alberta Health Services, Edmonton, AB T5J 3E4, Canada;
| | - Kamran Shazand
- Shriners Hospitals for Children Headquarters, Tampa, FL 33607, USA; (S.G.C.); (A.G.G.); (K.S.)
| | | | - Philip F. Giampietro
- Department of Pediatrics, University of Illinois-Chicago, Chicago, IL 60607, USA
- Correspondence: (P.F.G.); (K.D.)
| | - Klaus Dieterich
- Institut of Advanced Biosciences, Université Grenoble Alpes, Inserm, U1209, CHU Grenoble Alpes, 38000 Grenoble, France
- Correspondence: (P.F.G.); (K.D.)
| |
Collapse
|
23
|
Tran TQ, Kioussi C. Pitx genes in development and disease. Cell Mol Life Sci 2021; 78:4921-4938. [PMID: 33844046 PMCID: PMC11073205 DOI: 10.1007/s00018-021-03833-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/05/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
Homeobox genes encode sequence-specific transcription factors (SSTFs) that recognize specific DNA sequences and regulate organogenesis in all eukaryotes. They are essential in specifying spatial and temporal cell identity and as a result, their mutations often cause severe developmental defects. Pitx genes belong to the PRD class of the highly evolutionary conserved homeobox genes in all animals. Vertebrates possess three Pitx paralogs, Pitx1, Pitx2, and Pitx3 while non-vertebrates have only one Pitx gene. The ancient role of regulating left-right (LR) asymmetry is conserved while new functions emerge to afford more complex body plan and functionalities. In mouse, Pitx1 regulates hindlimb tissue patterning and pituitary development. Pitx2 is essential for the development of the oral cavity and abdominal wall while regulates the formation and symmetry of other organs including pituitary, heart, gut, lung among others by controlling growth control genes upon activation of the Wnt/ß-catenin signaling pathway. Pitx3 is essential for lens development and migration and survival of the dopaminergic neurons of the substantia nigra. Pitx gene mutations are linked to various congenital defects and cancers in humans. Pitx gene family has the potential to offer a new approach in regenerative medicine and aid in identifying new drug targets.
Collapse
Affiliation(s)
- Thai Q Tran
- Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA
| | - Chrissa Kioussi
- Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
24
|
Bioinformatics analysis of prognostic value of PITX1 gene in breast cancer. Biosci Rep 2020; 40:226181. [PMID: 32830857 PMCID: PMC7494990 DOI: 10.1042/bsr20202537] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Paired-like homeodomain transcription factor 1 (PITX1) participates in miscellaneous biological processes including cell growth, development, progression and invasion in various malignant tumors. However, the analysis of the association between PITX1 expression and the survival in breast cancer remains unclear. METHODS Clinical prognostic parameters and survival data related to PITX1 in breast cancer patients were performed using the bioinformatic analysis including Oncomine, Bc-GenExMiner v4.3, PrognoScan and UCSC Xena. RESULTS We found that PITX1 gene expression was significantly higher in different histological classification of breast cancer. The Scarff-Bloom-Richardson (SBR) grade, Nottingham prognostic index (NPI), estrogen receptor (ER) negative, epidermal growth factor receptor-2 (HER2) positive, lymph node positive, triple-negative status and basal-like status were positively correlated with PITX1 level, except for patients' age and the progesterone receptor (PR) status. We have found that the increased PITX1 expression correlated with worse relapse-free survival, disease specific survival and overall survival. PITX1 was positively correlated with metastatic relapse-free survival and distant metastasis-free survival. We also confirmed positive correlation between PITX1 and the nucleotide-binding oligomerization domain 2 (NOD2). CONCLUSION The lower expression of PITX1 was associated with better clinical prognostic parameters and clinical survival in breast cancer according to the bioinformatic analysis.
Collapse
|
25
|
Johnstone CP, Wang NB, Sevier SA, Galloway KE. Understanding and Engineering Chromatin as a Dynamical System across Length and Timescales. Cell Syst 2020; 11:424-448. [PMID: 33212016 DOI: 10.1016/j.cels.2020.09.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 12/20/2022]
Abstract
Connecting the molecular structure and function of chromatin across length and timescales remains a grand challenge to understanding and engineering cellular behaviors. Across five orders of magnitude, dynamic processes constantly reshape chromatin structures, driving spaciotemporal patterns of gene expression and cell fate. Through the interplay of structure and function, the genome operates as a highly dynamic feedback control system. Recent experimental techniques have provided increasingly detailed data that revise and augment the relatively static, hierarchical view of genomic architecture with an understanding of how dynamic processes drive organization. Here, we review how novel technologies from sequencing, imaging, and synthetic biology refine our understanding of chromatin structure and function and enable chromatin engineering. Finally, we discuss opportunities to use these tools to enhance understanding of the dynamic interrelationship of chromatin structure and function.
Collapse
Affiliation(s)
| | - Nathan B Wang
- Department of Chemical Engineering, MIT, 25 Ames St., Cambridge, MA 02139, USA
| | - Stuart A Sevier
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Kate E Galloway
- Department of Chemical Engineering, MIT, 25 Ames St., Cambridge, MA 02139, USA.
| |
Collapse
|
26
|
Khanshour AM, Kidane YH, Kozlitina J, Cornelia R, Rafipay A, De Mello V, Weston M, Paria N, Khalid A, Hecht JT, Dobbs MB, Richards BS, Vargesson N, Hamra FK, Wilson M, Wise C, Gurnett CA, Rios JJ. Genetic association and characterization of FSTL5 in isolated clubfoot. Hum Mol Genet 2020; 29:3717-3728. [PMID: 33105483 DOI: 10.1093/hmg/ddaa236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/28/2020] [Accepted: 10/14/2020] [Indexed: 01/01/2023] Open
Abstract
Talipes equinovarus (clubfoot, TEV) is a congenital rotational foot deformity occurring in 1 per 1000 births with increased prevalence in males compared with females. The genetic etiology of isolated clubfoot (iTEV) remains unclear. Using a genome-wide association study, we identified a locus within FSTL5, encoding follistatin-like 5, significantly associated with iTEV. FSTL5 is an uncharacterized gene whose potential role in embryonic and postnatal development was previously unstudied. Utilizing multiple model systems, we found that Fstl5 was expressed during later stages of embryonic hindlimb development, and, in mice, expression was restricted to the condensing cartilage anlage destined to form the limb skeleton. In the postnatal growth plate, Fstl5 was specifically expressed in prehypertrophic chondrocytes. As Fstl5 knockout rats displayed no gross malformations, we engineered a conditional transgenic mouse line (Fstl5LSL) to overexpress Fstl5 in skeletal osteochondroprogenitors. We observed that hindlimbs were slightly shorter and that bone mineral density was reduced in adult male, but not female, Prrx1-cre;Fstl5LSL mice compared with control. No overt clubfoot-like deformity was observed in Prrx1-cre;Fstl5LSL mice, suggesting FSTL5 may function in other cell types to contribute to iTEV pathogenesis. Interrogating published mouse embryonic single-cell expression data showed that Fstl5 was expressed in cell lineage subclusters whose transcriptomes were associated with neural system development. Moreover, our results suggest that lineage-specific expression of the Fstl genes correlates with their divergent roles as modulators of transforming growth factor beta and bone morphogenetic protein signaling. Results from this study associate FSTL5 with iTEV and suggest a potential sexually dimorphic role for Fstl5 in vivo.
