1
|
Williams RM, Taylor G, Ling ITC, Candido-Ferreira I, Fountain DM, Mayes S, Ateş-Kalkan PS, Haug JO, Price AJ, McKinney SA, Bozhilovh YK, Tyser RCV, Srinivas S, Hughes JR, Sauka-Spengler T. Chromatin remodeller Chd7 is developmentally regulated in the neural crest by tissue-specific transcription factors. PLoS Biol 2024; 22:e3002786. [PMID: 39418292 PMCID: PMC11521297 DOI: 10.1371/journal.pbio.3002786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 10/29/2024] [Accepted: 08/02/2024] [Indexed: 10/19/2024] Open
Abstract
Neurocristopathies such as CHARGE syndrome result from aberrant neural crest development. A large proportion of CHARGE cases are attributed to pathogenic variants in the gene encoding CHD7, chromodomain helicase DNA binding protein 7, which remodels chromatin. While the role for CHD7 in neural crest development is well documented, how this factor is specifically up-regulated in neural crest cells is not understood. Here, we use epigenomic profiling of chick and human neural crest to identify a cohort of enhancers regulating Chd7 expression in neural crest cells and other tissues. We functionally validate upstream transcription factor binding at candidate enhancers, revealing novel epistatic relationships between neural crest master regulators and Chd7, showing tissue-specific regulation of a globally acting chromatin remodeller. Furthermore, we find conserved enhancer features in human embryonic epigenomic data and validate the activity of the human equivalent CHD7 enhancers in the chick embryo. Our findings embed Chd7 in the neural crest gene regulatory network and offer potentially clinically relevant elements for interpreting CHARGE syndrome cases without causative allocation.
Collapse
Affiliation(s)
- Ruth M. Williams
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
| | - Guneş Taylor
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
| | - Irving T. C. Ling
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
- University of Oxford, Department of Paediatric Surgery, Children’s Hospital Oxford, Oxford, United Kingdom
| | - Ivan Candido-Ferreira
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
| | - Daniel M. Fountain
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
| | - Sarah Mayes
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
| | | | - Julianna O. Haug
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Andrew J. Price
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Sean A. McKinney
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Yavor K. Bozhilovh
- University of Oxford, MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Oxford, United Kingdom
- University of Oxford, MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
| | - Richard C. V. Tyser
- University of Oxford, Department of Physiology, Anatomy and Genetics, South Parks Road, Oxford, United Kingdom
| | - Shankar Srinivas
- University of Oxford, Department of Physiology, Anatomy and Genetics, South Parks Road, Oxford, United Kingdom
| | - Jim R. Hughes
- University of Oxford, MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Oxford, United Kingdom
- University of Oxford, MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
| | - Tatjana Sauka-Spengler
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford, United Kingdom
- University of Oxford, MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Oxford, United Kingdom
| |
Collapse
|
2
|
Breuer M, Rummler M, Singh J, Maher S, Zaouter C, Jamadagni P, Pilon N, Willie BM, Patten SA. CHD7 regulates craniofacial cartilage development via controlling HTR2B expression. J Bone Miner Res 2024; 39:498-512. [PMID: 38477756 PMCID: PMC11262153 DOI: 10.1093/jbmr/zjae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 12/19/2023] [Accepted: 01/17/2024] [Indexed: 03/14/2024]
Abstract
Mutations in the Chromodomain helicase DNA-binding protein 7 - coding gene (CHD7) cause CHARGE syndrome (CS). Although craniofacial and skeletal abnormalities are major features of CS patients, the role of CHD7 in bone and cartilage development remain largely unexplored. Here, using a zebrafish (Danio rerio) CS model, we show that chd7-/- larvae display abnormal craniofacial cartilage development and spinal deformities. The craniofacial and spine defects are accompanied by a marked reduction of bone mineralization. At the molecular level, we show that these phenotypes are associated with significant reduction in the expression levels of osteoblast differentiation markers. Additionally, we detected a marked depletion of collagen 2α1 in the cartilage of craniofacial regions and vertebrae, along with significantly reduced number of chondrocytes. Chondrogenesis defects are at least in part due to downregulation of htr2b, which we found to be also dysregulated in human cells derived from an individual with CHD7 mutation-positive CS. Overall, this study thus unveils an essential role for CHD7 in cartilage and bone development, with potential clinical relevance for the craniofacial defects associated with CS.
