1
|
Adhikari N, Wu Z, Huang Y, Lan Y, Jiang R. Twist1 Acts Upstream of the Dlx5-Hand2 Pathway to Pattern the Mammalian Jaw. J Dent Res 2025; 104:310-319. [PMID: 39707586 DOI: 10.1177/00220345241291527] [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] [Indexed: 12/23/2024] Open
Abstract
Both the upper and lower jaws develop from cranial neural crest cells (CNCCs) populating the first pharyngeal arch in all gnathostomes. Previous studies showed that the Edn1/Ednra-Dlx5/Dlx6-Hand2 signaling pathway is necessary for lower jaw formation and that ectopic expression of Edn1 or Hand2 throughout the CNCCs partly transformed the upper jaw to lower jaw structures, but the molecular mechanisms regulating upper jaw development remain unclear. Here we show that the basic helix-loop-helix transcription factor Twist1 is required for upper jaw development. Whereas the Twist1fl/fl;Wnt1-Cre mouse embryos, with tissue-specific inactivation of Twist1 in premigratory CNCCs, exhibited aberrantly persistent expression of the key neuroglial lineage regulator Sox10 in the postmigratory CNCCs populating the facial primordia, we found that genetic inactivation of Sox10 did not rescue the defects in CNCC survival and patterning in Twist1fl/fl;Wnt1-Cre embryos. However, analysis of Sox10fl/+;Twist1fl/fl;Wnt1-Cre mice revealed duplicated mandibular structures, including ectopic Meckel's cartilage, in place of the maxilla. Both Sox10fl/+;Twist1fl/fl;Wnt1-Cre and Sox10fl/fl;Twist1fl/fl;Wnt1-Cre embryos exhibited ectopic expression of Dlx5 and Hand2 in the developing maxillary processes at E10.5. Furthermore, we found that Twist1fl/fl;Wnt1-Cre embryos also expressed Dlx5 and Hand2 ectopically in the maxillary domain at E10.5 and subsequently developed Meckel's cartilage-like cartilage rods bilaterally at the maxillary region. However, the expression of Edn1 was unaltered in the developing Twist1fl/fl;Wnt1-Cre embryos, indicating that Twist1 functions in the CNCC-derived facial mesenchyme to regulate the Dlx5-Hand2 pathway without affecting Edn1 expression in the epithelial and mesodermal compartments. We further show that Twist1 represses reporter gene activation driven by the Dlx5/Dlx6 intergenic enhancer known to drive Dlx5/Dlx6 expression in the developing mandibular arch. Together, these data identify a new role of Twist1 in patterning the regional identities of the CNCC-derived facial mesenchyme and provide novel insight into the pathogenic mechanisms underlying TWIST1-related craniofacial developmental disorders.
Collapse
Affiliation(s)
- N Adhikari
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Z Wu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR
| | - Y Huang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Graduate Program in Development, Stem Cells, and Regenerative Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Y Lan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Departments of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - R Jiang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Departments of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
2
|
Tu M, Ge B, Li J, Pan Y, Zhao B, Han J, Wu J, Zhang K, Liu G, Hou M, Yue M, Han X, Sun T, An Y. Emerging biological functions of Twist1 in cell differentiation. Dev Dyn 2025; 254:8-25. [PMID: 39254141 DOI: 10.1002/dvdy.736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/03/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Twist1 is required for embryonic development and expresses after birth in mesenchymal stem cells derived from mesoderm, where it governs mesenchymal cell development. As a well-known regulator of epithelial-mesenchymal transition or embryonic organogenesis, Twist1 is important in a variety of developmental systems, including mesoderm formation, neurogenesis, myogenesis, cranial neural crest cell migration, and differentiation. In this review, we first highlight the physiological significance of Twist1 in cell differentiation, including osteogenic, chondrogenic, and myogenic differentiation, and then detail its probable molecular processes and signaling pathways. On this premise, we summarize the significance of Twist1 in distinct developmental disorders and diseases to provide a reference for studies on cell differentiation/development-related diseases.
Collapse
Affiliation(s)
- Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Bingqian Ge
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jialin Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Guangchao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengwen Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Man Yue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
3
|
Al Jadeedi S, Al Alawi KM, Al Bulushi T. Craniosynostosis: Epidemiology and Pattern at a Tertiary Referral Institute in Oman 2004 to 2023. Cleft Palate Craniofac J 2024:10556656241304544. [PMID: 39698983 DOI: 10.1177/10556656241304544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE To date, there are no published studies From the Sultanate of Oman on the incidence or characteristics of craniosynostosis (CS). This is a population-based epidemiological study of the incidence of CS. METHODS The prospective registry of the craniofacial surgery unit in Khoula Hospital was used to retrieve data on all individuals with CS treated between 2004 and 2023. The cohort was divided into four 5-year groups based on year of birth: 2004 to 2008, 2009 to 2013, 2014 to 2018, and 2019 to 2023. RESULTS We identified 312 individuals with CS. The incidence increased significantly during the study period and was 2.5 per 10 000 live births in the last 5-year period. There was a male preponderance (male/female ratio 1.5:1). Our study findings reveal a notable diversity in the trend of suture involvement, we observed a higher frequency of complex CS within our study population 35.9%. Half of the study population was nonsyndromic, accounting for 51.6%. The nonsyndromic population exhibits a higher proportion of midline suture involvement. CONCLUSIONS The incidence of CS increased during the study period. The majority of cases were identified as nonsyndromic. We found that multiple sutures CS were the most prevalent overall in our population. It is imperative to intensify efforts aimed at raising awareness among the general population regarding these deformities.
Collapse
Affiliation(s)
- Sondus Al Jadeedi
- Department of Plastic and Reconstructive Surgery, Craniofacial Surgery and Burn Department, Khoula Hospital, Mina Al Fahal, Muscat, Sultanate of Oman
| | - Khalifa Mohammed Al Alawi
- Department of Plastic and Reconstructive Surgery, Craniofacial Surgery and Burn Department, Khoula Hospital, Mina Al Fahal, Muscat, Sultanate of Oman
| | - Taimoor Al Bulushi
- Department of Plastic and Reconstructive Surgery, Craniofacial Surgery and Burn Department, Khoula Hospital, Mina Al Fahal, Muscat, Sultanate of Oman
| |
Collapse
|
4
|
Lovisa S, Vetrano S. TWISTed fibroblasts: New drivers of intestinal fibrosis in Crohn's disease. Heliyon 2024; 10:e40604. [PMID: 39654763 PMCID: PMC11626011 DOI: 10.1016/j.heliyon.2024.e40604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
Fibrosis is the pathological consequence of chronic inflammation. In Crohn's disease (CD), fibrostenotic complications occur with 50-70 % frequency as a failure to properly repair the tissue damage. Intestinal stenosis requires surgical intervention and relapses in most patients. Mesenchymal cells encompassed of heterogeneous cell subsets orchestrate this complex process. The lack of a full characterization of the stromal diversity and function in CD has consequently slowed the development of anti-fibrotic targets. Two recent studies align together demonstrating FAP+TWIST1+ fibroblasts as the primary mesenchymal population driving intestinal fibrosis in CD. Genetic and pharmacological targeting of Twist1 in mouse models proved the functional role of Fap+Twist1+ fibroblasts and indicate the use of the Twist1 inhibitor harmine as a potential therapeutic strategy to revert fibrosis.
Collapse
Affiliation(s)
- Sara Lovisa
- Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
- Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Stefania Vetrano
- Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
- Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
5
|
Sun L, Liu L, Jiang J, Liu K, Zhu J, Wu L, Lu X, Huang Z, Yuan Y, Crowley SD, Mao H, Xing C, Ren J. Transcription factor Twist1 drives fibroblast activation to promote kidney fibrosis via signaling proteins Prrx1/TNC. Kidney Int 2024; 106:840-855. [PMID: 39181396 DOI: 10.1016/j.kint.2024.07.028] [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: 11/30/2023] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024]
Abstract
The transcription factor Twist1 plays a vital role in normal development in many tissue systems and continues to be important throughout life. However, inappropriate Twist1 activity has been associated with kidney injury and fibrosis, though the underlying mechanisms involved remain incomplete. Here, we explored the role of Twist1 in regulating fibroblast behaviors and the development kidney fibrosis. Initially Twist1 protein and activity was found to be markedly increased within interstitial myofibroblasts in fibrotic kidneys in both humans and rodents. Treatment of rat kidney interstitial fibroblasts with transforming growth factor-β1 (a profibrotic factor) also induced Twist1 expression in vitro. Gain- and loss-of-function experiments supported that Twist1 signaling was responsible for transforming growth factor-β1-induced fibroblast activation and fetal bovine serum-induced fibroblast proliferation. Mechanistically, Twist1 protein promoted kidney fibroblast activation by driving the expression of downstream signaling proteins, Prrx1 and Tnc. Twist1 directly enhanced binding to the promoter of Prrx1 but not TNC, whereas the promoter of TNC was directly bound by Prrx1. Finally, mice with fibroblast-specific deletion of Twist1 exhibited less Prrx1 and TNC protein abundance, interstitial extracellular matrix deposition and kidney inflammation in both the unilateral ureteral obstruction and ischemic-reperfusion injury-induced-kidney fibrotic models. Inhibition of Twist1 signaling with Harmine, a β-carboline alkaloid, improved extracellular matrix deposition in both injury models. Thus, our results suggest that Twist1 signaling promotes the activation and proliferation of kidney fibroblasts, contributing to the development of interstitial fibrosis, offering a potential therapeutic target for chronic kidney disease.
Collapse
Affiliation(s)
- Lianqin Sun
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lishan Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Juanjuan Jiang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kang Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jingfeng Zhu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lin Wu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaohan Lu
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Zhimin Huang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yanggang Yuan
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Department of Medicine, Durham VA Medical Center, Durham, North Carolina, USA
| | - Huijuan Mao
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Changying Xing
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Jiafa Ren
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
6
|
Farmer DT, Dukov JE, Chen HJ, Arata C, Hernandez-Trejo J, Xu P, Teng CS, Maxson RE, Crump JG. Cellular transitions during cranial suture establishment in zebrafish. Nat Commun 2024; 15:6948. [PMID: 39138165 PMCID: PMC11322166 DOI: 10.1038/s41467-024-50780-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
Cranial sutures separate neighboring skull bones and are sites of bone growth. A key question is how osteogenic activity is controlled to promote bone growth while preventing aberrant bone fusions during skull expansion. Using single-cell transcriptomics, lineage tracing, and mutant analysis in zebrafish, we uncover key developmental transitions regulating bone formation at sutures during skull expansion. In particular, we identify a subpopulation of mesenchyme cells in the mid-suture region that upregulate a suite of genes including BMP antagonists (e.g. grem1a) and pro-angiogenic factors. Lineage tracing with grem1a:nlsEOS reveals that this mid-suture subpopulation is largely non-osteogenic. Moreover, combinatorial mutation of BMP antagonists enriched in this mid-suture subpopulation results in increased BMP signaling in the suture, misregulated bone formation, and abnormal suture morphology. These data reveal establishment of a non-osteogenic mesenchyme population in the mid-suture region that restricts bone formation through local BMP antagonism, thus ensuring proper suture morphology.
Collapse
Affiliation(s)
- D'Juan T Farmer
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, 90095, USA.
| | - Jennifer E Dukov
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Hung-Jhen Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Claire Arata
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jose Hernandez-Trejo
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Pengfei Xu
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Camilla S Teng
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Robert E Maxson
- Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - J Gage Crump
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
7
|
Zeng T, Spence JP, Mostafavi H, Pritchard JK. Bayesian estimation of gene constraint from an evolutionary model with gene features. Nat Genet 2024; 56:1632-1643. [PMID: 38977852 DOI: 10.1038/s41588-024-01820-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 05/29/2024] [Indexed: 07/10/2024]
Abstract
Measures of selective constraint on genes have been used for many applications, including clinical interpretation of rare coding variants, disease gene discovery and studies of genome evolution. However, widely used metrics are severely underpowered at detecting constraints for the shortest ~25% of genes, potentially causing important pathogenic mutations to be overlooked. Here we developed a framework combining a population genetics model with machine learning on gene features to enable accurate inference of an interpretable constraint metric, shet. Our estimates outperform existing metrics for prioritizing genes important for cell essentiality, human disease and other phenotypes, especially for short genes. Our estimates of selective constraint should have wide utility for characterizing genes relevant to human disease. Finally, our inference framework, GeneBayes, provides a flexible platform that can improve the estimation of many gene-level properties, such as rare variant burden or gene expression differences.