Collapse
Affiliation(s)
- Anas M Khanshour
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX 75219, USA
| | - Yared H Kidane
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX 75219, USA
| | - Julia Kozlitina
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Reuel Cornelia
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX 75219, USA
| | - Alexandra Rafipay
- School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Vanessa De Mello
- School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Mitchell Weston
- Department of Anatomy, University of Otago, Dunedin 9016, New Zealand
| | - Nandina Paria
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX 75219, USA
| | - Aysha Khalid
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX 75219, USA
| | - Jacqueline T Hecht
- Department of Pediatrics, McGovern Medical School, University of Texas Health, Houston, TX 77030, USA
| | - Matthew B Dobbs
- Paley Orthopedic and Spine Institute, West Palm Beach, FL 33407, USA
| | - B Stephens Richards
- Department of Orthopaedics, Scottish Rite for Children, Dallas, TX 75219, USA.,Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Neil Vargesson
- School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - F Kent Hamra
- Department of Obstetrics and Gynecology, Cecil H. & Ida Green Center for Reproductive Biology Sciences, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Megan Wilson
- Department of Anatomy, University of Otago, Dunedin 9016, New Zealand
| | - Carol Wise
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX 75219, USA.,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christina A Gurnett
- Department of Neurology, School of Medicine, Washington University, St. Louis, MO 63130, USA
| | - Jonathan J Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX 75219, USA.,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
27
|
Ding J, Liang Z, Feng W, Cai Q, Zhang Z. Integrated Bioinformatics Analysis Reveals Potential Pathway Biomarkers and Their Interactions for Clubfoot. Med Sci Monit 2020; 26:e925249. [PMID: 32829375 PMCID: PMC7462570 DOI: 10.12659/msm.925249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Congenital talipes equinovarus (clubfoot), one of the most regular pediatric congenital skeletal anomalies, seriously affects the normal growth and development of about 1 in 1000 newborns. Although it has been investigated widely, the etiology and pathogenesis of clubfoot are still controversial. Material/Methods g: Profiler, NetworkAnalyst and WebGestalt were used to probe the enriched signaling pathways by using the Gene Ontology (GO), Human Phenotype Ontology (HP), Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome (REAC), and WikiPathways (WP) databases. Large numbers of enriched signaling pathways were identified using the integrated bioinformatics enrichment analyses. Results Apoptosis or programmed cell death (PCD), disease, muscle contraction, metabolism, and immune system were the top functions. Embryo or organ morphogenesis and development, cell or muscle contraction, and apoptosis were the top biological processes, and cell/muscle contraction and apoptosis were the top molecular functions using enriched GO terms analysis. There were a large number of complex interactions in the genes, enriched pathways, and transcription factor (TF)-miRNA co-regulatory networks. Transcription factors such as FOXN3, GLI3, HOX, and NCOR2 family regulated the gene expression of APAF1, BCL2, BID, CASP, MTHFR, and TPM family. Conclusions The results of bioinformatics enrichment analysis not only supported the previously proposed hypotheses, e.g., extracellular matrix (ECM) abnormality, fetal movement reducing, genetic abnormality, muscle abnormality, neurological abnormality, skeletal abnormality and vascular abnormality, but also indicated that cellular or immune responses to external stimulus, molecular transport and metabolism may be new etiological mechanisms in clubfoot.
Collapse
Affiliation(s)
- Jing Ding
- Department of Pediatric Orthopaedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Zhenpeng Liang
- Department of Pediatric Orthopaedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Weijia Feng
- Department of Pediatric Orthopaedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Qixun Cai
- Department of Pediatric Orthopaedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Ziming Zhang
- Department of Pediatric Orthopaedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
28
|
Morel G, Duhamel C, Boussion S, Frénois F, Lesca G, Chatron N, Labalme A, Sanlaville D, Edery P, Thevenon J, Faivre L, Fassier A, Prodhomme O, Escande F, Manouvrier S, Petit F, Geneviève D, Rossi M. Mandibular-pelvic-patellar syndrome is a novel PITX1-related disorder due to alteration of PITX1 transactivation ability. Hum Mutat 2020; 41:1499-1506. [PMID: 32598510 DOI: 10.1002/humu.24070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/24/2020] [Accepted: 05/15/2020] [Indexed: 02/03/2023]
Abstract
PITX1 is a homeobox transcription factor essential for hindlimb morphogenesis. Two PITX1-related human disorders have been reported to date: PITX1 ectopic expression causes Liebenberg syndrome, characterized by malformation of upper limbs showing a "lower limb" appearance; PITX1 deletions or missense variation cause a syndromic picture including clubfoot, tibial hemimelia, and preaxial polydactyly. We report two novel PITX1 missense variants, altering PITX1 transactivation ability, in three individuals from two unrelated families showing a distinct recognizable autosomal dominant syndrome, including first branchial arch, pelvic, patellar, and male genital abnormalities. This syndrome shows striking similarities with the Pitx1-/- mouse model. A partial phenotypic overlap is also observed with Ischiocoxopodopatellar syndrome caused by TBX4 haploinsufficiency, and with the phenotypic spectrum caused by SOX9 anomalies, both genes being PITX1 downstream targets. Our study findings expand the spectrum of PITX1-related disorders and suggest a common pattern of developmental abnormalities in disorders of the PITX1-TBX4-SOX9 signaling pathway.