Collapse
Affiliation(s)
- Maximilian Breuer
- Institut National de la Recherche Scientifique (INRS) – Centre Armand Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada
| | - Maximilian Rummler
- Research Centre, Shriners Hospital for Children-Canada, Department of Biological and Biomedical Engineering, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal H4A 0A9, Canada
| | - Jaskaran Singh
- Institut National de la Recherche Scientifique (INRS) – Centre Armand Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada
| | - Sabrina Maher
- Institut National de la Recherche Scientifique (INRS) – Centre Armand Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada
- Research Centre, Shriners Hospital for Children-Canada, Department of Biological and Biomedical Engineering, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal H4A 0A9, Canada
- Département de Neurosciences, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Charlotte Zaouter
- Institut National de la Recherche Scientifique (INRS) – Centre Armand Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada
| | - Priyanka Jamadagni
- Institut National de la Recherche Scientifique (INRS) – Centre Armand Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada
| | - Nicolas Pilon
- Molecular Genetics of Development Laboratory, Départment des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montréal, QC H3C 3P8, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montréal, QC H3C 3P8, Canada
| | - Bettina M Willie
- Research Centre, Shriners Hospital for Children-Canada, Department of Biological and Biomedical Engineering, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal H4A 0A9, Canada
| | - Shunmoogum A Patten
- Institut National de la Recherche Scientifique (INRS) – Centre Armand Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada
- Département de Neurosciences, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montréal, QC H3C 3P8, Canada
| |
Collapse
|
3
|
Stathopoulou A, Wang P, Thellier C, Kelly RG, Zheng D, Scambler PJ. CHARGE syndrome-associated CHD7 acts at ISL1-regulated enhancers to modulate second heart field gene expression. Cardiovasc Res 2023; 119:2089-2105. [PMID: 37052590 PMCID: PMC10478754 DOI: 10.1093/cvr/cvad059] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/20/2022] [Accepted: 04/12/2023] [Indexed: 04/14/2023] Open
Abstract
AIMS Haploinsufficiency of the chromo-domain protein CHD7 underlies most cases of CHARGE syndrome, a multisystem birth defect including congenital heart malformation. Context specific roles for CHD7 in various stem, progenitor, and differentiated cell lineages have been reported. Previously, we showed severe defects when Chd7 is absent from cardiopharyngeal mesoderm (CPM). Here, we investigate altered gene expression in the CPM and identify specific CHD7-bound target genes with known roles in the morphogenesis of affected structures. METHODS AND RESULTS We generated conditional KO of Chd7 in CPM and analysed cardiac progenitor cells using transcriptomic and epigenomic analyses, in vivo expression analysis, and bioinformatic comparisons with existing datasets. We show CHD7 is required for correct expression of several genes established as major players in cardiac development, especially within the second heart field (SHF). We identified CHD7 binding sites in cardiac progenitor cells and found strong association with histone marks suggestive of dynamically regulated enhancers during the mesodermal to cardiac progenitor transition of mESC differentiation. Moreover, CHD7 shares a subset of its target sites with ISL1, a pioneer transcription factor in the cardiogenic gene regulatory network, including one enhancer modulating Fgf10 expression in SHF progenitor cells vs. differentiating cardiomyocytes. CONCLUSION We show that CHD7 interacts with ISL1, binds ISL1-regulated cardiac enhancers, and modulates gene expression across the mesodermal heart fields during cardiac morphogenesis.