Collapse
Affiliation(s)
- Tony Zeng
- Department of Genetics, Stanford University, Stanford, CA, USA.
| | | | - Hakhamanesh Mostafavi
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Population Health, New York University, New York, NY, USA
| | - Jonathan K Pritchard
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Department of Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Romeo DJ, Oral KT, Massenburg BB, Ng JJ, Wu M, Sussman JH, Du S, Bartlett SP, Swanson JW, Taylor JA. Genetic Heterogeneity, Craniofacial Surgical Burden, and Surgical Techniques in Patients With Saethre-Chotzen Syndrome. J Craniofac Surg 2024:00001665-990000000-01781. [PMID: 39058028 DOI: 10.1097/scs.0000000000010348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/03/2024] [Indexed: 07/28/2024] Open
Abstract
OBJECTIVE While genotype correlates with phenotype in patients with many forms of syndromic craniosynostosis, the relationship between molecular diagnosis and craniofacial surgical history in patients with Saethre-Chotzen syndrome (SCS) is more variable. This manuscript characterizes that relationship and evaluates operative trends in these patients over the past 3 decades. METHODS Demographic information, molecular diagnosis, and craniofacial surgical history in patients born with SCS between 1989 and 2023 were compared with appropriate statistics, including t tests and analysis of variance. RESULTS Thirty-five patients with SCS were included, and there was no difference in total craniofacial procedures among those with TWIST1 substitutions (2.1 ± 1.6), duplications (3.0 ± 4.2), insertions (3.5 ± 0.7), or deletions (2.4 ± 1.9; P = 0.97). Cranial expansion rates were also similar across all genetic diagnoses (P>0.05), and surgical incidence was similar across patients with unicoronal, bicoronal, and multisuture involvement (P > 0.05). Those with an initial fronto-orbital advancement had a lower incidence of secondary cranial vault procedures compared with those with an initial posterior vault distraction osteogenesis (29% versus 71%, P < 0.05), though this did not control for phenotypic severity. On average, total cranial vault surgical burden (1.35 ± 0.67 versus 1.75 ± 0.46) and cranial expansion surgical burden (1.40 ± 0.68 versus 1.88 ± 0.64) between the fronto-orbital advancement-first and posterior vault distraction osteogenesis-first cohorts were similar (P = 0.11, P = 0.17, respectively). CONCLUSION While SCS is molecularly and phenotypically heterogeneous, genetic diagnosis does not appear associated with rates of craniofacial surgery. Additional prospective study of correlations between genotype, severity of craniofacial manifestations, and treatment algorithms is warranted; but, in the end, it may be that this highly variable form of syndromic craniosynostosis warrants tailored, expectant management.
Collapse
Affiliation(s)
- Dominic J Romeo
- Division of Plastic, Reconstructive, and Oral Surgery, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Camp E, Garcia LG, Pribadi C, Paton S, Vasilev K, Anderson P, Gronthos S. Targeting of C-ROS-1 Activity Using a Controlled Release Carrier to Treat Craniosynostosis in a Preclinical Model of Saethre-Chotzen Syndrome. J Tissue Eng Regen Med 2024; 2024:8863925. [PMID: 40225751 PMCID: PMC11919205 DOI: 10.1155/2024/8863925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 04/15/2025]
Abstract
Saethre-Chotzen syndrome (SCS) is one of the most prevalent craniosynostosis, caused by a loss-of-function mutation in the TWIST-1 gene, with current treatment options relying on major invasive transcranial surgery. TWIST-1 haploinsufficient osteogenic progenitor cells exhibit increased osteogenic differentiation potential due to an upregulation of the transmembrane tyrosine kinase receptor, C-ROS-1, a TWIST-1 target gene known to promote bone formation. The present study assessed the efficacy of suppressing C-ROS-1 activity using a known chemical inhibitor to C-ROS-1, crizotinib, to halt premature coronal suture fusion in a preclinical mouse model of SCS. Crizotinib (1 μM, 2 μM, or 4 μM) was administered locally over the calvaria of Twist-1del/+ heterozygous mice prior to coronal suture fusion using either a nonresorbable collagen sponge (quick drug release) or a resorbable sodium carboxymethylcellulose microdisk (slow sustained release). Coronal suture fusion rates and bone parameters were determined by μCT imaging and histomorphometric analysis of calvaria postcoronal suture fusion. Results demonstrated a dose-dependent increase in the efficacy of crizotinib to maintain coronal suture patency, with no adverse effects to brain, kidney, liver, and spleen tissue, or blood cell parameters. Moreover, crizotinib delivered on microdisks resulted in a greater efficacy at a lower concentration to reduce bone formation at the coronal suture sites compared to sponges. However, the bone inhibitory effects were found to be diminished by over time following cessation of treatment. Our findings lay the foundation for the development of a pharmacological nonsurgical, targeted approach to temporarily maintain open coronal sutures in SCS patients. This study could potentially be used to develop similar therapeutic strategies to treat different syndromic craniosynostosis conditions caused by known genetic mutations.
Collapse
Affiliation(s)
- Esther Camp
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Laura Gonzalez Garcia
- School of Engineering and Future Industries Institute, Mawson Lakes Campus, University of South Australia, Mawson Lakes, SA, Australia
| | - Clara Pribadi
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Sharon Paton
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Krasimir Vasilev
- School of Engineering and Future Industries Institute, Mawson Lakes Campus, University of South Australia, Mawson Lakes, SA, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Peter Anderson
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Cleft & Craniofacial SA, Woman's and Children's Hospital, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
10
|
Zeng T, Spence JP, Mostafavi H, Pritchard JK. Bayesian estimation of gene constraint from an evolutionary model with gene features. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.19.541520. [PMID: 37292653 PMCID: PMC10245655 DOI: 10.1101/2023.05.19.541520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Measures of selective constraint on genes have been used for many applications including clinical interpretation of rare coding variants, disease gene discovery, and studies of genome evolution. However, widely-used metrics are severely underpowered at detecting constraint for the shortest ∼25% of genes, potentially causing important pathogenic mutations to be overlooked. We developed a framework combining a population genetics model with machine learning on gene features to enable accurate inference of an interpretable constraint metric, shet. Our estimates outperform existing metrics for prioritizing genes important for cell essentiality, human disease, and other phenotypes, especially for short genes. Our new estimates of selective constraint should have wide utility for characterizing genes relevant to human disease. Finally, our inference framework, GeneBayes, provides a flexible platform that can improve estimation of many gene-level properties, such as rare variant burden or gene expression differences.
Collapse
Affiliation(s)
- Tony Zeng
- Department of Genetics, Stanford University, Stanford CA
| | | | | | - Jonathan K. Pritchard
- Department of Genetics, Stanford University, Stanford CA
- Department of Biology, Stanford University, Stanford CA
| |
Collapse
|
11
|
Anjum AA, Lin MJ, Jin L, Li GQ. Twist is required for muscle development of the adult legs in Henosepilachna vigintioctopunctata. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2024; 115:e22063. [PMID: 37920138 DOI: 10.1002/arch.22063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023]
Abstract
Although muscle development has been widely studied in Drosophila melanogaster, it was a great challenge to apply to developmental processes of other insect muscles. This study was focused on the functional characterization of a basic helix-loop-helix transcription factor gene twist in an herbivorous ladybird Henosepilachna vigintioctopunctata. Its transcript (Hvtwist) levels were detected in all developmental stages. RNA interference (RNAi)-aided knockdown of Hvtwist at the penultimate larval instar stage impaired pupation, and caused a deformed adult in the legs. The tarsi were malformed and did not support the bodies in an upright position. The climbing ability was impaired. Moreover, around 50% of the impaired adults had a malformed elytrum. In addition, they consumed less foliage and did not lay eggs. A hematoxylin-eosin staining of the leg demonstrated that the tibial extensor (TE) and the tibial flexor (TF) muscles were originated from the femurs while levator and depressor muscles of the tarsus (TL and TD) were located in the tibia in the control adults, in which tarsal segments were devoid of muscles. RNAi treatment specific to Hvtwist expression markedly impaired TE and TF muscles in the femurs, and prevented the development of TL and TD muscles in the tibia. Therefore, our findings demonstrate Twist plays a vital role in the myogenesis in H. vigintioctopunctata adult legs.
Collapse
Affiliation(s)
- Ahmad Ali Anjum
- Department of Entomology, Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Meng-Jiao Lin
- Department of Entomology, Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Lin Jin
- Department of Entomology, Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Guo-Qing Li
- Department of Entomology, Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
12
|
Choi TM, Liu X, Abdel-Alim T, van Veelen ML, Mathijssen IMJ, Wolvius EB, Roshchupkin GV. Automated three-dimensional analysis of facial asymmetry in patients with syndromic coronal synostosis: A retrospective study. J Craniomaxillofac Surg 2024; 52:48-54. [PMID: 38135649 DOI: 10.1016/j.jcms.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/04/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Craniosynostosis, characterized by premature fusion of one or more cranial sutures, results in a distorted skull shape. Only three studies have assessed facial asymmetry manually in unicoronal synostosis patients. It is therefore important to understand how uni- and bicoronal synostosis affect facial asymmetry with a minimum risk of human bias. An automated algorithm was developed to quantify facial asymmetry from three-dimensional images, generating a mean facial asymmetry (MFA) value in millimeters to reflect the degree of asymmetry. The framework was applied to analyze postoperative 3D images of syndromic patients (N = 35) diagnosed with Muenke syndrome, Saethre-Chotzen syndrome, and TCF12-related craniosynostosis with respect to MFA values from a healthy control group (N = 89). Patients demonstrated substantially higher MFA values than controls: Muenke syndrome (unicoronal 1.74 ± 0.40 mm, bicoronal 0.77 ± 0.21 mm), Saethre-Chotzen syndrome (unicoronal 1.15 ± 0.20 mm, bicoronal 0.69 ± 0.16 mm), and TCF12-related craniosynostosis (unicoronal 1.40 ± 0.51 mm, bicoronal 0.66 ± 0.05 mm), compared with controls (0.49 ± 0.12 mm). Longitudinal analysis identified an increasing MFA trend in unicoronal synostosis patients. Our study revealed higher MFA in syndromic patients with uni- and bicoronal synostosis compared with controls, with the most pronounced MFA in Muenke syndrome patients with unilateral synostosis. Bicoronal synostosis patients demonstrated higher facial asymmetry than expected given the condition's symmetrical presentation.
Collapse
Affiliation(s)
- Tsun Man Choi
- Erasmus Medical Centre, Department of Oral Maxillofacial Surgery, Special Dental Care and Orthodontics, Dutch Craniofacial Centre, Rotterdam, the Netherlands.
| | - Xianjing Liu
- Erasmus Medical Centre, Department of Oral Maxillofacial Surgery, Special Dental Care and Orthodontics, Dutch Craniofacial Centre, Rotterdam, the Netherlands; Erasmus Medical Centre, Department of Radiology and Nuclear Medicine, Rotterdam, the Netherlands
| | - Tareq Abdel-Alim
- Erasmus Medical Centre, Department of Radiology and Nuclear Medicine, Rotterdam, the Netherlands; Erasmus Medical Centre, Department of Neurosurgery, Dutch Craniofacial Centre, Rotterdam, the Netherlands
| | - Marie-Lise van Veelen
- Erasmus Medical Centre, Department of Neurosurgery, Dutch Craniofacial Centre, Rotterdam, the Netherlands
| | - Irene Margreet Jacqueline Mathijssen
- Erasmus Medical Centre, Department of Plastic and Reconstructive Surgery and Hand Surgery, Dutch Craniofacial Centre, Rotterdam, the Netherlands
| | - Eppo Bonne Wolvius
- Erasmus Medical Centre, Department of Oral Maxillofacial Surgery, Special Dental Care and Orthodontics, Dutch Craniofacial Centre, Rotterdam, the Netherlands
| | - Gennady Vasilievich Roshchupkin
- Erasmus Medical Centre, Department of Radiology and Nuclear Medicine, Rotterdam, the Netherlands; Erasmus Medical Centre, Department of Epidemiology, Rotterdam, the Netherlands
| |
Collapse
|
13
|
Yang C, Ge Y, Zang Y, Xu M, Jin L, Wang Y, Xu X, Xue B, Wang Z, Wang L. CDC20 promotes radioresistance of prostate cancer by activating Twist1 expression. Apoptosis 2023; 28:1584-1595. [PMID: 37535214 DOI: 10.1007/s10495-023-01877-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2023] [Indexed: 08/04/2023]
Abstract
Currently, radiotherapy is one of the most attractive treatments for prostate cancer (PCa) patients. However, radioresistance remains a challenging issue and the underlying mechanism is unknown. Growing evidence has demonstrated that CDC20 (Cell division cycle protein 20) plays a pivotal role in a variety of tumors, including PCa. Here, GEPIA database mining and western blot analysis showed that higher expression of CDC20 was observed in PCa tissues and cells. We demonstrated that the expression of CDC20 was increased in PCa cells by irradiation, and knockdown of CDC20 resulted in inhibition of cell proliferation, migration, tumor formation, induced cell apoptosis and increased radiosensitivity in PCa in vitro and in vivo. Furthermore, we observed that CDC20 regulated Twist1 pathway, influencing cell proliferation and migration. These results suggest that targeting CDC20 and Twist1 may be an effective way to improve the radiosensitivity of PCa.
Collapse
Affiliation(s)
- Chuanlai Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Scientific Research Department, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yuegang Ge
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Institute of Radiotherapy and Oncology, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yachen Zang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Ming Xu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Lu Jin
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yang Wang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Xinyu Xu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Boxin Xue
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Zhiwei Wang
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, 233003, Anhui, China.
| | - Lixia Wang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
14
|
Yu X, He T, Tong Z, Liao L, Huang S, Fakhouri WD, Edwards DP, Xu J. Molecular mechanisms of TWIST1-regulated transcription in EMT and cancer metastasis. EMBO Rep 2023; 24:e56902. [PMID: 37680145 PMCID: PMC10626429 DOI: 10.15252/embr.202356902] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
TWIST1 induces epithelial-to-mesenchymal transition (EMT) to drive cancer metastasis. It is yet unclear what determines TWIST1 functions to activate or repress transcription. We found that the TWIST1 N-terminus antagonizes TWIST1-regulated gene expression, cancer growth and metastasis. TWIST1 interacts with both the NuRD complex and the NuA4/TIP60 complex (TIP60-Com) via its N-terminus. Non-acetylated TWIST1-K73/76 selectively interacts with and recruits NuRD to repress epithelial target gene transcription. Diacetylated TWIST1-acK73/76 binds BRD8, a component of TIP60-Com that also binds histone H4-acK5/8, to recruit TIP60-Com to activate mesenchymal target genes and MYC. Knockdown of BRD8 abolishes TWIST1 and TIP60-Com interaction and TIP60-Com recruitment to TWIST1-activated genes, resulting in decreasing TWIST1-activated target gene expression and cancer metastasis. Both TWIST1/NuRD and TWIST1/TIP60-Com complexes are required for TWIST1 to promote EMT, proliferation, and metastasis at full capacity. Therefore, the diacetylation status of TWIST1-K73/76 dictates whether TWIST1 interacts either with NuRD to repress epithelial genes, or with TIP60-Com to activate mesenchymal genes and MYC. Since BRD8 is essential for TWIST1-acK73/76 and TIP60-Com interaction, targeting BRD8 could be a means to inhibit TWIST1-activated gene expression.