Collapse
Affiliation(s)
- Godelieve Morel
- Service de Génétique, Centre de compétences Anomalies du Développement, CHU de Nice, Nice, France.,Service de Génétique, Centre de Référence Anomalies du Développement et Centre de Compétences Maladies Osseuses Constitutionnelles, Hospices Civils de Lyon, Bron, France
| | | | | | | | - Gaetan Lesca
- Service de Génétique, Centre de Référence Anomalies du Développement et Centre de Compétences Maladies Osseuses Constitutionnelles, Hospices Civils de Lyon, Bron, France.,INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Bron, France
| | - Nicolas Chatron
- Service de Génétique, Centre de Référence Anomalies du Développement et Centre de Compétences Maladies Osseuses Constitutionnelles, Hospices Civils de Lyon, Bron, France.,INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Bron, France
| | - Audrey Labalme
- Service de Génétique, Centre de Référence Anomalies du Développement et Centre de Compétences Maladies Osseuses Constitutionnelles, Hospices Civils de Lyon, Bron, France
| | - Damien Sanlaville
- Service de Génétique, Centre de Référence Anomalies du Développement et Centre de Compétences Maladies Osseuses Constitutionnelles, Hospices Civils de Lyon, Bron, France.,INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Bron, France
| | - Patrick Edery
- Service de Génétique, Centre de Référence Anomalies du Développement et Centre de Compétences Maladies Osseuses Constitutionnelles, Hospices Civils de Lyon, Bron, France.,INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Bron, France
| | | | - Laurence Faivre
- Service de Génétique, Centre de Référence Anomalies du Développement, FHU TRANSLAD, Hôpital d'Enfants, CHU de Dijon, and Inserm - Université de Bourgogne UMR1231 GAD, FHU-TRANSLAD, Dijon, France
| | - Alice Fassier
- Service d'orthopédie pédiatrique, Centre de Compétences Maladies Osseuses Constitutionnelles, Hospices Civils de Lyon, Bron, France
| | - Olivier Prodhomme
- Service d'imagerie pédiatrique, Hôpital Arnaud de Villeneuve, CHU Montpellier, France
| | - Fabienne Escande
- Université de Lille, Lille, France.,Institut de Biochimie et Génétique moléculaire, CBP, CHU de Lille, France
| | - Sylvie Manouvrier
- Université de Lille, Lille, France.,CHU Lille, Clinique de Génétique, Hôpital Jeanne de Flandre, Lille, France
| | - Florence Petit
- Université de Lille, Lille, France.,CHU Lille, Clinique de Génétique, Hôpital Jeanne de Flandre, Lille, France
| | - David Geneviève
- Département de Génétique, IRMB, Maladies Rares et Médecine Personnalisée, Centre de Référence Maladies Rares ADSOOR, Filière AnDDI-Rare, INSERM U1183, CHU Montpellier, Université Montpellier, Montpellier, France
| | - Massimiliano Rossi
- Service de Génétique, Centre de Référence Anomalies du Développement et Centre de Compétences Maladies Osseuses Constitutionnelles, Hospices Civils de Lyon, Bron, France.,INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Bron, France
| |
Collapse
|
29
|
Sadler B, Haller G, Antunes L, Nikolov M, Amarillo I, Coe B, Dobbs MB, Gurnett CA. Rare and de novo duplications containing SHOX in clubfoot. J Med Genet 2020; 57:851-857. [PMID: 32518174 PMCID: PMC7688552 DOI: 10.1136/jmedgenet-2020-106842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 11/12/2022]
Abstract
Introduction Congenital clubfoot is a common birth defect that affects at least 0.1% of all births. Nearly 25% cases are familial and the remaining are sporadic in inheritance. Copy number variants (CNVs) involving transcriptional regulators of limb development, including PITX1 and TBX4, have previously been shown to cause familial clubfoot, but much of the heritability remains unexplained. Methods Exome sequence data from 816 unrelated clubfoot cases and 2645 in-house controls were analysed using coverage data to identify rare CNVs. The precise size and location of duplications were then determined using high-density Affymetrix Cytoscan chromosomal microarray (CMA). Segregation in families and de novo status were determined using qantitative PCR. Results Chromosome Xp22.33 duplications involving SHOX were identified in 1.1% of cases (9/816) compared with 0.07% of in-house controls (2/2645) (p=7.98×10−5, OR=14.57) and 0.27% (38/13592) of Atherosclerosis Risk in Communities/the Wellcome Trust Case Control Consortium 2 controls (p=0.001, OR=3.97). CMA validation confirmed an overlapping 180.28 kb duplicated region that included SHOX exons as well as downstream non-coding regions. In four of six sporadic cases where DNA was available for unaffected parents, the duplication was de novo. The probability of four de novo mutations in SHOX by chance in a cohort of 450 sporadic clubfoot cases is 5.4×10–10. Conclusions Microduplications of the pseudoautosomal chromosome Xp22.33 region (PAR1) containing SHOX and downstream enhancer elements occur in ~1% of patients with clubfoot. SHOX and regulatory regions have previously been implicated in skeletal dysplasia as well as idiopathic short stature, but have not yet been reported in clubfoot. SHOX duplications likely contribute to clubfoot pathogenesis by altering early limb development.
Collapse
Affiliation(s)
- Brooke Sadler
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Gabe Haller
- Department of Orthopedic Surgery, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Lilian Antunes
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Momchil Nikolov
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Ina Amarillo
- Department of Pathology and Immunology, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Bradley Coe
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington, USA.,Department of Pathology & Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew B Dobbs
- Department of Orthopedic Surgery, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| | - Christina A Gurnett
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
30
|
Zhang J, Li S, Ma S, Liu Y, Wang X, Li Y. Whole‑exome sequencing study identifies two novel rare variations associated with congenital talipes equinovarus. Mol Med Rep 2020; 21:2597-2602. [PMID: 32236576 DOI: 10.3892/mmr.2020.11038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 03/06/2020] [Indexed: 11/05/2022] Open
Abstract
Congenital talipes equinovarus (CTEV) is a common birth defect with an unclear genetic pathogenesis that results from both genetic and environmental factors. The present study aimed to identify novel variants in patients with CTEV using whole‑exome sequencing (WES) and to investigate the genetic factors responsible for the development of CTEV.A cohort of nine neonates/infants with suspected CTEV was recruited. Subsequently, sequential tests, including chromosome karyotyping and WES, were performed for each of the participants. Familial validation was performed using Sanger sequencing and low‑coverage copy‑number variation (CNV) sequencing. A novel CNV containing the mediator complex subunit 13L gene at 12q24.21‑q24.23 was detected by WES and further investigated by CNVseq. Additionally, a novel de novo missense variation, transforming growth factor‑β receptor 2: c.1280T>C, was identified by WES and further investigated by Sanger sequencing. The two identified variations were hypothesized to be causative genetic factors for the development of CTEV in the two cases the variations were identified in. In the present study, two pathogenic variations (one CNV and one single‑base variation) were detected in two Chinese families with CTEV. The results of the present study may aid in investigating the molecular basis of CTEV; however, further investigation is required.
Collapse
Affiliation(s)
- Jing Zhang
- Center of Prenatal Diagnosis, Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Shang Li
- Department of Anesthesiology and Operating Room, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Suling Ma
- Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Yan Liu
- Department of Pediatric Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Xuan Wang
- Department of Pediatric Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yazhou Li
- Department of Pediatric Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
31
|
Zhao X, Huang P, Li G, Feng Y, Zhendong L, Zhou C, Hu G, Xu Q. Overexpression of Pitx1 attenuates the senescence of chondrocytes from osteoarthritis degeneration cartilage-A self-controlled model for studying the etiology and treatment of osteoarthritis. Bone 2020; 131:115177. [PMID: 31783149 DOI: 10.1016/j.bone.2019.115177] [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: 05/25/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
To explore the role of low expression of Pitx1 in degenerative cartilage tissue. A cartilage injury model was established by using the cartilage scratch method. The newly generated tissue by BrdU labeled in injured cartilage region expressed SOX-9 and Col2A1 in 5-week-old rats. Compared with that, the number of BrdU-positive cells was lower in 4-month-old cartilage injury model rats. Compared with that in lateral cartilage, the expression of Pitx1 was lower in medial cartilage. Compared with chondrocytes derived from the lateral cartilage, chondrocytes derived from the medial cartilage exhibited significantly increased cell aging, as determined by SA-β-GAL staining; downregulated Pitx1 expression; reduced autophagy levels; and decreased Col2A1 expression in a chondrogenic differentiation assay. Inhibition of Pitx1 expression in chondrocytes from the lateral cartilage significantly increased the ratio of cell senescence. Overexpression of Pitx1 in chondrocytes derived from the medial cartilage decreased the cell senescence ratio. In a luciferase assay, Pitx1 was found to promote Sirt1 gene transcription. Decreased Pitx1 expression is an essential cause of cartilage degeneration in the medial tibial plateau. The described self-controlled model is an excellent way to study OA etiology and screen therapeutic drugs for OA.