Collapse
Affiliation(s)
- Athanasia Stathopoulou
- Developmental Biology of Birth Defects, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | | | - Robert G Kelly
- Aix-Marseille University, CNRS UMR 7288, IBDM, Marseille, France
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Neurology and Neurosciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Peter J Scambler
- Developmental Biology of Birth Defects, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
4
|
Hodorovich DR, Lindsley PM, Berry AA, Burton DF, Marsden KC. Morphological and sensorimotor phenotypes in a zebrafish CHARGE syndrome model are domain-dependent. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12839. [PMID: 36717082 PMCID: PMC10242184 DOI: 10.1111/gbb.12839] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/22/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023]
Abstract
CHARGE syndrome is a heterogeneous disorder characterized by a spectrum of defects affecting multiple tissues and behavioral difficulties such as autism, attention-deficit/hyperactivity disorder, obsessive-compulsive disorder, anxiety, and sensory deficits. Most CHARGE cases arise from de novo, loss-of-function mutations in chromodomain-helicase-DNA-binding-protein-7 (CHD7). CHD7 is required for processes such as neuronal differentiation and neural crest cell migration, but how CHD7 affects neural circuit function to regulate behavior is unclear. To investigate the pathophysiology of behavioral symptoms in CHARGE, we established a mutant chd7 zebrafish line that recapitulates multiple CHARGE phenotypes including ear, cardiac, and craniofacial defects. Using a panel of behavioral assays, we found that chd7 mutants have specific auditory and visual behavior deficits that are independent of defects in sensory structures. Mauthner cell-dependent short-latency acoustic startle responses are normal in chd7 mutants, while Mauthner-independent long-latency responses are reduced. Responses to sudden decreases in light are also reduced in mutants, while responses to sudden increases in light are normal, suggesting that the retinal OFF pathway may be affected. Furthermore, by analyzing multiple chd7 alleles we observed that the penetrance of morphological and behavioral phenotypes is influenced by genetic background but that it also depends on the mutation location, with a chromodomain mutation causing the highest penetrance. This pattern is consistent with analysis of a CHARGE patient dataset in which symptom penetrance was highest in subjects with mutations in the CHD7 chromodomains. These results provide new insight into the heterogeneity of CHARGE and will inform future work to define CHD7-dependent neurobehavioral mechanisms.
Collapse
Affiliation(s)
- Dana R. Hodorovich
- Department of Biological SciencesNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Patrick M. Lindsley
- Department of Biological SciencesNorth Carolina State UniversityRaleighNorth CarolinaUSA
- University of Virginia School of Medicine, University of VirginiaCharlottesvilleVAUSA
| | - Austen A. Berry
- Department of Biological SciencesNorth Carolina State UniversityRaleighNorth CarolinaUSA
- BiogenDurhamNCUSA
| | - Derek F. Burton
- Department of Biological SciencesNorth Carolina State UniversityRaleighNorth CarolinaUSA
- Washington UniversitySt. LouisMOUSA
| | - Kurt C. Marsden
- Department of Biological SciencesNorth Carolina State UniversityRaleighNorth CarolinaUSA
- Washington UniversitySt. LouisMOUSA
| |
Collapse
|
5
|
MacLean JE, Wertman JN, Prykhozhij SV, Chedrawe E, Langley S, Steele SL, Ban K, Blake K, Berman JN. phox2ba: The Potential Genetic Link behind the Overlap in the Symptomatology between CHARGE and Central Congenital Hypoventilation Syndromes. Genes (Basel) 2023; 14:genes14051086. [PMID: 37239446 DOI: 10.3390/genes14051086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
CHARGE syndrome typically results from mutations in the gene encoding chromodomain helicase DNA-binding protein 7 (CHD7). CHD7 is involved in regulating neural crest development, which gives rise to tissues of the skull/face and the autonomic nervous system (ANS). Individuals with CHARGE syndrome are frequently born with anomalies requiring multiple surgeries and often experience adverse events post-anesthesia, including oxygen desaturations, decreased respiratory rates, and heart rate abnormalities. Central congenital hypoventilation syndrome (CCHS) affects ANS components that regulate breathing. Its hallmark feature is hypoventilation during sleep, clinically resembling observations in anesthetized CHARGE patients. Loss of PHOX2B (paired-like homeobox 2b) underlies CCHS. Employing a chd7-null zebrafish model, we investigated physiologic responses to anesthesia and compared these to loss of phox2b. Heart rates were lower in chd7 mutants compared to the wild-type. Exposure to tricaine, a zebrafish anesthetic/muscle relaxant, revealed that chd7 mutants took longer to become anesthetized, with higher respiratory rates during recovery. chd7 mutant larvae demonstrated unique phox2ba expression patterns. The knockdown of phox2ba reduced larval heart rates similar to chd7 mutants. chd7 mutant fish are a valuable preclinical model to investigate anesthesia in CHARGE syndrome and reveal a novel functional link between CHARGE syndrome and CCHS.