Collapse
Affiliation(s)
- Xiaobin Yu
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| | - Tao He
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Present address:
Institute for Cancer MedicineSouthwest Medical UniversitySichuanChina
| | - Zhangwei Tong
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
| | - Lan Liao
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Shixia Huang
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Walid D Fakhouri
- Department of Diagnostic and Biomedical Sciences, Center for Craniofacial Research, School of DentistryUniversity of Texas Health Science Center at HoustonHoustonTXUSA
| | - Dean P Edwards
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| | - Jianming Xu
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTXUSA
- Dan L. Duncan Comprehensive Cancer CenterBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
15
|
Kurzbuch AR, Cooper B, Duncan C, Ellenbogen J, Richardson D, Sinha A, Weber A, Sithambaram S, Hennedige A, Parks C. Patient Tailored Surgery in Saethre-Chotzen Syndrome: Analysis of Reoperation for Intracranial Hypertension. J Craniofac Surg 2023; 34:2099-2103. [PMID: 37226293 DOI: 10.1097/scs.0000000000009429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/08/2023] [Indexed: 05/26/2023] Open
Abstract
Saethre-Chotzen syndrome (SCS) is a syndromic craniosynostosis with pathogenic variants in the TWIST1 gene showing a broad phenotypic spectrum. Controversies exist in the literature regarding surgical management with single one-stage versus patient-tailored surgery and the related reoperation rate for intracranial hypertension of up to 42%. At our center, SCS patients are offered patient-tailored surgery with single-stage fronto-orbital advancement and remodeling or fronto-orbital advancement and remodeling and posterior distraction in an individually determined order. The authors' database identified 35 confirmed SCS patients between 1999 and 2022. Involved sutures in craniosynostosis were left unicoronal (22.9%), bicoronal (22.9%), sagittal (8.6%), bicoronal and sagittal (5.7%), right unicoronal (2.9%), bicoronal and metopic (2.9%), bicoronal, sagittal and metopic (2.9%), and bilateral lambdoid (2.9%). There was pansynostosis in 8.6% and no craniosynostosis in 14.3% of the patients. Twenty-six patients, 10 females, and 16 males were operated on. Mean age at the first surgery was 1.70 years, and 3.86 years at the second surgery. Eleven of 26 patients had invasive intracranial pressure monitoring. Three patients presented with papilledema before the first surgery and 4 afterward. Four of the 26 operated patients were operated initially elsewhere. The other 22 patients were initially referred to our unit and underwent patient-tailored surgery. Nine of these patients (41%) had a second surgery, and 3 (14%) of them were because of raised intracranial pressure. Seven (27%) of all operated patients had a complication. Median follow-up was 13.98 years (range, 1.85-18.08). Patient-tailored surgery in a specialized center and long-term follow-up allow for a low reoperation rate for intracranial hypertension.
Collapse
Affiliation(s)
- Arthur R Kurzbuch
- Department of Neurosurgery, Craniofacial Unit, Alder Hey Children's NHS Foundation Trust
| | - Ben Cooper
- Department of Neurosurgery, Craniofacial Unit, Alder Hey Children's NHS Foundation Trust
| | - Christian Duncan
- Department of Maxillofacial and Craniofacial Surgery, Alder Hey Children's NHS Foundation Trust
| | - Jonathan Ellenbogen
- Department of Neurosurgery, Craniofacial Unit, Alder Hey Children's NHS Foundation Trust
| | - David Richardson
- Department of Maxillofacial and Craniofacial Surgery, Alder Hey Children's NHS Foundation Trust
| | - Ajay Sinha
- Department of Neurosurgery, Craniofacial Unit, Alder Hey Children's NHS Foundation Trust
| | - Astrid Weber
- Liverpool Centre for Genomic Medicine, Liverpool Women's NHS Foundation Trust
| | | | - Anusha Hennedige
- Department of Maxillofacial and Craniofacial Surgery, Alder Hey Children's NHS Foundation Trust
| | - Chris Parks
- Department of Neurosurgery, Craniofacial Unit, Alder Hey Children's NHS Foundation Trust
| |
Collapse
|
16
|
Bok S, Yallowitz AR, Sun J, McCormick J, Cung M, Hu L, Lalani S, Li Z, Sosa BR, Baumgartner T, Byrne P, Zhang T, Morse KW, Mohamed FF, Ge C, Franceschi RT, Cowling RT, Greenberg BH, Pisapia DJ, Imahiyerobo TA, Lakhani S, Ross ME, Hoffman CE, Debnath S, Greenblatt MB. A multi-stem cell basis for craniosynostosis and calvarial mineralization. Nature 2023; 621:804-812. [PMID: 37730988 PMCID: PMC10799660 DOI: 10.1038/s41586-023-06526-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 08/09/2023] [Indexed: 09/22/2023]
Abstract
Craniosynostosis is a group of disorders of premature calvarial suture fusion. The identity of the calvarial stem cells (CSCs) that produce fusion-driving osteoblasts in craniosynostosis remains poorly understood. Here we show that both physiologic calvarial mineralization and pathologic calvarial fusion in craniosynostosis reflect the interaction of two separate stem cell lineages; a previously identified cathepsin K (CTSK) lineage CSC1 (CTSK+ CSC) and a separate discoidin domain-containing receptor 2 (DDR2) lineage stem cell (DDR2+ CSC) that we identified in this study. Deletion of Twist1, a gene associated with craniosynostosis in humans2,3, solely in CTSK+ CSCs is sufficient to drive craniosynostosis in mice, but the sites that are destined to fuse exhibit an unexpected depletion of CTSK+ CSCs and a corresponding expansion of DDR2+ CSCs, with DDR2+ CSC expansion being a direct maladaptive response to CTSK+ CSC depletion. DDR2+ CSCs display full stemness features, and our results establish the presence of two distinct stem cell lineages in the sutures, with both populations contributing to physiologic calvarial mineralization. DDR2+ CSCs mediate a distinct form of endochondral ossification without the typical haematopoietic marrow formation. Implantation of DDR2+ CSCs into suture sites is sufficient to induce fusion, and this phenotype was prevented by co-transplantation of CTSK+ CSCs. Finally, the human counterparts of DDR2+ CSCs and CTSK+ CSCs display conserved functional properties in xenograft assays. The interaction between these two stem cell populations provides a new biologic interface for the modulation of calvarial mineralization and suture patency.
Collapse
Affiliation(s)
- Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jason McCormick
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Michelle Cung
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lingling Hu
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
| | - Sarfaraz Lalani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Zan Li
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Branden R Sosa
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Tomas Baumgartner
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Paul Byrne
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Kyle W Morse
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
| | - Fatma F Mohamed
- Department of Periodontics, Prevention and Geriatrics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Chunxi Ge
- Department of Periodontics, Prevention and Geriatrics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Renny T Franceschi
- Department of Periodontics, Prevention and Geriatrics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Randy T Cowling
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, CA, USA
| | - Barry H Greenberg
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, CA, USA
| | - David J Pisapia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Thomas A Imahiyerobo
- Division of Plastic Surgery, Department of Surgery, New York-Presbyterian Hospital and Columbia University Medical Center, New York, NY, USA
| | - Shenela Lakhani
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Caitlin E Hoffman
- Department of Neurological Surgery, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, NY, USA
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Research Division, Hospital for Special Surgery, New York, NY, USA.
| |
Collapse
|
17
|
Luzzio A, Edie S, Palmer K, Caddle LB, Urban R, Goodwin LO, Welsh IC, Reinholdt LG, Bergstrom DE, Cox TC, Donahue LR, Murray SA. The spontaneous mouse mutant low set ears (Lse) is caused by tandem duplication of Fgf3 and Fgf4. Mamm Genome 2023; 34:453-463. [PMID: 37341808 PMCID: PMC11601887 DOI: 10.1007/s00335-023-09999-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 05/18/2023] [Indexed: 06/22/2023]
Abstract
The external ear develops from an organized convergence of ventrally migrating neural crest cells into the first and second branchial arches. Defects in external ear position are often symptomatic of complex syndromes such as Apert, Treacher-Collins, and Crouzon Syndrome. The low set ears (Lse) spontaneous mouse mutant is characterized by the dominant inheritance of a ventrally shifted external ear position and an abnormal external auditory meatus (EAM). We identified the causative mutation as a 148 Kb tandem duplication on Chromosome 7, which includes the entire coding sequences of Fgf3 and Fgf4. Duplications of FGF3 and FGF4 occur in 11q duplication syndrome in humans and are associated with craniofacial anomalies, among other features. Intercrosses of Lse-affected mice revealed perinatal lethality in homozygotes, and Lse/Lse embryos display additional phenotypes including polydactyly, abnormal eye morphology, and cleft secondary palate. The duplication results in increased Fgf3 and Fgf4 expression in the branchial arches and additional discrete domains in the developing embryo. This ectopic overexpression resulted in functional FGF signaling, demonstrated by increased Spry2 and Etv5 expression in overlapping domains of the developing arches. Finally, a genetic interaction between Fgf3/4 overexpression and Twist1, a regulator of skull suture development, resulted in perinatal lethality, cleft palate, and polydactyly in compound heterozygotes. These data indicate a role for Fgf3 and Fgf4 in external ear and palate development and provide a novel mouse model for further interrogation of the biological consequences of human FGF3/4 duplication.
Collapse
Affiliation(s)
| | - Sarah Edie
- The Jackson Laboratory, Bar Harbor, ME, USA
| | | | | | | | | | | | | | | | - Timothy C Cox
- Departments of Oral & Craniofacial Sciences and Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA
| | | | | |
Collapse
|
18
|
Haerinck J, Goossens S, Berx G. The epithelial-mesenchymal plasticity landscape: principles of design and mechanisms of regulation. Nat Rev Genet 2023; 24:590-609. [PMID: 37169858 DOI: 10.1038/s41576-023-00601-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/13/2023]
Abstract
Epithelial-mesenchymal plasticity (EMP) enables cells to interconvert between several states across the epithelial-mesenchymal landscape, thereby acquiring hybrid epithelial/mesenchymal phenotypic features. This plasticity is crucial for embryonic development and wound healing, but also underlies the acquisition of several malignant traits during cancer progression. Recent research using systems biology and single-cell profiling methods has provided novel insights into the main forces that shape EMP, which include the microenvironment, lineage specification and cell identity, and the genome. Additionally, key roles have emerged for hysteresis (cell memory) and cellular noise, which can drive stochastic transitions between cell states. Here, we review these forces and the distinct but interwoven layers of regulatory control that stabilize EMP states or facilitate epithelial-mesenchymal transitions (EMTs) and discuss the therapeutic potential of manipulating the EMP landscape.
Collapse
Affiliation(s)
- Jef Haerinck
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Unit for Translational Research in Oncology, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
19
|
Casasnovas-Nieves JJ, Rodríguez Y, Franco HL, Cadilla CL. Mechanisms of Regulation of the CHRDL1 Gene by the TWIST2 and ADD1/SREBP1c Transcription Factors. Genes (Basel) 2023; 14:1733. [PMID: 37761873 PMCID: PMC10530651 DOI: 10.3390/genes14091733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Setleis syndrome (SS) is a rare focal facial dermal dysplasia caused by recessive mutations in the basic helix-loop-helix (bHLH) transcription factor, TWIST2. Expression microarray analysis showed that the chordin-like 1 (CHRDL1) gene is up-regulated in dermal fibroblasts from three SS patients with the Q119X TWIST2 mutation. METHODS Putative TWIST binding sites were found in the upstream region of the CHRDL1 gene and examined by electrophoretic mobility shift (EMSA) and reporter gene assays. RESULTS EMSAs showed specific binding of TWIST1 and TWIST2 homodimers, as well as heterodimers with E12, to the more distal E-boxes. An adjoining E-box was bound by ADD1/SREBP1c. EMSA analysis suggested that TWIST2 and ADD1/SREBP1c could compete for binding. Luciferase (luc) reporter assays revealed that the CHRDL1 gene upstream region drives its expression and ADD1/SREBP1c increased it 2.6 times over basal levels. TWIST2, but not the TWIST2-Q119X mutant, blocked activation by ADD1/SREBP1c, but overexpression of TWIST2-Q119X increased luc gene expression. In addition, EMSA competition assays showed that TWIST2, but not TWIST1, competes with ADD1/SREBP1c for DNA binding to the same site. CONCLUSIONS Formation of an inactive complex between the TWIST2 Q119X and Q65X mutant proteins and ADD1/SREBP1c may prevent repressor binding and allow the binding of other regulators to activate CHRDL1 gene expression.