Collapse
Affiliation(s)
- Xiang Zhao
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Huang
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gen Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yu Feng
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lv Zhendong
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chun Zhou
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guangyu Hu
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qingrong Xu
- Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai JiaoTong University, 160 Pujian Road, Shanghai 200127, China; Department of Orthopaedics, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2000, Jiangyue Road, Shanghai 201112, China.
| |
Collapse
|
32
|
Skuplik I, Cobb J. Animal Models for Understanding Human Skeletal Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:157-188. [DOI: 10.1007/978-981-15-2389-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
33
|
Abstract
PURPOSE OF REVIEW The goal of the review is to provide a comprehensive overview of the current understanding of the mechanisms underlying variation in human stature. RECENT FINDINGS Human height is an anthropometric trait that varies considerably within human populations as well as across the globe. Historically, much research focus was placed on understanding the biology of growth plate chondrocytes and how modifications to core chondrocyte proliferation and differentiation pathways potentially shaped height attainment in normal as well as pathological contexts. Recently, much progress has been made to improve our understanding regarding the mechanisms underlying the normal and pathological range of height variation within as well as between human populations, and today, it is understood to reflect complex interactions among a myriad of genetic, environmental, and evolutionary factors. Indeed, recent improvements in genetics (e.g., GWAS) and breakthroughs in functional genomics (e.g., whole exome sequencing, DNA methylation analysis, ATAC-sequencing, and CRISPR) have shed light on previously unknown pathways/mechanisms governing pathological and common height variation. Additionally, the use of an evolutionary perspective has also revealed important mechanisms that have shaped height variation across the planet. This review provides an overview of the current knowledge of the biological mechanisms underlying height variation by highlighting new research findings on skeletal growth control with an emphasis on previously unknown pathways/mechanisms influencing pathological and common height variation. In this context, this review also discusses how evolutionary forces likely shaped the genomic architecture of height across the globe.
Collapse
Affiliation(s)
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
34
|
Zhao X, Yang X. Retinoic Acid Promotes Retinoic Acid Signaling by Suppression of Pitx1 In Tendon Cells: A Possible Mechanism of a Clubfoot-Like Phenotype Induced by Retinoic Acid. Med Sci Monit 2019; 25:6980-6989. [PMID: 31527569 PMCID: PMC6761847 DOI: 10.12659/msm.917740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background The pathogenesis of idiopathic congenital clubfoot (CCF) is unknown. Although some familial patients have Pitx1 mutations, and the Pitx1+/− genotype causes a clubfoot-like phenotype in mice, the mechanism of Pitx1-induced CCF is unknown. Material/Methods We used tibialis anterior tendon samples to detect the expression of Pitx1 in idiopathic and neurogenic clubfoot patients. After obtaining Sprague-Dawley (SD) rat Achilles tendon cells, the expression of Pitx1 was knocked down by SiRNA. After 48 h of culture, mass spectrometry was used to quantitatively analyze proteins. Then, Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were used to assess the downstream pathway of PITX1. The relationship between Pitx1 and the promoter region of deacetylase 1 (Sirtuin-1 and Sirt1) was examined by luciferase and ChIP assays. Results We found that Pitx1 expression in the tendon samples of idiopathic CCF patients was downregulated. Mass spectrometry analysis revealed that the inhibition of Pitx1 induced the downregulation of Sirt1 expression in tendon cells. Luciferase and ChIP assays confirmed that Pitx1 binds to the promoter region of SIRT1 and promotes Sirt1 gene transcription. Further results showed that, after the inhibition of Pitx1 in tendon cells, CRABP2 acetylation increased, the nuclear import of CRABP2 was enhanced, and the expression of RARβ2 increased. After the inhibition of Pitx1, RARβ2 expression was further increased by RA treatment in tendon cells. In the presence of retinoic acid, the expression of Pitx1 was inhibited in tendon cells. Conclusions Pitx1 binds to the promoter region of SIRT1 and promotes the transcription of SIRT1. Positive feedback occurs between RA signaling and Pitx1.
Collapse
Affiliation(s)
- Xiang Zhao
- Department of Pediatric Orthopaedics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Xuan Yang
- Department of Pediatric Orthopaedics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (mainland)
| |
Collapse
|
35
|
Abstract
PURPOSE Congenital clubfoot is a serious birth defect that affects nearly 0.1% of all births. Though there is strong evidence for a genetic basis of isolated clubfoot, aside from a handful of associations, much of the heritability remains unexplained. METHODS By systematically examining the genes involved in syndromic clubfoot, we may find new candidate genes and pathways to investigate in isolated clubfoot. RESULTS In addition to the expected enrichment of extracellular matrix and transforming growth factor beta (TGF-β) signalling genes, we find many genes involved in syndromic clubfoot encode peroxisomal matrix proteins, as well as enzymes necessary for sulfation of proteoglycans, an important part of connective tissue. Further, the association of Filamin B with isolated clubfoot as well as syndromic clubfoot is an encouraging finding. CONCLUSION We should examine these categories for enrichment in isolated clubfoot patients to increase our understanding of the underlying biology and pathophysiology of this deformity. Understanding the spectrum of syndromes that have clubfoot as a feature enables a better understanding of the underlying pathophysiology of the disorder and directs future genetic screening efforts toward certain genes and genetic pathways. LEVEL OF EVIDENCE V.