Collapse
Affiliation(s)
- Jessica E MacLean
- Department of Pediatrics, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Jaime N Wertman
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Sergey V Prykhozhij
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Emily Chedrawe
- Department of Pediatrics, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Stewart Langley
- Department of Pediatrics, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Shelby L Steele
- Department of Pediatrics, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Kevin Ban
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Kim Blake
- Department of Pediatrics, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Jason N Berman
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Departments of Pediatrics and Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
6
|
Kocere A, Lalonde RL, Mosimann C, Burger A. Lateral thinking in syndromic congenital cardiovascular disease. Dis Model Mech 2023; 16:dmm049735. [PMID: 37125615 PMCID: PMC10184679 DOI: 10.1242/dmm.049735] [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] [Indexed: 05/02/2023] Open
Abstract
Syndromic birth defects are rare diseases that can present with seemingly pleiotropic comorbidities. Prime examples are rare congenital heart and cardiovascular anomalies that can be accompanied by forelimb defects, kidney disorders and more. Whether such multi-organ defects share a developmental link remains a key question with relevance to the diagnosis, therapeutic intervention and long-term care of affected patients. The heart, endothelial and blood lineages develop together from the lateral plate mesoderm (LPM), which also harbors the progenitor cells for limb connective tissue, kidneys, mesothelia and smooth muscle. This developmental plasticity of the LPM, which founds on multi-lineage progenitor cells and shared transcription factor expression across different descendant lineages, has the potential to explain the seemingly disparate syndromic defects in rare congenital diseases. Combining patient genome-sequencing data with model organism studies has already provided a wealth of insights into complex LPM-associated birth defects, such as heart-hand syndromes. Here, we summarize developmental and known disease-causing mechanisms in early LPM patterning, address how defects in these processes drive multi-organ comorbidities, and outline how several cardiovascular and hematopoietic birth defects with complex comorbidities may be LPM-associated diseases. We also discuss strategies to integrate patient sequencing, data-aggregating resources and model organism studies to mechanistically decode congenital defects, including potentially LPM-associated orphan diseases. Eventually, linking complex congenital phenotypes to a common LPM origin provides a framework to discover developmental mechanisms and to anticipate comorbidities in congenital diseases affecting the cardiovascular system and beyond.
Collapse
Affiliation(s)
- Agnese Kocere
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
- Department of Molecular Life Science, University of Zurich, 8057 Zurich, Switzerland
| | - Robert L. Lalonde
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| | - Alexa Burger
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO 80045, USA
| |
Collapse
|
7
|
Cellular and Molecular Mechanisms of Pathogenesis Underlying Inherited Retinal Dystrophies. Biomolecules 2023; 13:biom13020271. [PMID: 36830640 PMCID: PMC9953031 DOI: 10.3390/biom13020271] [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: 12/21/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are congenital retinal degenerative diseases that have various inheritance patterns, including dominant, recessive, X-linked, and mitochondrial. These diseases are most often the result of defects in rod and/or cone photoreceptor and retinal pigment epithelium function, development, or both. The genes associated with these diseases, when mutated, produce altered protein products that have downstream effects in pathways critical to vision, including phototransduction, the visual cycle, photoreceptor development, cellular respiration, and retinal homeostasis. The aim of this manuscript is to provide a comprehensive review of the underlying molecular mechanisms of pathogenesis of IRDs by delving into many of the genes associated with IRD development, their protein products, and the pathways interrupted by genetic mutation.