Collapse
Affiliation(s)
- José J. Casasnovas-Nieves
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.J.C.-N.); (Y.R.); (H.L.F.)
| | - Yacidzohara Rodríguez
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.J.C.-N.); (Y.R.); (H.L.F.)
| | - Hector L. Franco
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.J.C.-N.); (Y.R.); (H.L.F.)
- Department of Genetics, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carmen L. Cadilla
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.J.C.-N.); (Y.R.); (H.L.F.)
| |
Collapse
|
20
|
Radhakrishnan K, Truong L, Carmichael CL. An "unexpected" role for EMT transcription factors in hematological development and malignancy. Front Immunol 2023; 14:1207360. [PMID: 37600794 PMCID: PMC10435889 DOI: 10.3389/fimmu.2023.1207360] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a fundamental developmental process essential for normal embryonic development. It is also important during various pathogenic processes including fibrosis, wound healing and epithelial cancer cell metastasis and invasion. EMT is regulated by a variety of cell signalling pathways, cell-cell interactions and microenvironmental cues, however the key drivers of EMT are transcription factors of the ZEB, TWIST and SNAIL families. Recently, novel and unexpected roles for these EMT transcription factors (EMT-TFs) during normal blood cell development have emerged, which appear to be largely independent of classical EMT processes. Furthermore, EMT-TFs have also begun to be implicated in the development and pathogenesis of malignant hematological diseases such as leukemia and lymphoma, and now present themselves or the pathways they regulate as possible new therapeutic targets within these malignancies. In this review, we discuss the ZEB, TWIST and SNAIL families of EMT-TFs, focusing on what is known about their normal roles during hematopoiesis as well as the emerging and "unexpected" contribution they play during development and progression of blood cancers.
Collapse
Affiliation(s)
- Karthika Radhakrishnan
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Lynda Truong
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Catherine L. Carmichael
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Monash University, Faculty of Medicine, Nursing and Health Sciences, Clayton, VIC, Australia
| |
Collapse
|
21
|
Vesuna F, Penet MF, Mori N, Bhujwalla ZM, Raman V. Twist alters the breast tumor microenvironment via choline kinase to facilitate an aggressive phenotype. Mol Cell Biochem 2023; 478:939-948. [PMID: 36136285 PMCID: PMC11299248 DOI: 10.1007/s11010-022-04555-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022]
Abstract
Twist (TWIST1) is a gene required for cell fate specification in embryos and its expression in mammary epithelium can initiate tumorigenesis through the epithelial-mesenchymal transition. To identify downstream target genes of Twist in breast cancer, we performed microarray analysis on the transgenic breast cancer cell line, MCF-7/Twist. One of the targets identified was choline kinase whose upregulation resulted in increased cellular phosphocholine and total choline containing compounds-a characteristic observed in highly aggressive metastatic cancers. To study the interactions between Twist, choline kinase, and their effect on the microenvironment, we used 1H magnetic resonance spectroscopy and found significantly higher phosphocholine and total choline, as well as increased phosphocholine/glycerophosphocholine ratio in MCF-7/Twist cells. We also observed significant increases in extracellular glucose, lactate, and [H +] ion concentrations in the MCF-7/Twist cells. Magnetic resonance imaging of MCF-7/Twist orthotopic breast tumors showed a significant increase in vascular volume and permeability surface area product compared to control tumors. In addition, by reverse transcription-quantitative polymerase chain reaction, we discovered that Twist upregulated choline kinase expression in estrogen receptor negative breast cancer cell lines through FOXA1 downregulation. Moreover, using The Cancer Genome Atlas database, we observed a significant inverse relationship between FOXA1 and choline kinase expression and propose that it could act as a modulator of the Twist/choline kinase axis. The data presented indicate that Twist is a driver of choline kinase expression in breast cancer cells via FOXA1 resulting in the generation of an aggressive breast cancer phenotype.
Collapse
Affiliation(s)
- Farhad Vesuna
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marie-France Penet
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Noriko Mori
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Venu Raman
- Division of Cancer Imaging Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Department of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Hoshino Y, Takechi M, Moazen M, Steacy M, Koyabu D, Furutera T, Ninomiya Y, Nuri T, Pauws E, Iseki S. Synchondrosis fusion contributes to the progression of postnatal craniofacial dysmorphology in syndromic craniosynostosis. J Anat 2023; 242:387-401. [PMID: 36394990 PMCID: PMC9919486 DOI: 10.1111/joa.13790] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/16/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
Syndromic craniosynostosis (CS) patients exhibit early, bony fusion of calvarial sutures and cranial synchondroses, resulting in craniofacial dysmorphology. In this study, we chronologically evaluated skull morphology change after abnormal fusion of the sutures and synchondroses in mouse models of syndromic CS for further understanding of the disease. We found fusion of the inter-sphenoid synchondrosis (ISS) in Apert syndrome model mice (Fgfr2S252W/+ ) around 3 weeks old as seen in Crouzon syndrome model mice (Fgfr2cC342Y/+ ). We then examined ontogenic trajectories of CS mouse models after 3 weeks of age using geometric morphometrics analyses. Antero-ventral growth of the face was affected in Fgfr2S252W/+ and Fgfr2cC342Y/+ mice, while Saethre-Chotzen syndrome model mice (Twist1+/- ) did not show the ISS fusion and exhibited a similar growth pattern to that of control littermates. Further analysis revealed that the coronal suture synostosis in the CS mouse models induces only the brachycephalic phenotype as a shared morphological feature. Although previous studies suggest that the fusion of the facial sutures during neonatal period is associated with midface hypoplasia, the present study suggests that the progressive postnatal fusion of the cranial synchondrosis also contributes to craniofacial dysmorphology in mouse models of syndromic CS. These morphological trajectories increase our understanding of the progression of syndromic CS skull growth.
Collapse
Affiliation(s)
- Yukiko Hoshino
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- Office of New Drug V, Pharmaceuticals and Medical Devices Agency (PMDA)TokyoJapan
| | - Masaki Takechi
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- Department of Anatomy and Life StructureJuntendo University Graduate School of MedicineTokyoJapan
| | - Mehran Moazen
- Department of UCL Mechanical EngineeringUniversity College LondonLondonUK
| | - Miranda Steacy
- Institute of Child Health, Great Ormond StreetUniversity College LondonLondonUK
| | - Daisuke Koyabu
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- Research and Development Center for Precision MedicineTsukuba UniversityTsukubaJapan
| | - Toshiko Furutera
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
- Department of Anatomy and Life StructureJuntendo University Graduate School of MedicineTokyoJapan
| | - Youichirou Ninomiya
- Research Organization of Information and SystemsNational Institute of InformaticsTokyoJapan
| | - Takashi Nuri
- Department of Plastic and Reconstructive SurgeryOsaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Erwin Pauws
- Institute of Child Health, Great Ormond StreetUniversity College LondonLondonUK
| | - Sachiko Iseki
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| |
Collapse
|
23
|
Diaz-Gonzalez F, Sacedo-Gutiérrez JM, Twigg SRF, Calpena E, Carceller-Benito FE, Parrón-Pajares M, Santos-Simarro F, Heath KE. Case report: A third variant in the 5' UTR of TWIST1 creates a novel upstream translation initiation site in a child with Saethre-Chotzen syndrome. Front Genet 2023; 13:1089417. [PMID: 36685936 PMCID: PMC9845400 DOI: 10.3389/fgene.2022.1089417] [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: 11/04/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction: Saethre-Chotzen syndrome, a craniosynostosis syndrome characterized by the premature closure of the coronal sutures, dysmorphic facial features and limb anomalies, is caused by haploinsufficiency of TWIST1. Although the majority of variants localize in the coding region of the gene, two variants in the 5' UTR have been recently reported to generate novel upstream initiation codons. Methods: Skeletal dysplasia Next-generation sequencing (NGS) panel was used for genetic analysis in a patient with bicoronal synostosis, facial dysmorphisms and limb anomalies. The variant pathogenicity was assessed by a luciferase reporter promoter assay. Results: Here, we describe the identification of a third ATG-creating de novo variant, c.-18C>T, in the 5' UTR of TWIST1 in the patient with a clinical diagnosis of Saethre-Chotzen syndrome. It was predicted to create an out-of-frame new upstream translation initiation codon resulting in a 40 amino acid larger functionally inactive protein. We performed luciferase reporter promoter assays to demonstrate that the variant does indeed reduce translation from the main open reading frame. Conclusion: This is the third variant identified in this region and confirms the introduction of upstream ATGs in the 5' UTR of TWIST1 as a pathogenic mechanism in Saethre-Chotzen syndrome. This case report shows the necessity for performing functional characterization of variants of unknown significance within national health services.
Collapse
Affiliation(s)
- Francisca Diaz-Gonzalez
- Institute of Medical & Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain,Skeletal Dysplasia Multidisciplinary Unit (UMDE) and ERN-BOND, Hospital Universitario La Paz, Madrid, Spain
| | - Javier M. Sacedo-Gutiérrez
- Skeletal Dysplasia Multidisciplinary Unit (UMDE) and ERN-BOND, Hospital Universitario La Paz, Madrid, Spain,Department of Neurosurgery, Hospital Universitario la Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain
| | - Stephen R. F. Twigg
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Eduardo Calpena
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Fernando E. Carceller-Benito
- Skeletal Dysplasia Multidisciplinary Unit (UMDE) and ERN-BOND, Hospital Universitario La Paz, Madrid, Spain,Department of Neurosurgery, Hospital Universitario la Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain
| | - Manuel Parrón-Pajares
- Skeletal Dysplasia Multidisciplinary Unit (UMDE) and ERN-BOND, Hospital Universitario La Paz, Madrid, Spain,Department of Radiology, Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Fernando Santos-Simarro
- Institute of Medical & Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain,Skeletal Dysplasia Multidisciplinary Unit (UMDE) and ERN-BOND, Hospital Universitario La Paz, Madrid, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U753), Instituto Carlos III, Madrid, Spain
| | - Karen E. Heath
- Institute of Medical & Molecular Genetics (INGEMM), Hospital Universitario La Paz, Universidad Autónoma de Madrid, IdiPAZ, Madrid, Spain,Skeletal Dysplasia Multidisciplinary Unit (UMDE) and ERN-BOND, Hospital Universitario La Paz, Madrid, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U753), Instituto Carlos III, Madrid, Spain,*Correspondence: Karen E. Heath,
| |
Collapse
|
24
|
Luyckx I, Verstraeten A, Goumans MJ, Loeys B. SMAD6-deficiency in human genetic disorders. NPJ Genom Med 2022; 7:68. [DOI: 10.1038/s41525-022-00338-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
AbstractSMAD6 encodes an intracellular inhibitor of the bone morphogenetic protein (BMP) signalling pathway. Until now, SMAD6-deficiency has been associated with three distinctive human congenital conditions, i.e., congenital heart diseases, including left ventricular obstruction and conotruncal defects, craniosynostosis and radioulnar synostosis. Intriguingly, a similar spectrum of heterozygous loss-of-function variants has been reported to cause these clinically distinct disorders without a genotype–phenotype correlation. Even identical nucleotide changes have been described in patients with either a cardiovascular phenotype, craniosynostosis or radioulnar synostosis. These findings suggest that the primary pathogenic variant alone cannot explain the resultant patient phenotype. In this review, we summarise clinical and (patho)genetic (dis)similarities between these three SMAD6-related conditions, compare published Madh6 mouse models, in which the importance and impact of the genetic background with respect to the observed phenotype is highlighted, and elaborate on the cellular key mechanisms orchestrated by SMAD6 in the development of these three discrete inherited disorders. In addition, we discuss future research needed to elucidate the pathogenetic mechanisms underlying these diseases in order to improve their molecular diagnosis, advance therapeutic strategies and facilitate counselling of patients and their families.
Collapse
|
25
|
Ang PS, Matrongolo MJ, Zietowski ML, Nathan SL, Reid RR, Tischfield MA. Cranium growth, patterning and homeostasis. Development 2022; 149:dev201017. [PMID: 36408946 PMCID: PMC9793421 DOI: 10.1242/dev.201017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Craniofacial development requires precise spatiotemporal regulation of multiple signaling pathways that crosstalk to coordinate the growth and patterning of the skull with surrounding tissues. Recent insights into these signaling pathways and previously uncharacterized progenitor cell populations have refined our understanding of skull patterning, bone mineralization and tissue homeostasis. Here, we touch upon classical studies and recent advances with an emphasis on developmental and signaling mechanisms that regulate the osteoblast lineage for the calvaria, which forms the roof of the skull. We highlight studies that illustrate the roles of osteoprogenitor cells and cranial suture-derived stem cells for proper calvarial growth and homeostasis. We also discuss genes and signaling pathways that control suture patency and highlight how perturbing the molecular regulation of these pathways leads to craniosynostosis. Finally, we discuss the recently discovered tissue and signaling interactions that integrate skull and cerebrovascular development, and the potential implications for both cerebrospinal fluid hydrodynamics and brain waste clearance in craniosynostosis.