Collapse
Affiliation(s)
- B. Sadler
- Department of Neurology, Washington University in St. Louis, St Louis, Missouri, USA
| | - C. A. Gurnett
- Department of Neurology, Washington University in St. Louis, St Louis, Missouri, USA
| | - M. B. Dobbs
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA,Correspondence should be sent to Matthew B. Dobbs, MD, 1 Children’s Place, Suite 4S-60, Department of Orthopedic Surgery, 660 S Euclid Ave, Campus Box 8233, Washington University in St Louis, St Louis, Missouri 63110, USA. E-mail:
| |
Collapse
|
36
|
Thompson AC, Capellini TD, Guenther CA, Chan YF, Infante CR, Menke DB, Kingsley DM. A novel enhancer near the Pitx1 gene influences development and evolution of pelvic appendages in vertebrates. eLife 2018; 7:38555. [PMID: 30499775 PMCID: PMC6269122 DOI: 10.7554/elife.38555] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/12/2018] [Indexed: 12/29/2022] Open
Abstract
Vertebrate pelvic reduction is a classic example of repeated evolution. Recurrent loss of pelvic appendages in sticklebacks has previously been linked to natural mutations in a pelvic enhancer that maps upstream of Pitx1. The sequence of this upstream PelA enhancer is not conserved to mammals, so we have surveyed a large region surrounding the mouse Pitx1 gene for other possible hind limb control sequences. Here we identify a new pelvic enhancer, PelB, that maps downstream rather than upstream of Pitx1. PelB drives expression in the posterior portion of the developing hind limb, and deleting the sequence from mice alters the size of several hind limb structures. PelB sequences are broadly conserved from fish to mammals. A wild stickleback population lacking the pelvis has an insertion/deletion mutation that disrupts the structure and function of PelB, suggesting that changes in this ancient enhancer contribute to evolutionary modification of pelvic appendages in nature.
Collapse
Affiliation(s)
- Abbey C Thompson
- Department of Developmental Biology, Stanford University School of Medicine, California, United States.,Department of Genetics, Stanford University School of Medicine, California, United States
| | - Terence D Capellini
- Department of Developmental Biology, Stanford University School of Medicine, California, United States
| | - Catherine A Guenther
- Department of Developmental Biology, Stanford University School of Medicine, California, United States.,Howard Hughes Medical Institute, Stanford University, California, United States
| | - Yingguang Frank Chan
- Department of Developmental Biology, Stanford University School of Medicine, California, United States
| | - Carlos R Infante
- Department of Genetics, University of Georgia, Georgia, United States
| | - Douglas B Menke
- Department of Genetics, University of Georgia, Georgia, United States
| | - David M Kingsley
- Department of Developmental Biology, Stanford University School of Medicine, California, United States.,Howard Hughes Medical Institute, Stanford University, California, United States
| |
Collapse
|
37
|
Dynamic 3D chromatin architecture contributes to enhancer specificity and limb morphogenesis. Nat Genet 2018; 50:1463-1473. [PMID: 30262816 PMCID: PMC10154999 DOI: 10.1038/s41588-018-0221-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022]
Abstract
The regulatory specificity of enhancers and their interaction with gene promoters is thought to be controlled by their sequence and the binding of transcription factors. By studying Pitx1, a regulator of hindlimb development, we show that dynamic changes in chromatin conformation can restrict the activity of enhancers. Inconsistent with its hindlimb-restricted expression, Pitx1 is controlled by an enhancer (Pen) that shows activity in forelimbs and hindlimbs. By Capture Hi-C and three-dimensional modeling of the locus, we demonstrate that forelimbs and hindlimbs have fundamentally different chromatin configurations, whereby Pen and Pitx1 interact in hindlimbs and are physically separated in forelimbs. Structural variants can convert the inactive into the active conformation, thereby inducing Pitx1 misexpression in forelimbs, causing partial arm-to-leg transformation in mice and humans. Thus, tissue-specific three-dimensional chromatin conformation can contribute to enhancer activity and specificity in vivo and its disturbance can result in gene misexpression and disease.
Collapse
|
38
|
Kinold JC, Pfarr C, Aberle H. Sidestep-induced neuromuscular miswiring causes severe locomotion defects in Drosophila larvae. Development 2018; 145:145/17/dev163279. [DOI: 10.1242/dev.163279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/17/2018] [Indexed: 01/12/2023]
Abstract
ABSTRACT
Mutations in motor axon guidance molecules cause aberrant projection patterns of motor nerves. As most studies in Drosophila have analysed these molecules in fixed embryos, the consequences for larval locomotion are entirely unexplored. Here, we took advantage of sidestep (side)-mutant larvae that display severe locomotion defects because of irreparable innervation errors. Mutations in side affected all motor nerve branches and all body wall regions. Innervation defects were non-stereotypical, showing unique innervation patterns in each hemisegment. Premature activation of Side in muscle precursors abrogated dorsal migration of motor nerves, resulting in larvae with a complete loss of neuromuscular junctions on dorsal-most muscles. High-speed videography showed that these larvae failed to maintain substrate contact and inappropriately raised both head and tail segments above the substrate, resulting in unique ‘arching’ and ‘lifting’ phenotypes. These results show that guidance errors in side mutants are maintained throughout larval life and are asymmetrical with respect to the bilateral body axis. Together with similar findings in mice, this study also suggests that miswiring could be an underlying cause of inherited movement disorders.
Collapse
Affiliation(s)
- Jaqueline C. Kinold
- Heinrich Heine University Düsseldorf, Functional Cell Morphology Lab, Building 26-12-00, Universitaetsstrasse 1, 40225 Düsseldorf, Germany
| | - Carsten Pfarr
- Heinrich Heine University Düsseldorf, Functional Cell Morphology Lab, Building 26-12-00, Universitaetsstrasse 1, 40225 Düsseldorf, Germany
| | - Hermann Aberle
- Heinrich Heine University Düsseldorf, Functional Cell Morphology Lab, Building 26-12-00, Universitaetsstrasse 1, 40225 Düsseldorf, Germany
| |
Collapse
|
39
|
Pavone V, Chisari E, Vescio A, Lucenti L, Sessa G, Testa G. The etiology of idiopathic congenital talipes equinovarus: a systematic review. J Orthop Surg Res 2018; 13:206. [PMID: 30134936 PMCID: PMC6104023 DOI: 10.1186/s13018-018-0913-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/15/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Also known as clubfoot, idiopathic congenital talipes equinovarus (ICTEV) is the most common pediatric deformity and occurs in 1 in every 1000 live births. Even though it has been widely researched, the etiology of ICTEV remains poorly understood and is often described as being based on a multifactorial genesis. Genetic and environmental factors seem to have a major role in the development of this disease. Thus, the aim of this review is to analyze the available literature to document the current evidence on ICTEV etiology. METHODS The literature on ICTEV etiology was systematically reviewed using the following inclusion criteria: studies of any level of evidence, reporting clinical or preclinical results, published in the last 20 years (1998-2018), and dealing with the etiology of ICTEV. RESULTS A total of 48 articles were included. ICTEV etiology is still controversial. Several hypotheses have been researched, but none of them are decisive. Emerging evidence suggests a role of several pathways and gene families associated with limb development (HOX family; PITX1-TBX4), the apoptotic pathway (caspases), and muscle contractile protein (troponin and tropomyosin), but a major candidate gene has still not been identified. Strong recent evidence emerging from twin studies confirmed major roles of genetics and the environment in the disease pathogenesis. CONCLUSIONS The available literature on the etiology of ICTEV presents major limitations in terms of great heterogeneity and a lack of high-profile studies. Although many studies focus on the genetic background of the disease, there is lack of consensus on one or multiple targets. Genetics and smoking seem to be strongly associated with ICTEV etiology, but more studies are needed to understand the complex and multifactorial genesis of this common congenital lower-limb disease.