Collapse
|
8
|
Krueger LA, Bills JD, Lim ZY, Skidmore JM, Martin DM, Morris AC. Chromatin remodeler Chd7 regulates photoreceptor development and outer segment length. Exp Eye Res 2023; 226:109299. [PMID: 36343670 PMCID: PMC10354686 DOI: 10.1016/j.exer.2022.109299] [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: 06/01/2022] [Revised: 09/29/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Mutations in the chromatin remodeling factor CHD7 are the predominant cause of CHARGE syndrome, a congenital disorder that frequently includes ocular coloboma. Although CHD7 is known to be required for proper ocular morphogenesis, its role in retinal development has not been thoroughly investigated. Given that individuals with CHARGE syndrome can experience visual impairment even in the absence of coloboma, a better understanding of CHD7 function in the retina is needed. In this study, we characterized the expression pattern of Chd7 in the developing zebrafish and mouse retina and documented ocular and retinal phenotypes in Chd7 loss-of-function mutants. Zebrafish Chd7 was expressed throughout the retinal neuroepithelium when retinal progenitor cells were actively proliferating, and later in subsets of newly post-mitotic retinal cells. At stages of retinal development when most retinal cell types had terminally differentiated, Chd7 expression remained strong in the ganglion cell layer and in some cells in the inner nuclear layer. Intriguingly, strong expression of Chd7 was also observed in the outer nuclear layer where it was co-expressed with markers of post-mitotic cone and rod photoreceptors. Expression of mouse CHD7 displayed a similar pattern, including expression in the ganglion cells, subsets of inner nuclear layer cells, and in the distal outer nuclear layer as late as P15. Two different mutant chd7 zebrafish lines were characterized for ocular and retinal defects. These mutants displayed microphthalmia, reduced numbers of cone photoreceptors, and truncated rod and cone photoreceptor outer segments. Reduced cone photoreceptor number and abnormal outer segments were also observed in heterozygous Chd7 mutant mice. Taken together, our results in zebrafish and mouse reveal a conserved, previously undescribed role for Chd7 in retinal development and photoreceptor outer segment morphogenesis. Moreover, our work suggests an avenue of future investigation into the pathogenesis of visual system defects in CHARGE syndrome.
Collapse
Affiliation(s)
- Laura A Krueger
- Department of Biology, University of Kentucky, Lexington, KY, 40506-0225, USA
| | - Jessica D Bills
- Department of Biology, University of Kentucky, Lexington, KY, 40506-0225, USA
| | - Zun Yi Lim
- Department of Biology, University of Kentucky, Lexington, KY, 40506-0225, USA
| | | | - Donna M Martin
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ann C Morris
- Department of Biology, University of Kentucky, Lexington, KY, 40506-0225, USA.
| |
Collapse
|
9
|
Sun Y, Kumar SR, Wong CED, Tian Z, Bai H, Crump JG, Bajpai R, Lien CL. Craniofacial and cardiac defects in chd7 zebrafish mutants mimic CHARGE syndrome. Front Cell Dev Biol 2022; 10:1030587. [PMID: 36568983 PMCID: PMC9768498 DOI: 10.3389/fcell.2022.1030587] [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: 08/29/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022] Open
Abstract
Congenital heart defects occur in almost 80% of patients with CHARGE syndrome, a sporadically occurring disease causing craniofacial and other abnormalities due to mutations in the CHD7 gene. Animal models have been generated to mimic CHARGE syndrome; however, heart defects are not extensively described in zebrafish disease models of CHARGE using morpholino injections or genetic mutants. Here, we describe the co-occurrence of craniofacial abnormalities and heart defects in zebrafish chd7 mutants. These mutant phenotypes are enhanced in the maternal zygotic mutant background. In the chd7 mutant fish, we found shortened craniofacial cartilages and extra cartilage formation. Furthermore, the length of the ventral aorta is altered in chd7 mutants. Many CHARGE patients have aortic arch anomalies. It should be noted that the aberrant branching of the first branchial arch artery is observed for the first time in chd7 fish mutants. To understand the cellular mechanism of CHARGE syndrome, neural crest cells (NCCs), that contribute to craniofacial and cardiovascular tissues, are examined using sox10:Cre lineage tracing. In contrast to its function in cranial NCCs, we found that the cardiac NCC-derived mural cells along the ventral aorta and aortic arch arteries are not affected in chd7 mutant fish. The chd7 fish mutants we generated recapitulate some of the craniofacial and cardiovascular phenotypes found in CHARGE patients and can be used to further determine the roles of CHD7.