Collapse
Affiliation(s)
- Phillip S. Ang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Matt J. Matrongolo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | | | - Shelby L. Nathan
- Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Russell R. Reid
- Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Max A. Tischfield
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
26
|
The speckle-type POZ protein (SPOP) inhibits breast cancer malignancy by destabilizing TWIST1. Cell Death Dis 2022; 8:389. [PMID: 36115849 PMCID: PMC9482615 DOI: 10.1038/s41420-022-01182-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 12/21/2022]
Abstract
Epithelial-mesenchymal transition (EMT) inducing transcription factor TWIST1 plays a vital role in cancer metastasis. How the tumor-suppressive E3 ligase, speckle-type POZ protein (SPOP), regulates TWIST1 in breast cancer remains unknown. In this study, we report that SPOP physically interacts with, ubiquitinates, and destabilizes TWIST1. SPOP promotes K63-and K48-linked ubiquitination of TWIST1, predominantly at K73, thereby suppressing cancer cell migration and invasion. Silencing SPOP significantly enhances EMT, which accelerates breast cancer cell migration and invasiveness in vitro and lung metastasis in vivo. Clinically, SPOP is negatively correlated with the levels of TWIST1 in highly invasive breast carcinomas. Reduced SPOP expression, along with elevated TWIST1 levels, is associated with poor prognosis in advanced breast cancer patients, particularly those with metastatic triple-negative breast cancer (TNBC). Taken together, we have disclosed a new mechanism linking SPOP to TWIST1 degradation. Thus SPOP may serve as a prognostic marker and a potential therapeutic target for advanced TNBC patients.
Collapse
|
27
|
Bertol JW, Johnston S, Ahmed R, Xie VK, Hubka KM, Cruz L, Nitschke L, Stetsiv M, Goering JP, Nistor P, Lowell S, Hoskens H, Claes P, Weinberg SM, Saadi I, Farach-Carson MC, Fakhouri WD. TWIST1 interacts with β/δ-catenins during neural tube development and regulates fate transition in cranial neural crest cells. Development 2022; 149:dev200068. [PMID: 35781329 PMCID: PMC9440756 DOI: 10.1242/dev.200068] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 05/30/2022] [Indexed: 08/10/2023]
Abstract
Cell fate determination is a necessary and tightly regulated process for producing different cell types and structures during development. Cranial neural crest cells (CNCCs) are unique to vertebrate embryos and emerge from the neural plate borders into multiple cell lineages that differentiate into bone, cartilage, neurons and glial cells. We have previously reported that Irf6 genetically interacts with Twist1 during CNCC-derived tissue formation. Here, we have investigated the mechanistic role of Twist1 and Irf6 at early stages of craniofacial development. Our data indicate that TWIST1 is expressed in endocytic vesicles at the apical surface and interacts with β/δ-catenins during neural tube closure, and Irf6 is involved in defining neural fold borders by restricting AP2α expression. Twist1 suppresses Irf6 and other epithelial genes in CNCCs during the epithelial-to-mesenchymal transition (EMT) process and cell migration. Conversely, a loss of Twist1 leads to a sustained expression of epithelial and cell adhesion markers in migratory CNCCs. Disruption of TWIST1 phosphorylation in vivo leads to epidermal blebbing, edema, neural tube defects and CNCC-derived structural abnormalities. Altogether, this study describes a previously uncharacterized function of mammalian Twist1 and Irf6 in the neural tube and CNCCs, and provides new target genes for Twist1 that are involved in cytoskeletal remodeling.
Collapse
Affiliation(s)
- Jessica W. Bertol
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Shelby Johnston
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Rabia Ahmed
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Victoria K. Xie
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Kelsea M. Hubka
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Lissette Cruz
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Larissa Nitschke
- Department of Pathology and Immunology,Baylor College of Medicine, Houston, TX 77030, USA
| | - Marta Stetsiv
- Department of Anatomy and Cell Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jeremy P. Goering
- Department of Anatomy and Cell Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Paul Nistor
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Little France Drive, Edinburgh EH16 4UU, UK
| | - Sally Lowell
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Little France Drive, Edinburgh EH16 4UU, UK
| | - Hanne Hoskens
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven 3001, Belgium
- Medical Imaging Research Center, UZ Leuven, Leuven 3000, Belgium
| | - Peter Claes
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven 3001, Belgium
- Medical Imaging Research Center, UZ Leuven, Leuven 3000, Belgium
- Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Seth M. Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Irfan Saadi
- Department of Anatomy and Cell Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Mary C. Farach-Carson
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Walid D. Fakhouri
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
28
|
Kalmar CL, Lang SS, Heuer GG, Schreiber JE, Tucker AM, Swanson JW, Beslow LA. Neurocognitive outcomes of children with non-syndromic single-suture craniosynostosis. Childs Nerv Syst 2022; 38:893-901. [PMID: 35192026 DOI: 10.1007/s00381-022-05448-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/06/2022] [Indexed: 01/17/2023]
Abstract
While the focus of craniosynostosis surgery is to improve head shape, neurocognitive sequelae are common and are incompletely understood. Neurodevelopmental problems that children with craniosynostosis face include cognitive and language impairments, motor delays or deficits, learning disabilities, executive dysfunction, and behavioral problems. Studies have shown that children with multiple suture craniosynostosis have more impairment than children with single-suture craniosynostosis. Children with isolated single-suture subtypes of craniosynostosis such as sagittal, metopic, and unicoronal craniosynostosis can have distinct neurocognitive profiles. In this review, we discuss the unique neurodevelopmental profiles of children with single-suture subtypes of craniosynostosis.
Collapse
Affiliation(s)
- Christopher L Kalmar
- Division of Plastic and Reconstructive Surgery, Children's Hospital of Philadelphia, PA, Philadelphia, USA
| | - Shih-Shan Lang
- Division of Neurosurgery, Children's Hospital of Philadelphia, PA, Philadelphia, USA.,Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, PA, Philadelphia, USA
| | - Gregory G Heuer
- Division of Neurosurgery, Children's Hospital of Philadelphia, PA, Philadelphia, USA.,Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, PA, Philadelphia, USA
| | - Jane E Schreiber
- Department of Child & Adolescent Psychiatry and Behavioral Sciences, Children's Hospital of Philadelphia, PA, Philadelphia, USA.,Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, PA, Philadelphia, USA
| | - Alexander M Tucker
- Division of Neurosurgery, Children's Hospital of Philadelphia, PA, Philadelphia, USA
| | - Jordan W Swanson
- Division of Plastic and Reconstructive Surgery, Children's Hospital of Philadelphia, PA, Philadelphia, USA.,Division of Plastic Surgery, Perelman School of Medicine at the University of Pennsylvania, PA, Philadelphia, USA
| | - Lauren A Beslow
- Division of Neurology, Children's Hospital of Philadelphia, PA, Philadelphia, USA. .,Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, PA, Philadelphia, USA. .,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, PA, Philadelphia, USA.
| |
Collapse
|
29
|
Kemppainen AV, Finnilä MA, Heikkinen A, Härönen H, Izzi V, Kauppinen S, Saarakkala S, Pihlajaniemi T, Koivunen J. The CMS19 disease model specifies a pivotal role for collagen XIII in bone homeostasis. Sci Rep 2022; 12:5866. [PMID: 35393492 PMCID: PMC8990013 DOI: 10.1038/s41598-022-09653-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Mutations in the COL13A1 gene result in congenital myasthenic syndrome type 19 (CMS19), a disease of neuromuscular synapses and including various skeletal manifestations, particularly facial dysmorphisms. The phenotypic consequences in Col13a1 null mice (Col13a1−/−) recapitulate the muscle findings of the CMS19 patients. Collagen XIII (ColXIII) is exists as two forms, a transmembrane protein and a soluble molecule. While the Col13a1−/− mice have poorly formed neuromuscular junctions, the prevention of shedding of the ColXIII ectodomain in the Col13a1tm/tm mice results in acetylcholine receptor clusters of increased size and complexity. In view of the bone abnormalities in CMS19, we here studied the tubular and calvarial bone morphology of the Col13a1−/− mice. We discovered several craniofacial malformations, albeit less pronounced ones than in the human disease, and a reduction of cortical bone mass in aged mice. In the Col13a1tm/tm mice, where ColXIII is synthesized but the ectodomain shedding is prevented due to a mutation in a protease recognition sequence, the cortical bone mass decreased as well with age and the cephalometric analyses revealed significant craniofacial abnormalities but no clear phenotypical pattern. To conclude, our data indicates an intrinsic role for ColXIII, particularly the soluble form, in the upkeep of bone with aging and suggests the possibility of previously undiscovered bone pathologies in patients with CMS19.
Collapse
Affiliation(s)
- A V Kemppainen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland
| | - M A Finnilä
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - A Heikkinen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland
| | - H Härönen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland
| | - V Izzi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland.,Faculty of Medicine, University of Oulu, 90014, Oulu, Finland.,Foundation for the Finnish Cancer Institute, Tukholmankatu 8, 00130, Helsinki, Finland
| | - S Kauppinen
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - S Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.,Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - T Pihlajaniemi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland
| | - J Koivunen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5400, 90014, Oulu, Finland.
| |
Collapse
|
30
|
Alawneh RJ, Johnson AL, Hoover-Fong JE, Jackson EM, Steinberg JP, MacCarrick G. Postnatal Progressive Craniosynostosis in Syndromic Conditions: Two Patients With Saethre-Chotzen Due to TWIST1 Gene Deletions and Review of the Literature. Cleft Palate Craniofac J 2022:10556656221090844. [PMID: 35354337 DOI: 10.1177/10556656221090844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Saethre-Chotzen syndrome (SCS) is a known craniosynostosis syndrome with a variable presentation of craniofacial and somatic involvement. Congenital coronal craniosynostosis is most commonly observed in SCS; however, progressive postnatal craniosynostosis of other sutures has been reported. The authors present 2 infants with progressive postnatal craniosynostosis and SCS caused by chromosome 7p deletions including the TWIST1 gene. The evolution of their clinical features and a literature review of patients with syndromic, postnatal progressive craniosynostosis illustrate the importance of longitudinal observation and management of these patients.
Collapse
Affiliation(s)
- Rama J Alawneh
- Faculty of Medicine, King Abdullah University Hospital, 37251Jordan University of Science and Technology, Irbid, Jordan
| | - Andrea L Johnson
- Department of Cellular Biology and Molecular Genetics, 1068University of Maryland College Park, College Park, MD, USA
| | - Julie Elizabeth Hoover-Fong
- Greenberg Center for Skeletal Dysplasias, Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Eric M Jackson
- Department of Neurosurgery, 1500Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jordan P Steinberg
- Division of Plastic Surgery, Nicklaus Children's Hospital, Miami, FL, USA
| | - Gretchen MacCarrick
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
31
|
Quarto N, Menon S, Griffin M, Huber J, Longaker MT. Harnessing a Feasible and Versatile ex vivo Calvarial Suture 2-D Culture System to Study Suture Biology. Front Physiol 2022; 13:823661. [PMID: 35222087 PMCID: PMC8871685 DOI: 10.3389/fphys.2022.823661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
As a basic science, craniofacial research embraces multiple facets spanning from molecular regulation of craniofacial development, cell biology/signaling and ultimately translational craniofacial biology. Calvarial sutures coordinate development of the skull, and the premature fusion of one or more, leads to craniosynostosis. Animal models provide significant contributions toward craniofacial biology and clinical/surgical treatments of patients with craniofacial disorders. Studies employing mouse models are costly and time consuming for housing/breeding. Herein, we present the establishment of a calvarial suture explant 2-D culture method that has been proven to be a reliable system showing fidelity with the in vivo harvesting procedure to isolate high yields of skeletal stem/progenitor cells from small number of mice. Moreover, this method allows the opportunity to phenocopying models of craniosynostosis and in vitro tamoxifen-induction of ActincreERT2;R26Rainbow suture explants to trace clonal expansion. This versatile method tackles needs of large number of mice to perform calvarial suture research.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli Federico II, Naples, Italy
- *Correspondence: Natalina Quarto,
| | - Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Michelle Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Julika Huber
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Plastic Surgery, BG University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Michael T. Longaker,
| |
Collapse
|
32
|
Tønne E, Due-Tønnessen BJ, Vigeland MD, Amundsen SS, Ribarska T, Åsten PM, Sheng Y, Helseth E, Gilfillan GD, Mero IL, Heimdal KR. Whole-exome sequencing in syndromic craniosynostosis increases diagnostic yield and identifies candidate genes in osteogenic signaling pathways. Am J Med Genet A 2022; 188:1464-1475. [PMID: 35080095 DOI: 10.1002/ajmg.a.62663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/26/2021] [Accepted: 12/26/2021] [Indexed: 11/07/2022]
Abstract
Craniosynostosis (CS) is a common congenital anomaly defined by premature fusion of one or more cranial sutures. Syndromic CS involves additional organ anomalies or neurocognitive deficits and accounts for 25%-30% of the cases. In a recent population-based study by our group, 84% of the syndromic CS cases had a genetically verified diagnosis after targeted analyses. A number of different genetic causes were detected, confirming that syndromic CS is highly heterogeneous. In this study, we performed whole-exome sequencing of 10 children and parents from the same cohort where previous genetic results were negative. We detected pathogenic, or likely pathogenic, variants in four additional genes (NFIA, EXTL3, POLR2A, and FOXP2) associated with rare conditions. In two of these (POLR2A and FOXP2), CS has not previously been reported. We further detected a rare predicted damaging variant in SH3BP4, which has not previously been related to human disease. All findings were clustered in genes involved in the pathways of osteogenesis and suture patency. We conclude that whole-exome sequencing expands the list of genes associated with syndromic CS, and provides new candidate genes in osteogenic signaling pathways.