Collapse
Affiliation(s)
- Vito Pavone
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Via Plebiscito, 628, 95124 Catania, Italy
| | - Emanuele Chisari
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Via Plebiscito, 628, 95124 Catania, Italy
| | - Andrea Vescio
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Via Plebiscito, 628, 95124 Catania, Italy
| | - Ludovico Lucenti
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Via Plebiscito, 628, 95124 Catania, Italy
| | - Giuseppe Sessa
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Via Plebiscito, 628, 95124 Catania, Italy
| | - Gianluca Testa
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Via Plebiscito, 628, 95124 Catania, Italy
| |
Collapse
|
40
|
Shi B, Xu L, Mao S, Xu L, Liu Z, Sun X, Zhu Z, Qiu Y. Abnormal PITX1 gene methylation in adolescent idiopathic scoliosis: a pilot study. BMC Musculoskelet Disord 2018; 19:138. [PMID: 29743058 PMCID: PMC5941792 DOI: 10.1186/s12891-018-2054-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 04/23/2018] [Indexed: 01/18/2023] Open
Abstract
Background The gene of pituitary homeobox 1 (PITX1) has been reported to be down-regulated in adolescent idiopathic scoliosis (AIS), of which the cause has not been well addressed. The abnormal DNA methylation was recently assumed to be an important mechanism for the down-regulated genes expression. However, the association between PITX1 promoter methylation and the etiology of AIS was not clear. Methods The peripheral blood samples of 50 AIS patients and 50 healthy controls were collected and the genomic DNA was extracted. The pyrosequencing assay was used to assess the methylation status of PITX1 promoter and real-time quantitative polymerase chain reaction (PCR) was used to detect the PITX1 gene expression. Comparison analysis was performed using independent t test and Chi-square tests, while correlation analysis were performed with 2-tailed Pearson coefficients. Results The mean methylation level was (3.52 ± 0.96)% in AIS and (1.40 ± 0.81)% in healthy controls (P < 0.0001). The PITX1 gene expression was 0.15 ± 0.08 in AIS and 0.80 ± 0.55 in healthy controls (P < 0.0001). The comparative analysis showed significant difference in age (P = 0.021) and Cobb angle of the main curve (P = 0.0001) between AIS groups with positive and negative methylation. The methylation level of 6 CpG sites in PITX1 promoters was significantly associated with Cobb angle of the main curve (P < 0.001) in AIS. No statistical relationship between PITX1 promoter methylation and gene expression was found in AIS (P = 0.842). Conclusion Significantly higher methylation level and lower PITX1 gene expression are found in AIS patients. PITX1 methylation is associated with Cobb angles of the main curves in AIS. DNA methylation thus plays an important role in the etiology and curve progression in AIS.
Collapse
Affiliation(s)
- Benlong Shi
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Liang Xu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Saihu Mao
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Leilei Xu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Zhen Liu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Xu Sun
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| | - Zezhang Zhu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China.
| | - Yong Qiu
- Spine Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road No. 321, Nanjing, 210008, China
| |
Collapse
|
41
|
Sarro R, Kocher AA, Emera D, Uebbing S, Dutrow EV, Weatherbee SD, Nottoli T, Noonan JP. Disrupting the three-dimensional regulatory topology of the Pitx1 locus results in overtly normal development. Development 2018; 145:dev158550. [PMID: 29549111 PMCID: PMC5963865 DOI: 10.1242/dev.158550] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 03/09/2018] [Indexed: 12/28/2022]
Abstract
Developmental gene expression patterns are orchestrated by thousands of distant-acting transcriptional enhancers. However, identifying enhancers essential for the expression of their target genes has proven challenging. Maps of long-range regulatory interactions may provide the means to identify enhancers crucial for developmental gene expression. To investigate this hypothesis, we used circular chromosome conformation capture coupled with interaction maps in the mouse limb to characterize the regulatory topology of Pitx1, which is essential for hindlimb development. We identified a robust hindlimb-specific interaction between Pitx1 and a putative hindlimb-specific enhancer. To interrogate the role of this interaction in Pitx1 regulation, we used genome editing to delete this enhancer in mouse. Although deletion of the enhancer completely disrupts the interaction, Pitx1 expression in the hindlimb is only mildly affected, without any detectable compensatory interactions between the Pitx1 promoter and potentially redundant enhancers. Pitx1 enhancer null mice did not exhibit any of the characteristic morphological defects of the Pitx1-/- mutant. Our results suggest that robust, tissue-specific physical interactions at essential developmental genes have limited predictive value for identifying enhancer mutations with strong loss-of-function phenotypes.
Collapse
Affiliation(s)
- Richard Sarro
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Acadia A Kocher
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Deena Emera
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Severin Uebbing
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Emily V Dutrow
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Scott D Weatherbee
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Timothy Nottoli
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Genome Editing Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - James P Noonan
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
42
|
Collinson JM, Lindström NO, Neves C, Wallace K, Meharg C, Charles RH, Ross ZK, Fraser AM, Mbogo I, Oras K, Nakamoto M, Barker S, Duce S, Miedzybrodzka Z, Vargesson N. The developmental and genetic basis of 'clubfoot' in the peroneal muscular atrophy mutant mouse. Development 2018; 145:145/3/dev160093. [PMID: 29439133 DOI: 10.1242/dev.160093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/28/2017] [Indexed: 12/19/2022]
Abstract
Genetic factors underlying the human limb abnormality congenital talipes equinovarus ('clubfoot') remain incompletely understood. The spontaneous autosomal recessive mouse 'peroneal muscular atrophy' mutant (PMA) is a faithful morphological model of human clubfoot. In PMA mice, the dorsal (peroneal) branches of the sciatic nerves are absent. In this study, the primary developmental defect was identified as a reduced growth of sciatic nerve lateral motor column (LMC) neurons leading to failure to project to dorsal (peroneal) lower limb muscle blocks. The pma mutation was mapped and a candidate gene encoding LIM-domain kinase 1 (Limk1) identified, which is upregulated in mutant lateral LMC motor neurons. Genetic and molecular analyses showed that the mutation acts in the EphA4-Limk1-Cfl1/cofilin-actin pathway to modulate growth cone extension/collapse. In the chicken, both experimental upregulation of Limk1 by electroporation and pharmacological inhibition of actin turnover led to defects in hindlimb spinal motor neuron growth and pathfinding, and mimicked the clubfoot phenotype. The data support a neuromuscular aetiology for clubfoot and provide a mechanistic framework to understand clubfoot in humans.