Collapse
Affiliation(s)
- Elin Tønne
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.,Norwegian National Unit for Craniofacial Surgery, Oslo University Hospital, Oslo, Norway
| | - Bernt Johan Due-Tønnessen
- Norwegian National Unit for Craniofacial Surgery, Oslo University Hospital, Oslo, Norway.,Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Magnus Dehli Vigeland
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | | | - Teodora Ribarska
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | | | - Ying Sheng
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Eirik Helseth
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Gregor Duncan Gilfillan
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Inger-Lise Mero
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ketil Riddervold Heimdal
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.,Norwegian National Unit for Craniofacial Surgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
33
|
Ting MC, Farmer DT, Teng CS, He J, Chai Y, Crump JG, Maxson RE. Embryonic requirements for Tcf12 in the development of the mouse coronal suture. Development 2022; 149:273884. [PMID: 34878091 PMCID: PMC8783042 DOI: 10.1242/dev.199575] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 11/22/2021] [Indexed: 01/07/2023]
Abstract
A major feature of Saethre-Chotzen syndrome is coronal craniosynostosis, the fusion of the frontal and parietal bones at the coronal suture. It is caused by heterozygous loss-of-function mutations in either of the bHLH transcription factors TWIST1 and TCF12. Although compound heterozygous Tcf12; Twist1 mice display severe coronal synostosis, the individual role of Tcf12 had remained unexplored. Here, we show that Tcf12 controls several key processes in calvarial development, including the rate of frontal and parietal bone growth, and the boundary between sutural and osteogenic cells. Genetic analysis supports an embryonic requirement for Tcf12 in suture formation, as combined deletion of Tcf12 in embryonic neural crest and mesoderm, but not in postnatal suture mesenchyme, disrupts the coronal suture. We also detected asymmetric distribution of mesenchymal cells on opposing sides of the wild-type frontal and parietal bones, which prefigures later bone overlap at the sutures. In Tcf12 mutants, reduced asymmetry is associated with bones meeting end-on-end, possibly contributing to synostosis. Our results support embryonic requirements of Tcf12 in proper formation of the overlapping coronal suture.
Collapse
Affiliation(s)
- Man-chun Ting
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - D'Juan T. Farmer
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Camilla S. Teng
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jinzhi He
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA,Authors for correspondence (, )
| | - Robert E. Maxson
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA,Authors for correspondence (, )
| |
Collapse
|
34
|
Nuri T, Ota M, Ueda K, Iseki S. Quantitative Morphologic Analysis of Cranial Vault in Twist1+/- Mice: Implications in Craniosynostosis. Plast Reconstr Surg 2022; 149:28e-37e. [PMID: 34936613 DOI: 10.1097/prs.0000000000008665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The haploinsufficiency in the TWIST1 gene encoding a basic helix-loop-helix transcription factor is a cause of one of the craniosynostosis syndromes, Saethre-Chotzen syndrome. Patients with craniosynostosis usually require operative release of affected sutures, which makes it difficult to observe the long-term consequence of suture fusion on craniofacial growth. METHODS In this study, we performed quantitative analysis of morphologic changes of the skull in Twist1 heterozygously-deleted mice (Twist1+/-) with micro-computed tomographic images. RESULTS In Twist1+/- mice, fusion of the coronal suture began before postnatal day 14 and progressed until postnatal day 56, during which morphologic changes occurred. The growth of the skull was not achieved by a constant increase in the measured distances in wild type mice; some distances in the top-basal axis were decreased during the observation period. In the Twist1+/- mouse, growth in the top-basal axis was accelerated and that of the frontal cranium was reduced. In the unicoronal suture fusion mouse, the length of the zygomatic arch of affected side was shorter in the Twist1+/- mouse. In one postnatal day 56 Twist1+/- mouse with bilateral coronal suture fusion, asymmetric zygomatic arch length was identified. CONCLUSION The authors'results suggest that measuring the length of the left and right zygomatic arches may be useful for early diagnosis of coronal suture fusion and for estimation of the timing of synostosis, and that more detailed study on the growth pattern of the normal and the synostosed skull could provide prediction of the risk of resynostosis. CLINICAL RELEVANCE STATEMENT The data from this study can be useful to better understand the cranial growth pattern in patients with craniosynostosis.
Collapse
Affiliation(s)
- Takashi Nuri
- From the Department of Plastic Reconstructive Surgery, Osaka Medical College; Food and Nutrition, Japan Women's University; and Molecular Craniofacial Embryology, Tokyo Medical and Dental University
| | - Masato Ota
- From the Department of Plastic Reconstructive Surgery, Osaka Medical College; Food and Nutrition, Japan Women's University; and Molecular Craniofacial Embryology, Tokyo Medical and Dental University
| | - Koichi Ueda
- From the Department of Plastic Reconstructive Surgery, Osaka Medical College; Food and Nutrition, Japan Women's University; and Molecular Craniofacial Embryology, Tokyo Medical and Dental University
| | - Sachiko Iseki
- From the Department of Plastic Reconstructive Surgery, Osaka Medical College; Food and Nutrition, Japan Women's University; and Molecular Craniofacial Embryology, Tokyo Medical and Dental University
| |
Collapse
|
35
|
Craniofacial morphology and growth in Muenke syndrome, Saethre-Chotzen syndrome, and TCF12-related craniosynostosis. Clin Oral Investig 2021; 26:2927-2936. [PMID: 34904178 PMCID: PMC8898243 DOI: 10.1007/s00784-021-04275-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/07/2021] [Indexed: 11/20/2022]
Abstract
Objectives To determine whether the midface of patients with Muenke syndrome, Saethre-Chotzen syndrome, or TCF12-related craniosynostosis is hypoplastic compared to skeletal facial proportions of a Dutch control group. Material and methods We included seventy-four patients (43 patients with Muenke syndrome, 22 patients with Saethre-Chotzen syndrome, and 9 patients with TCF12-related craniosynostosis) who were referred between 1990 and 2020 (age range 4.84 to 16.83 years) and were treated at the Department of Oral Maxillofacial Surgery, Special Dental Care and Orthodontics, Children’s Hospital Erasmus University Medical Center, Sophia, Rotterdam, the Netherlands. The control group consisted of 208 healthy children. Results Cephalometric values comprising the midface were decreased in Muenke syndrome (ANB: β = –1.87, p = 0.001; and PC1: p < 0,001), Saethre-Chotzen syndrome (ANB: β = –1.76, p = 0.001; and PC1: p < 0.001), and TCF12-related craniosynostosis (ANB: β = –1.70, p = 0.015; and PC1: p < 0.033). Conclusions In this study, we showed that the midface is hypoplastic in Muenke syndrome, Saethre-Chotzen syndrome, and TCF12-related craniosynostosis compared to the Dutch control group. Furthermore, the rotation of the maxilla and the typical craniofacial buildup is significantly different in these three craniosynostosis syndromes compared to the controls. Clinical relevance The maxillary growth in patients with Muenke syndrome, Saethre-Chotzen syndrome, or TCF12-related craniosynostosis is impaired, leading to a deviant dental development. Therefore, timely orthodontic follow-up is recommended. In order to increase expertise and support treatment planning by medical and dental specialists for these patients, and also because of the specific differences between the syndromes, we recommend the management of patients with Muenke syndrome, Saethre-Chotzen syndrome, or TCF12-related craniosynostosis in specialized multidisciplinary teams. Supplementary Information The online version contains supplementary material available at 10.1007/s00784-021-04275-y.
Collapse
|
36
|
Twist1 Influences the Expression of Leading Members of the IL-17 Signaling Pathway in HER2-Positive Breast Cancer Cells. Int J Mol Sci 2021; 22:ijms222212144. [PMID: 34830027 PMCID: PMC8620489 DOI: 10.3390/ijms222212144] [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: 08/28/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 12/16/2022] Open
Abstract
Breast cancer (BC) is a heterogeneous disease composed of multiple subtypes with different molecular characteristics and clinical outcomes. The metastatic process in BC depends on the transcription factors (TFs) related to epithelial-mesenchymal transition (EMT), including the master regulator Twist1. However, its role beyond EMT in BC subtypes remains unclear. Our study aimed to investigate the role of Twist1, beyond EMT, in the molecular subtypes of BC. In patients, we observed the overexpression of TWIST1 in the HER2+ group. The silencing of TWIST1 in HER2+ BC cells resulted in the upregulation of 138 genes and the downregulation of 174 genes compared to control cells in a microarray assay. In silico analysis revealed correlations between Twist1 and important biological processes such as the Th17-mediated immune response, suggesting that Twist1 could be relevant for IL-17 signaling in HER2+ BC. IL-17 signaling was then examined, and it was shown that TWIST1 knockdown caused the downregulation of leading members of IL-17 signaling pathway. Taken together, our findings suggest that Twist1 plays a role on IL-17 signaling in HER2+ BC.
Collapse
|
37
|
Romanelli Tavares VL, Guimarães-Ramos SL, Zhou Y, Masotti C, Ezquina S, Moreira DDP, Buermans H, Freitas RS, Den Dunnen JT, Twigg SRF, Passos-Bueno MR. New locus underlying auriculocondylar syndrome (ARCND): 430 kb duplication involving TWIST1 regulatory elements. J Med Genet 2021; 59:895-905. [PMID: 34750192 PMCID: PMC9411924 DOI: 10.1136/jmedgenet-2021-107825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022]
Abstract
Background Auriculocondylar syndrome (ARCND) is a rare genetic disease that affects structures derived from the first and second pharyngeal arches, mainly resulting in micrognathia and auricular malformations. To date, pathogenic variants have been identified in three genes involved in the EDN1-DLX5/6 pathway (PLCB4, GNAI3 and EDN1) and some cases remain unsolved. Here we studied a large unsolved four-generation family. Methods We performed linkage analysis, resequencing and Capture-C to investigate the causative variant of this family. To test the pathogenicity of the CNV found, we modelled the disease in patient craniofacial progenitor cells, including induced pluripotent cell (iPSC)-derived neural crest and mesenchymal cells. Results This study highlights a fourth locus causative of ARCND, represented by a tandem duplication of 430 kb in a candidate region on chromosome 7 defined by linkage analysis. This duplication segregates with the disease in the family (LOD score=2.88) and includes HDAC9, which is located over 200 kb telomeric to the top candidate gene TWIST1. Notably, Capture-C analysis revealed multiple cis interactions between the TWIST1 promoter and possible regulatory elements within the duplicated region. Modelling of the disease revealed an increased expression of HDAC9 and its neighbouring gene, TWIST1, in neural crest cells. We also identified decreased migration of iPSC-derived neural crest cells together with dysregulation of osteogenic differentiation in iPSC-affected mesenchymal stem cells. Conclusion Our findings support the hypothesis that the 430 kb duplication is causative of the ARCND phenotype in this family and that deregulation of TWIST1 expression during craniofacial development can contribute to the phenotype.
Collapse
Affiliation(s)
| | | | - Yan Zhou
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Cibele Masotti
- Genética e Biologia Evolutiva, Universidade de São Paulo Instituto de Biociências, Sao Paulo, Brazil.,Molecular Oncology Center, Hospital Sírio-Libanês, Sao Paulo, Brazil
| | - Suzana Ezquina
- Genética e Biologia Evolutiva, Universidade de São Paulo Instituto de Biociências, Sao Paulo, Brazil.,Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Danielle de Paula Moreira
- Genética e Biologia Evolutiva, Universidade de São Paulo Instituto de Biociências, Sao Paulo, Brazil
| | - Henk Buermans
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Renato S Freitas
- Centro de Atendimento Integral ao Fissurado Lábio Palatal, Curitiba, Brazil
| | - Johan T Den Dunnen
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Stephen R F Twigg
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Maria Rita Passos-Bueno
- Genética e Biologia Evolutiva, Universidade de São Paulo Instituto de Biociências, Sao Paulo, Brazil
| |
Collapse
|
38
|
Fonteles CS, Finnell RH, Lei Y, Zurita-Jimenez ME, Monteiro AJ, George TM, Harshbarger RJ. De novo ALX4 variant detected in child with non-syndromic craniosynostosis. Braz J Med Biol Res 2021; 54:e11396. [PMID: 34586326 DOI: 10.1590/1414-431x2021e11396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 08/11/2021] [Indexed: 02/03/2023] Open
Abstract
Current understanding of the genetic factors contributing to the etiology of non-syndromic craniosynostosis (NSC) remains scarce. The present work investigated the presence of variants in ALX4, EFNA4, and TWIST1 genes in children with NSC to verify if variants within these genes may contribute to the occurrence of these abnormal phenotypes. A total of 101 children (aged 45.07±40.94 months) with NSC participated in this cross-sectional study. Parents and siblings of the probands were invited to participate. Medical and family history of craniosynostosis were documented. Biological samples were collected to obtain genomic DNA. Coding exons of human TWIST1, ALX4, and EFNA4 genes were amplified by polymerase chain reaction and Sanger sequenced. Five missense variants were identified in ALX4 in children with bilateral coronal, sagittal, and metopic synostosis. A de novo ALX4 variant, c.799G>A: p.Ala267Thr, was identified in a proband with sagittal synostosis. Three missense variants were identified in the EFNA4 gene in children with metopic and sagittal synostosis. A TWIST1 variant occurred in a child with unilateral coronal synostosis. Variants were predicted to be among the 0.1% (TWIST1, c.380C>A: p. Ala127Glu) and 1% (ALX4, c.769C>T: p.Arg257Cys, c.799G>A: p.Ala267Thr, c.929G>A: p.Gly310Asp; EFNA4, c.178C>T: p.His60Tyr, C.283A>G: p.Lys95Glu, c.349C>A: Pro117Thr) most deleterious variants in the human genome. With the exception of ALX4, c.799G>A: p.Ala267Thr, all other variants were present in at least one non-affected family member, suggesting incomplete penetrance. Thus, these variants may contribute to the development of craniosynostosis, and should not be discarded as potential candidate genes in the diagnosis of this condition.