Collapse
Affiliation(s)
- J Martin Collinson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Nils O Lindström
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Carlos Neves
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Karen Wallace
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Caroline Meharg
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Rebecca H Charles
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Zoe K Ross
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Amy M Fraser
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Ivan Mbogo
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Kadri Oras
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Masaru Nakamoto
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Simon Barker
- Royal Aberdeen Children's Hospital, Foresterhill, Aberdeen AB25 2ZN, UK
| | - Suzanne Duce
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Zosia Miedzybrodzka
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| |
Collapse
|
43
|
Hadjiargyrou M. Mustn1: A Developmentally Regulated Pan-Musculoskeletal Cell Marker and Regulatory Gene. Int J Mol Sci 2018; 19:ijms19010206. [PMID: 29329193 PMCID: PMC5796155 DOI: 10.3390/ijms19010206] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/26/2017] [Accepted: 01/06/2018] [Indexed: 02/07/2023] Open
Abstract
The Mustn1 gene encodes a small nuclear protein (~9.6 kDa) that does not belong to any known family. Its genomic organization consists of three exons interspersed by two introns and it is highly homologous across vertebrate species. Promoter analyses revealed that its expression is regulated by the AP family of transcription factors, especially c-Fos, Fra-2 and JunD. Mustn1 is predominantly expressed in the major tissues of the musculoskeletal system: bone, cartilage, skeletal muscle and tendon. Its expression has been associated with normal embryonic development, postnatal growth, exercise, and regeneration of bone and skeletal muscle. Moreover, its expression has also been detected in various musculoskeletal pathologies, including arthritis, Duchenne muscular dystrophy, other skeletal muscle myopathies, clubfoot and diabetes associated muscle pathology. In vitro and in vivo functional perturbation revealed that Mustn1 is a key regulatory molecule in myogenic and chondrogenic lineages. This comprehensive review summarizes our current knowledge of Mustn1 and proposes that it is a new developmentally regulated pan-musculoskeletal marker as well as a key regulatory protein for cell differentiation and tissue growth.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY 11568-8000, USA.
| |
Collapse
|
44
|
Wang JS, Infante CR, Park S, Menke DB. PITX1 promotes chondrogenesis and myogenesis in mouse hindlimbs through conserved regulatory targets. Dev Biol 2017; 434:186-195. [PMID: 29273440 DOI: 10.1016/j.ydbio.2017.12.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/05/2017] [Accepted: 12/18/2017] [Indexed: 10/18/2022]
Abstract
The PITX1 transcription factor is expressed during hindlimb development, where it plays a critical role in directing hindlimb growth and the specification of hindlimb morphology. While it is known that PITX1 regulates hindlimb formation, in part, through activation of the Tbx4 gene, other transcriptional targets remain to be elucidated. We have used a combination of ChIP-seq and RNA-seq to investigate enhancer regions and target genes that are directly regulated by PITX1 in embryonic mouse hindlimbs. In addition, we have analyzed PITX1 binding sites in hindlimbs of Anolis lizards to identify ancient PITX1 regulatory targets. We find that PITX1-bound regions in both mouse and Anolis hindlimbs are strongly associated with genes implicated in limb and skeletal system development. Gene expression analyses reveal a large number of misexpressed genes in the hindlimbs of Pitx1-/- mouse embryos. By intersecting misexpressed genes with genes that have neighboring mouse PITX1 binding sites, we identified 440 candidate targets of PITX1. Of these candidates, 68 exhibit ultra-conserved PITX1 binding events that are shared between mouse and Anolis hindlimbs. Among the ancient targets of PITX1 are important regulators of cartilage and skeletal muscle development, including Sox9 and Six1. Our data suggest that PITX1 promotes chondrogenesis and myogenesis in the hindlimb by direct regulation of several key members of the cartilage and muscle transcriptional networks.
Collapse
Affiliation(s)
- Jialiang S Wang
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Carlos R Infante
- Department of Genetics, University of Georgia, Athens, GA 30602, USA; Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Sungdae Park
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Douglas B Menke
- Department of Genetics, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
45
|
Basit S, Khoshhal KI. Genetics of clubfoot; recent progress and future perspectives. Eur J Med Genet 2017; 61:107-113. [PMID: 28919208 DOI: 10.1016/j.ejmg.2017.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/05/2017] [Accepted: 09/10/2017] [Indexed: 12/20/2022]
Abstract
Clubfoot or talipes equinovarus (TEV) is an inborn three-dimensional deformity of leg, ankle and foot. It results from structural defects of several tissues of foot and lower leg leading to abnormal positioning of foot and ankle joints. TEV can lead to long-lasting functional disability, malformation and discomfort if left untreated. Substantial progress has been achieved in the management and diagnosis of limb defects; however, not much is known about the molecular players and signalling pathways underlying TEV disorder. The homeostasis and development of the limb depends on the complex interactions between the lateral plate mesoderm cells and outer ectoderm. These complex interactions include HOX signalling and PITX1-TBX4 pathways. The susceptibility to develop TEV is determined by a number of environmental and genetic factors, although the nature and level of interplay between them remains unclear. Familial occurrence and inter and intra phenotypic variability of TEV is well documented. Variants in genes that code for contractile proteins of skeletal myofibers might play a role in the aetiology of TEV but, to date, no strong candidate genes conferring increased risk have emerged, although variants in TBX4, PITX1, HOXA, HOXC and HOXD clusters genes, NAT2 and others have been shown to be associated with TEV. The mechanisms by which variants in these genes confer risk and the nature of the physical and genetic interaction between them remains to be determined. Elucidation of genetic players and cellular pathways underlying TEV will certainly increase our understanding of the pathophysiology of this deformity.
Collapse
Affiliation(s)
- Sulman Basit
- Centre for Genetics and Inherited Diseases, Taibah University Almadinah Almunawwarah, Saudi Arabia.
| | - Khalid I Khoshhal
- College of Medicine, Taibah University Almadinah Almunawwarah, Saudi Arabia
| |
Collapse
|
46
|
Hernigou P. History of clubfoot treatment; part III (twentieth century): back to the future. INTERNATIONAL ORTHOPAEDICS 2017; 41:2407-2414. [DOI: 10.1007/s00264-017-3629-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 08/21/2017] [Indexed: 12/15/2022]
|
47
|
Dobbs MB, Gurnett CA. The 2017 ABJS Nicolas Andry Award: Advancing Personalized Medicine for Clubfoot Through Translational Research. Clin Orthop Relat Res 2017; 475:1716-1725. [PMID: 28236079 PMCID: PMC5406347 DOI: 10.1007/s11999-017-5290-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/16/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND Clubfoot is one of the most common pediatric orthopaedic disorders. While the Ponseti method has revolutionized clubfoot treatment, it is not effective for all patients. When the Ponseti method does not correct the foot, patients are at risk for lifelong disability and may require more-extensive surgery. QUESTIONS/PURPOSES (1) What genetic and morphologic abnormalities contribute to the development of clubfoot? (2) How can this information be used to devise personalized treatment paradigms for patients with clubfoot? METHODS Human gene sequencing, molecular genetic engineering of mouse models of clubfoot, MRI of clubfoot, and development of new treatment methods all have been used by our group to understand the biological basis and improve therapy for this group of disorders. RESULTS We gained new insight into clubfoot pathogenesis from our discovery that mutations in the PITX1-TBX4-HOXC transcriptional pathway cause familial clubfoot and vertical talus in a small number of families, with the unique lower limb expression of these genes providing an explanation for the lack of upper extremity involvement in these disorders. MRI studies revealed corresponding morphologic abnormalities, including hypomorphic muscle, bone, and vasculature, that are not only associated with these gene mutations, but also are biomarkers for treatment-resistant clubfoot. CONCLUSIONS Based on an understanding of the underlying biology, we improved treatment methods for neglected and syndromic clubfoot, developed new treatment for congenital vertical talus based on the principles of the Ponseti method, and designed a new dynamic clubfoot brace to improve strength and compliance.