Collapse
Affiliation(s)
- C S Fonteles
- Programa de Pós-graduação em Odontologia, Faculdade de Farmácia, Odontologia e Enfermagem, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - R H Finnell
- Center for Precision Environmental Health, Departments of Molecular and Cellular Biology, Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Y Lei
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - M E Zurita-Jimenez
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - A J Monteiro
- Departamento de Estatística e Matemática Aplicada, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - T M George
- Plastic Surgery, Craniofacial Team at the Dell Children's Medical Center of Central Texas, Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - R J Harshbarger
- Plastic Surgery, Craniofacial Team at the Dell Children's Medical Center of Central Texas, Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
39
|
Menon S, Salhotra A, Shailendra S, Tevlin R, Ransom RC, Januszyk M, Chan CKF, Behr B, Wan DC, Longaker MT, Quarto N. Skeletal stem and progenitor cells maintain cranial suture patency and prevent craniosynostosis. Nat Commun 2021; 12:4640. [PMID: 34330896 PMCID: PMC8324898 DOI: 10.1038/s41467-021-24801-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/07/2021] [Indexed: 12/29/2022] Open
Abstract
Cranial sutures are major growth centers for the calvarial vault, and their premature fusion leads to a pathologic condition called craniosynostosis. This study investigates whether skeletal stem/progenitor cells are resident in the cranial sutures. Prospective isolation by FACS identifies this population with a significant difference in spatio-temporal representation between fusing versus patent sutures. Transcriptomic analysis highlights a distinct signature in cells derived from the physiological closing PF suture, and scRNA sequencing identifies transcriptional heterogeneity among sutures. Wnt-signaling activation increases skeletal stem/progenitor cells in sutures, whereas its inhibition decreases. Crossing Axin2LacZ/+ mouse, endowing enhanced Wnt activation, to a Twist1+/- mouse model of coronal craniosynostosis enriches skeletal stem/progenitor cells in sutures restoring patency. Co-transplantation of these cells with Wnt3a prevents resynostosis following suturectomy in Twist1+/- mice. Our study reveals that decrease and/or imbalance of skeletal stem/progenitor cells representation within sutures may underlie craniosynostosis. These findings have translational implications toward therapeutic approaches for craniosynostosis.
Collapse
Affiliation(s)
- Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ankit Salhotra
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Siny Shailendra
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ruth Tevlin
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan C Ransom
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Charles K F Chan
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Björn Behr
- Department of Plastic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Napoli, Italy.
| |
Collapse
|
40
|
Naqvi S, Sleyp Y, Hoskens H, Indencleef K, Spence JP, Bruffaerts R, Radwan A, Eller RJ, Richmond S, Shriver MD, Shaffer JR, Weinberg SM, Walsh S, Thompson J, Pritchard JK, Sunaert S, Peeters H, Wysocka J, Claes P. Shared heritability of human face and brain shape. Nat Genet 2021; 53:830-839. [PMID: 33821002 PMCID: PMC8232039 DOI: 10.1038/s41588-021-00827-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 02/16/2021] [Indexed: 02/08/2023]
Abstract
Evidence from model organisms and clinical genetics suggests coordination between the developing brain and face, but the role of this link in common genetic variation remains unknown. We performed a multivariate genome-wide association study of cortical surface morphology in 19,644 individuals of European ancestry, identifying 472 genomic loci influencing brain shape, of which 76 are also linked to face shape. Shared loci include transcription factors involved in craniofacial development, as well as members of signaling pathways implicated in brain-face cross-talk. Brain shape heritability is equivalently enriched near regulatory regions active in either forebrain organoids or facial progenitors. However, we do not detect significant overlap between shared brain-face genome-wide association study signals and variants affecting behavioral-cognitive traits. These results suggest that early in embryogenesis, the face and brain mutually shape each other through both structural effects and paracrine signaling, but this interplay may not impact later brain development associated with cognitive function.
Collapse
Affiliation(s)
- Sahin Naqvi
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Departments of Genetics and Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Yoeri Sleyp
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Hanne Hoskens
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
| | - Karlijne Indencleef
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Jeffrey P Spence
- Departments of Genetics and Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Rose Bruffaerts
- Department of Neurosciences, KU Leuven, Leuven, Belgium, Hasselt University, Hasselt, Belgium
- Neurology Department, University Hospitals Leuven, Leuven, Belgium, Hasselt University, Hasselt, Belgium
- Biomedical Research Institute Hasselt University Hasselt Belgium, Hasselt University, Hasselt, Belgium
| | - Ahmed Radwan
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
| | - Ryan J Eller
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Stephen Richmond
- Applied Clinical Research and Public Health, School of Dentistry, Cardiff University, Cardiff, UK
| | - Mark D Shriver
- Department of Anthropology, Pennsylvania State University, State College, PA, USA
| | - John R Shaffer
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seth M Weinberg
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anthropology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susan Walsh
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - James Thompson
- Department of Psychology, George Mason University, Fairfax, VA, USA
| | - Jonathan K Pritchard
- Departments of Genetics and Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Stefan Sunaert
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Imaging and Pathology, Translational MRI, KU Leuven, Leuven, Belgium
| | - Hilde Peeters
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| | - Peter Claes
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium.
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
41
|
Zhou X, Hu S, Zhang Y, Du G, Li Y. The mechanism by which noncoding RNAs regulate muscle wasting in cancer cachexia. PRECISION CLINICAL MEDICINE 2021; 4:136-147. [PMID: 35694153 DOI: 10.1093/pcmedi/pbab008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 02/05/2023] Open
Abstract
Abstract
Cancer cachexia (CC) is a complex metabolic syndrome that accelerates muscle wasting and affects up to 80% of patients with cancer; however, timely diagnostic methods and effective cures are lacking. Although a considerable number of studies have focused on the mechanism of CC-induced muscle atrophy, few novel therapies have been applied in the last decade. In recent years, noncoding RNAs (ncRNAs) have attracted great attention as many differentially expressed ncRNAs in cancer cachectic muscles have been reported to participate in the inhibition of myogenesis and activation of proteolysis. In addition, extracellular vesicles (EVs), which function as ncRNA carriers in intercellular communication, are closely involved in changing ncRNA expression profiles in muscle and promoting the development of muscle wasting; thus, EV-related ncRNAs may represent potential therapeutic targets. This review comprehensively describes the process of ncRNA transmission through EVs and summarizes the pathways and targets of ncRNAs that lead to CC-induced muscle atrophy.
Collapse
Affiliation(s)
- Xueer Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shoushan Hu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yunan Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Guannan Du
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yi Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
42
|
Alghamdi M, Alhumsi TR, Altweijri I, Alkhamis WH, Barasain O, Cardona-Londoño KJ, Ramakrishnan R, Guzmán-Vega FJ, Arold ST, Ali G, Adly N, Ali H, Basudan A, Bakhrebah MA. Clinical and Genetic Characterization of Craniosynostosis in Saudi Arabia. Front Pediatr 2021; 9:582816. [PMID: 33937142 PMCID: PMC8085561 DOI: 10.3389/fped.2021.582816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 03/03/2021] [Indexed: 11/20/2022] Open
Abstract
Background: Craniosynostosis (CS) is defined as pre-mature fusion of one or more of the cranial sutures. CS is classified surgically as either simple or complex based on the number of cranial sutures involved. CS can also be classified genetically as isolated CS or syndromic CS if the patient has extracranial deformities. Currently, the link between clinical and genetic patterns of CS in the Saudi population is poorly understood. Methodology: We conducted a retrospective cohort study among 28 CS patients, of which 24 were operated and four were not. Clinical and genetic data were collected between February 2015 and February 2019, from consenting patient's families. The electronic chart data were collected and analyzed including patient demographics, craniofacial features, other anomalies and dysmorphic features, operative data, intra cranial pressure (ICP), parent consanguinity and genetic testing results. Results: The most common deformity in our population was trigonocephaly. The most performed procedure was cranial vault reconstruction with fronto-orbital advancement, followed by posterior vault distraction osteogenesis and suturectomy with barrel staving. Genetics analysis revealed pathogenic mutations in FGFR2 (6 cases), TWIST1 (3 cases), ALPL (2 cases), and TCF12 (2 cases), and FREM1 (2 case). Conclusion: Compared to Western countries, our Saudi cohort displays significant differences in the prevalence of CS features, such as the types of sutures and prevalence of inherited CS. The genomic background allows our phenotype-genotype study to reclassify variants of unknown significance. Worldwide, the sagittal suture is the most commonly affected suture in simple CS, but in the Saudi population, the metopic suture fusion was most commonly seen in our clinic. Further studies are needed to investigate the characteristics of CS in our population in a multicenter setting.
Collapse
Affiliation(s)
- Malak Alghamdi
- Medical Genetic Division, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Department of Pediatrics, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Taghreed R. Alhumsi
- Department of Plastic Surgery, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Ikhlass Altweijri
- Department of Neurosurgery, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Waleed H. Alkhamis
- Obstetrics and Gynecology, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Omar Barasain
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Kelly J. Cardona-Londoño
- Biological and Environmental Science and Engineering (BESE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Reshmi Ramakrishnan
- Biological and Environmental Science and Engineering (BESE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Francisco J. Guzmán-Vega
- Biological and Environmental Science and Engineering (BESE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Stefan T. Arold
- Biological and Environmental Science and Engineering (BESE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Center de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Ghaida Ali
- College of Medicine, Imam Muhammad Ibn Saud University, Riyadh, Saudi Arabia
| | - Nouran Adly
- College of Medicine Research Centre, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hebatallah Ali
- College of Medicine Research Centre, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Basudan
- Chair of Medical and Molecular Genetics, Department of Clinical Laboratory Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Muhammed A. Bakhrebah
- Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| |
Collapse
|
43
|
White HE, Goswami A, Tucker AS. The Intertwined Evolution and Development of Sutures and Cranial Morphology. Front Cell Dev Biol 2021; 9:653579. [PMID: 33842480 PMCID: PMC8033035 DOI: 10.3389/fcell.2021.653579] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Phenotypic variation across mammals is extensive and reflects their ecological diversification into a remarkable range of habitats on every continent and in every ocean. The skull performs many functions to enable each species to thrive within its unique ecological niche, from prey acquisition, feeding, sensory capture (supporting vision and hearing) to brain protection. Diversity of skull function is reflected by its complex and highly variable morphology. Cranial morphology can be quantified using geometric morphometric techniques to offer invaluable insights into evolutionary patterns, ecomorphology, development, taxonomy, and phylogenetics. Therefore, the skull is one of the best suited skeletal elements for developmental and evolutionary analyses. In contrast, less attention is dedicated to the fibrous sutural joints separating the cranial bones. Throughout postnatal craniofacial development, sutures function as sites of bone growth, accommodating expansion of a growing brain. As growth frontiers, cranial sutures are actively responsible for the size and shape of the cranial bones, with overall skull shape being altered by changes to both the level and time period of activity of a given cranial suture. In keeping with this, pathological premature closure of sutures postnatally causes profound misshaping of the skull (craniosynostosis). Beyond this crucial role, sutures also function postnatally to provide locomotive shock absorption, allow joint mobility during feeding, and, in later postnatal stages, suture fusion acts to protect the developed brain. All these sutural functions have a clear impact on overall cranial function, development and morphology, and highlight the importance that patterns of suture development have in shaping the diversity of cranial morphology across taxa. Here we focus on the mammalian cranial system and review the intrinsic relationship between suture development and morphology and cranial shape from an evolutionary developmental biology perspective, with a view to understanding the influence of sutures on evolutionary diversity. Future work integrating suture development into a comparative evolutionary framework will be instrumental to understanding how developmental mechanisms shaping sutures ultimately influence evolutionary diversity.
Collapse
Affiliation(s)
- Heather E White
- Department of Life Sciences, Natural History Museum, London, United Kingdom.,Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom.,Division of Biosciences, University College London, London, United Kingdom
| | - Anjali Goswami
- Department of Life Sciences, Natural History Museum, London, United Kingdom.,Division of Biosciences, University College London, London, United Kingdom
| | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
44
|
Truong BT, Artinger KB. The power of zebrafish models for understanding the co-occurrence of craniofacial and limb disorders. Genesis 2021; 59:e23407. [PMID: 33393730 PMCID: PMC8153179 DOI: 10.1002/dvg.23407] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
Craniofacial and limb defects are two of the most common congenital anomalies in the general population. Interestingly, these defects are not mutually exclusive. Many patients with craniofacial phenotypes, such as orofacial clefting and craniosynostosis, also present with limb defects, including polydactyly, syndactyly, brachydactyly, or ectrodactyly. The gene regulatory networks governing craniofacial and limb development initially seem distinct from one another, and yet these birth defects frequently occur together. Both developmental processes are highly conserved among vertebrates, and zebrafish have emerged as an advantageous model due to their high fecundity, relative ease of genetic manipulation, and transparency during development. Here we summarize studies that have used zebrafish models to study human syndromes that present with both craniofacial and limb phenotypes. We discuss the highly conserved processes of craniofacial and limb/fin development and describe recent zebrafish studies that have explored the function of genes associated with human syndromes with phenotypes in both structures. We attempt to identify commonalities between the two to help explain why craniofacial and limb anomalies often occur together.