Collapse
Affiliation(s)
- Matthew B. Dobbs
- 0000 0000 9953 7617grid.416775.6Department of Orthopaedics, St. Louis Children’s Hospital, 1 Children’s Place, Suite 4S-60, St. Louis, MO 63110 USA
| | - Christina A. Gurnett
- 0000 0001 2355 7002grid.4367.6Department of Neurology, Washington University School of Medicine in St Louis, St. Louis, MO USA
| |
Collapse
|
48
|
Sinha N, 1 Department of Orthopaedic Surgery, Geisinger Medical Center, Danville, PA, USA;, A. Seeley M, S. Horwitz D, Maniar H, H. Seeley A, 2 Department of Pediatrics, Geisinger Medical Center, Danville, PA, USA. Pediatric Orthogenomics: The Latest Trends and Controversies. AIMS MEDICAL SCIENCE 2017. [DOI: 10.3934/medsci.2017.2.192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
49
|
Domyan ET, Kronenberg Z, Infante CR, Vickrey AI, Stringham SA, Bruders R, Guernsey MW, Park S, Payne J, Beckstead RB, Kardon G, Menke DB, Yandell M, Shapiro MD. Molecular shifts in limb identity underlie development of feathered feet in two domestic avian species. eLife 2016; 5:e12115. [PMID: 26977633 PMCID: PMC4805547 DOI: 10.7554/elife.12115] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/19/2016] [Indexed: 12/15/2022] Open
Abstract
Birds display remarkable diversity in the distribution and morphology of scales and feathers on their feet, yet the genetic and developmental mechanisms governing this diversity remain unknown. Domestic pigeons have striking variation in foot feathering within a single species, providing a tractable model to investigate the molecular basis of skin appendage differences. We found that feathered feet in pigeons result from a partial transformation from hindlimb to forelimb identity mediated by cis-regulatory changes in the genes encoding the hindlimb-specific transcription factor Pitx1 and forelimb-specific transcription factor Tbx5. We also found that ectopic expression of Tbx5 is associated with foot feathers in chickens, suggesting similar molecular pathways underlie phenotypic convergence between these two species. These results show how changes in expression of regional patterning genes can generate localized changes in organ fate and morphology, and provide viable molecular mechanisms for diversity in hindlimb scale and feather distribution.
Collapse
Affiliation(s)
- Eric T Domyan
- Department of Biology, University of Utah, Salt Lake City, United States
| | - Zev Kronenberg
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Carlos R Infante
- Department of Genetics, University of Georgia, Athens, United States
| | - Anna I Vickrey
- Department of Biology, University of Utah, Salt Lake City, United States
| | - Sydney A Stringham
- Department of Biology, University of Utah, Salt Lake City, United States
| | - Rebecca Bruders
- Department of Biology, University of Utah, Salt Lake City, United States
| | - Michael W Guernsey
- Department of Biology, University of Utah, Salt Lake City, United States
| | - Sungdae Park
- Department of Genetics, University of Georgia, Athens, United States
| | - Jason Payne
- Poultry Science Department, University of Georgia, Athens, United States
| | - Robert B Beckstead
- Poultry Science Department, University of Georgia, Athens, United States
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Douglas B Menke
- Department of Genetics, University of Georgia, Athens, United States
| | - Mark Yandell
- Department of Human Genetics, University of Utah, Salt Lake City, United States
- Utah Center for Genetic Discovery, University of Utah, Salt Lake City, United States
| | - Michael D Shapiro
- Department of Biology, University of Utah, Salt Lake City, United States
| |
Collapse
|
50
|
Alvarado DM, McCall K, Hecht JT, Dobbs MB, Gurnett CA. Deletions of 5' HOXC genes are associated with lower extremity malformations, including clubfoot and vertical talus. J Med Genet 2016; 53:250-5. [PMID: 26729820 DOI: 10.1136/jmedgenet-2015-103505] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/29/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND Deletions of the HOXC gene cluster result in variable phenotypes in mice, but have been rarely described in humans. OBJECTIVE To report chromosome 12q13.13 microdeletions ranging from 13 to 175 kb and involving the 5' HOXC genes in four families, segregating congenital lower limb malformations, including clubfoot, vertical talus and hip dysplasia. METHODS Probands (N=253) with clubfoot or vertical talus were screened for point mutations and copy number variants using multiplexed direct genomic selection, a pooled BAC targeted capture approach. SNP genotyping included 1178 probands with clubfoot or vertical talus and 1775 controls. RESULTS The microdeletions share a minimal non-coding region overlap upstream of HOXC13, with variable phenotypes depending upon HOXC13, HOXC12 or the HOTAIR lncRNA inclusion. SNP analysis revealed HOXC11 p.Ser191Phe segregating with clubfoot in a small family and enrichment of HOXC12 p.Asn176Lys in patients with clubfoot or vertical talus (rs189468720, p=0.0057, OR=3.8). Defects in limb morphogenesis include shortened and overlapping toes, as well as peroneus muscle hypoplasia. Finally, HOXC and HOXD gene expression is reduced in fibroblasts from a patient with a 5' HOXC deletion, consistent with previous studies demonstrating that dosage of lncRNAs alters expression of HOXD genes in trans. CONCLUSIONS Because HOXD10 has been implicated in the aetiology of congenital vertical talus, variation in its expression may contribute to the lower limb phenotypes occurring with 5' HOXC microdeletions. Identification of 5' HOXC microdeletions highlights the importance of transcriptional regulators in the aetiology of severe lower limb malformations and will improve their diagnosis and management.
Collapse
Affiliation(s)
- David M Alvarado
- Department of Orthopedic Surgery, Washington University, St. Louis, Missouri, USA
| | - Kevin McCall
- Department of Orthopedic Surgery, Washington University, St. Louis, Missouri, USA
| | - Jacqueline T Hecht
- Department of Pediatrics, University of Texas Medical School, Houston, Texas, USA
| | - Matthew B Dobbs
- Department of Orthopedic Surgery, Washington University, St. Louis, Missouri, USA Shriners Hospital for Children, St Louis, Missouri, USA
| | - Christina A Gurnett
- Department of Orthopedic Surgery, Washington University, St. Louis, Missouri, USA Department of Neurology, Washington University, St. Louis, Missouri, USA Department of Pediatrics, Washington University, St. Louis, Missouri, USA
| |
Collapse
|