Collapse
Affiliation(s)
- Brittany T. Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Kristin Bruk Artinger
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
45
|
Den Ottelander BK, Van Veelen MC, De Goederen R, Van De Beeten SDC, Dremmen MHG, Loudon SE, Versnel SL, Van Den Ouweland AMW, Van Dooren MF, Joosten KFM, Mathijssen IMJ. Saethre-Chotzen syndrome: long-term outcome of a syndrome-specific management protocol. Dev Med Child Neurol 2021; 63:104-110. [PMID: 32909287 PMCID: PMC7754116 DOI: 10.1111/dmcn.14670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2020] [Indexed: 11/29/2022]
Abstract
AIM To assess the long-term outcomes of our management protocol for Saethre-Chotzen syndrome, which includes one-stage fronto-orbital advancement. METHOD All patients born with Saethre-Chotzen syndrome between January 1992 and March 2017 were included. Evaluated parameters included occipital frontal head circumference (OFC), fundoscopy, neuroimaging (ventricular size, tonsillar position, and the presence of collaterals/an abnormal transverse sinus), polysomnography, and ophthalmological outcomes. The relationship between papilledema and its associated risk factors was evaluated with Fisher's exact test. RESULTS Thirty-two patients (21 females, 11 males) were included. Median (SD) age at first surgery was 9.6 months (3.1mo) for patients who were primarily referred to our center (range: 3.6-13.0mo), the median (SD) age at last follow-up was 13 years (5y 7mo; range: 3-25y). Seven patients had papilledema preoperatively, which recurred in two. Two patients had papilledema solely after first surgery. Second cranial vault expansion was indicated in 20%. Thirteen patients had an OFC deflection, indicating restricted skull growth, one patient had ventriculomegaly, and none developed hydrocephalus. Eleven patients had emissary veins, while the transverse sinus was aberrant unilaterally in 13 (hypoplastic n=10 and absent n=3). Four patients had mild tonsillar descent, one of which was a Chiari type I malformation. Four patients had obstructive sleep apnoea (two mild, one moderate, and one severe). An aberrant transverse sinus was associated with papilledema (p=0.01). INTERPRETATION Single one-stage fronto-orbital advancement was sufficient to prevent intracranial hypertension for 80% of our patients with Saethre-Chotzen syndrome. Follow-up should focus on OFC deflection and venous anomalies.
Collapse
Affiliation(s)
- Bianca K Den Ottelander
- Department of Plastic and Reconstructive Surgery and Hand SurgeryDutch Craniofacial CenterErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Marie‐Lise C Van Veelen
- Department of NeurosurgeryErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Robbin De Goederen
- Department of Plastic and Reconstructive Surgery and Hand SurgeryDutch Craniofacial CenterErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Stephanie DC Van De Beeten
- Department of Plastic and Reconstructive Surgery and Hand SurgeryDutch Craniofacial CenterErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Marjolein HG Dremmen
- Department of RadiologyErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Sjoukje E Loudon
- Department of OphthalmologyErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Sarah L Versnel
- Department of Plastic and Reconstructive Surgery and Hand SurgeryDutch Craniofacial CenterErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Ans MW Van Den Ouweland
- Department of Clinical GeneticsErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Marieke F Van Dooren
- Department of Clinical GeneticsErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Koen FM Joosten
- Pediatric Intensive Care UnitErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| | - Irene MJ Mathijssen
- Department of Plastic and Reconstructive Surgery and Hand SurgeryDutch Craniofacial CenterErasmus MC – Sophia Children’s HospitalUniversity Medical Center RotterdamRotterdamthe Netherlands
| |
Collapse
|
46
|
Pribadi C, Camp E, Cakouros D, Anderson P, Glackin C, Gronthos S. Pharmacological targeting of KDM6A and KDM6B, as a novel therapeutic strategy for treating craniosynostosis in Saethre-Chotzen syndrome. Stem Cell Res Ther 2020; 11:529. [PMID: 33298158 PMCID: PMC7726873 DOI: 10.1186/s13287-020-02051-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/26/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND During development, excessive osteogenic differentiation of mesenchymal progenitor cells (MPC) within the cranial sutures can lead to premature suture fusion or craniosynostosis, leading to craniofacial and cognitive issues. Saethre-Chotzen syndrome (SCS) is a common form of craniosynostosis, caused by TWIST-1 gene mutations. Currently, the only treatment option for craniosynostosis involves multiple invasive cranial surgeries, which can lead to serious complications. METHODS The present study utilized Twist-1 haploinsufficient (Twist-1del/+) mice as SCS mouse model to investigate the inhibition of Kdm6a and Kdm6b activity using the pharmacological inhibitor, GSK-J4, on calvarial cell osteogenic potential. RESULTS This study showed that the histone methyltransferase EZH2, an osteogenesis inhibitor, is downregulated in calvarial cells derived from Twist-1del/+ mice, whereas the counter histone demethylases, Kdm6a and Kdm6b, known promoters of osteogenesis, were upregulated. In vitro studies confirmed that siRNA-mediated inhibition of Kdm6a and Kdm6b expression suppressed osteogenic differentiation of Twist-1del/+ calvarial cells. Moreover, pharmacological targeting of Kdm6a and Kdm6b activity, with the inhibitor, GSK-J4, caused a dose-dependent suppression of osteogenic differentiation by Twist-1del/+ calvarial cells in vitro and reduced mineralized bone formation in Twist-1del/+ calvarial explant cultures. Chromatin immunoprecipitation and Western blot analyses found that GSK-J4 treatment elevated the levels of the Kdm6a and Kdm6b epigenetic target, the repressive mark of tri-methylated lysine 27 on histone 3, on osteogenic genes leading to repression of Runx2 and Alkaline Phosphatase expression. Pre-clinical in vivo studies showed that local administration of GSK-J4 to the calvaria of Twist-1del/+ mice prevented premature suture fusion and kept the sutures open up to postnatal day 20. CONCLUSION The inhibition of Kdm6a and Kdm6b activity by GSK-J4 could be used as a potential non-invasive therapeutic strategy for preventing craniosynostosis in children with SCS. Pharmacological targeting of Kdm6a/b activity can alleviate craniosynostosis in Saethre-Chotzen syndrome. Aberrant osteogenesis by Twist-1 mutant cranial suture mesenchymal progenitor cells occurs via deregulation of epigenetic modifiers Ezh2 and Kdm6a/Kdm6b. Suppression of Kdm6a- and Kdm6b-mediated osteogenesis with GSK-J4 inhibitor can prevent prefusion of cranial sutures.
Collapse
Affiliation(s)
- Clara Pribadi
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Esther Camp
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Dimitrios Cakouros
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Peter Anderson
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Craniofacial Unit, Women and Children Hospital, North Adelaide, South Australia, Australia
| | - Carlotta Glackin
- Molecular Medicine and Neurosciences, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia. .,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.
| |
Collapse
|
47
|
A de novo frameshift mutation in ZEB2 causes polledness, abnormal skull shape, small body stature and subfertility in Fleckvieh cattle. Sci Rep 2020; 10:17032. [PMID: 33046754 PMCID: PMC7550345 DOI: 10.1038/s41598-020-73807-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/15/2020] [Indexed: 01/17/2023] Open
Abstract
Polledness in cattle is an autosomal dominant trait. Previous studies have revealed allelic heterogeneity at the polled locus and four different variants were identified, all in intergenic regions. In this study, we report a case of polled bull (FV-Polled1) born to horned parents, indicating a de novo origin of this polled condition. Using 50K genotyping and whole genome sequencing data, we identified on chromosome 2 an 11-bp deletion (AC_000159.1:g.52364063_52364073del; Del11) in the second exon of ZEB2 gene as the causal mutation for this de novo polled condition. We predicted that the deletion would shorten the protein product of ZEB2 by almost 91%. Moreover, we showed that all animals carrying Del11 mutation displayed symptoms similar to Mowat-Wilson syndrome (MWS) in humans, which is also associated with genetic variations in ZEB2. The symptoms in cattle include delayed maturity, small body stature and abnormal shape of skull. This is the first report of a de novo dominant mutation affecting only ZEB2 and associated with a genetic absence of horns. Therefore our results demonstrate undoubtedly that ZEB2 plays an important role in the process of horn ontogenesis as well as in the regulation of overall development and growth of animals.
Collapse
|
48
|
Genetic background dependent modifiers of craniosynostosis severity. J Struct Biol 2020; 212:107629. [PMID: 32976998 DOI: 10.1016/j.jsb.2020.107629] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 12/14/2022]
Abstract
Craniosynostosis severity varies in patients with identical genetic mutations. To understand causes of this phenotypic variation, we backcrossed the FGFR2+/C342Y mouse model of Crouzon syndrome onto congenic C57BL/6 and BALB/c backgrounds. Coronal suture fusion was observed in C57BL/6 (88% incidence, p < .001 between genotypes) but not in BALB/c FGFR2+/C342Y mutant mice at 3 weeks after birth, establishing that that the two models differ in phenotype severity. To begin identifying pre-existing modifiers of craniosynostosis severity, we compared transcriptome signatures of cranial tissues from C57BL/6 vs. BALB/c FGFR2+/+ mice. We separately analyzed frontal bone with coronal suture tissue from parietal bone with sagittal suture tissues because the coronal suture but not the sagittal suture fuses in FGFR2+/C342Y mice. The craniosynostosis associated Twist and En1 transcription factors were down-regulated, while Runx2 was up-regulated, in C57BL/6 compared to BALB/c tissues, which could predispose to craniosynostosis. Transcriptome analyses under the GO term MAPK cascade revealed that genes associated with calcium ion channels, angiogenesis, protein quality control and cell stress response were central to transcriptome differences associated with genetic background. FGFR2 and HSPA2 protein levels plus ERK1/2 activity were higher in cells isolated from C57BL/6 than BALB/c cranial tissues. Notably, the HSPA2 protein chaperone is central to craniofacial genetic epistasis, and we find that FGFR2 protein is abnormally processed in primary cells from FGFR2+/C342Y but not FGFR2+/+ mice. Therefore, we propose that differences in protein quality control responses may contribute to genetic background influences on craniosynostosis phenotype severity.
Collapse
|
49
|
Razzaque MS, Atfi A. TGIF1-Twist1 axis in pancreatic ductal adenocarcinoma. Comput Struct Biotechnol J 2020; 18:2568-2572. [PMID: 33005315 PMCID: PMC7520386 DOI: 10.1016/j.csbj.2020.09.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 12/18/2022] Open
Abstract
TG-interacting factor 1 (TGIF1) exerts inhibitory effects on transforming growth factor-beta (TGF-β) signaling by suppressing Smad signaling pathway at multiple levels. TGIF1 activity is important for normal embryogenesis and organogenesis, yet its dysregulation can culminate in tumorigenesis. For instance, increased expression of TGIF1 correlates with poor prognosis in triple-negative breast cancer patients, and enforced expression of TGIF1 facilitates Wnt-driven mammary tumorigenesis, suggesting that TGIF1 might function as an oncoprotein. Quite surprisingly, TGIF1 has recently been shown to function as a tumor suppressor in pancreatic ductal adenocarcinoma (PDAC), possibly owing to its ability to antagonize the pro-malignant transcription factor Twist1. In this article, we will briefly elaborate on the biological and clinical significance of the unique tumor-suppressive function of TGIF1 and its functional interaction with Twist1 in the context of PDAC pathogenesis and progression.
Collapse
Affiliation(s)
- Mohammed S Razzaque
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| | - Azeddine Atfi
- Department of Pathology and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
50
|
Ertosun MG, PehlİvanoĞlu S, DİlmaÇ S, TanriÖver G, ÖzeŞ ON. AKT-mediated phosphorylation of TWIST1 is essential for breast cancer cell metastasis. ACTA ACUST UNITED AC 2020; 44:158-165. [PMID: 32922123 PMCID: PMC7478131 DOI: 10.3906/biy-1912-74] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previously, it was shown that human TWIST1 (basic helix-loop-helix (b-HLH) is phosphorylated by Akt kinase at S42, T121, and S123. To show in vivo effect of these phosphorylations, we created mouse TWIST1 expression vector and converted the codons of S42, T125, and S127 to unphosphorylatable alanine and phosphorylation mimicking Glutamic acid. We hypothesized that alanine mutants would inhibit the metastatic ability of 4T1 cells while glutamic acid mutants would convert nonmetastatic 67NR cells into metastatic phenotype. To confirm this hypothesis, we created metastatic 4T1 and nonmetastatic 67NR cells expressing alanine mutants and glutamic acid mutants mouse TWIST1, respectively. Then, we injected 1 × 106 67NR and 1 × 105 4T1 cells overexpressing mutants of TWIST1 into the breast tissue of BALB/c mice. At the end of the 4th week, we sacrificed the animals, determined the numbers of tumors at lungs and liver. Although 67NR cells overexpressing wild-type TWIST1 did not show any metastasis, cells overexpressing S42E and T125E mutants showed 15–30 macroscopic metastasis to liver and lungs. Parallel to this, 4T1 cells expressing S42A and T125A mutants of TWIST1 showed no macroscopic metastasis. Our results indicate that phosphorylation of S42 and T125 by AKT is essential for TWIST1-mediated tumor growth and metastasis.
Collapse
Affiliation(s)
- Mustafa Gökhan Ertosun
- Department Plastic, Reconstructive and Aesthetic Surgery, Faculty of Medicine, Akdeniz University, Antalya Turkey
| | - Suray PehlİvanoĞlu
- Department of Molecular Biology, Faculty of Science, Necmettin Erbakan University, Konya Turkey
| | - Sayra DİlmaÇ
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya Turkey
| | - Gamze TanriÖver
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya Turkey
| | | |
Collapse
|