Collapse
Affiliation(s)
- Yuhan Sun
- Saban Research Institute and Heart Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States,Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, United States
| | - S. Ram Kumar
- Saban Research Institute and Heart Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States,Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Chee Ern David Wong
- Saban Research Institute and Heart Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Zhiyu Tian
- Saban Research Institute and Heart Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Haipeng Bai
- Saban Research Institute and Heart Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States,State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - J. Gage Crump
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ruchi Bajpai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, United States,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ching Ling Lien
- Saban Research Institute and Heart Institute, Children’s Hospital Los Angeles, Los Angeles, CA, United States,Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States,*Correspondence: Ching Ling Lien,
| |
Collapse
|
10
|
Luo H, Luo S, Fang W, Lin Q, Chen X, Zhou X. Genomic insight into the nocturnal adaptation of the black-crowned night heron (Nycticorax nycticorax). BMC Genomics 2022; 23:683. [PMID: 36192687 PMCID: PMC9531477 DOI: 10.1186/s12864-022-08904-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The black-crowned night heron (Nycticorax nycticorax) is an ardeid bird successfully adapted to the nocturnal environment. Previous studies had indicated that the eyes of the night herons have evolved several specialized morphological traits favoring nocturnal vision. However, the molecular mechanisms of the nocturnal vision adaptation of night herons remained inattentions. In this study, the whole genome of N. nycticorax was sequenced and comparative analyses were performed on the vision-related and olfactory receptor (OR) genes to understand the molecular mechanisms of the visual and olfactory adaptation of night herons. RESULTS The results indicated that a number of vision genes were under positive or relaxed selection in N. nycticorax, whereas a number of other vision genes were under relaxed or intensified selection in the boat-billed heron (Cochlearius cochlearius), which suggested that the two species adapt to nocturnality with different genetic mechanisms. The different selections acting on vision genes are probably associated with the enlargement of eye size and the enhancement of visual sensitivity in night herons. The analyses on olfactory receptor (OR) genes indicated that the total number of OR genes in the genomes of N. nycticorax and C. cochlearius were about half those in the little egret (Egretta garzetta), whereas the diversity of their OR genes was not remarkably different. Additionally, the number of expressed OR genes in the transcriptomes of N. nycticorax was also fewer than that in E. garzetta. These results suggest a reduced olfactory capability in night herons compared with E. garzetta. CONCLUSIONS Our results provided evidence that several vision genes of the night herons were subjected to different natural selections, which can contribute to a better understanding of the genetic mechanisms of visual adaptions of the night heron. In addition, the finding of the reduced number of total and expressed OR genes in night herons may reflect a trade-off between olfaction and vision.
Collapse
Affiliation(s)
- Haoran Luo
- Key Laboratory of Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Site Luo
- Key Laboratory of Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Wenzhen Fang
- Key Laboratory of Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Qingxian Lin
- Key Laboratory of Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Xiaolin Chen
- Key Laboratory of Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, 361102, People's Republic of China.
| | - Xiaoping Zhou
- Key Laboratory of Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, 361102, People's Republic of China.
| |
Collapse
|
11
|
Krueger LA, Morris AC. Eyes on CHARGE syndrome: Roles of CHD7 in ocular development. Front Cell Dev Biol 2022; 10:994412. [PMID: 36172288 PMCID: PMC9512043 DOI: 10.3389/fcell.2022.994412] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
The development of the vertebrate visual system involves complex morphogenetic interactions of cells derived from multiple embryonic lineages. Disruptions in this process are associated with structural birth defects such as microphthalmia, anophthalmia, and coloboma (collectively referred to as MAC), and inherited retinal degenerative diseases such as retinitis pigmentosa and allied dystrophies. MAC and retinal degeneration are also observed in systemic congenital malformation syndromes. One important example is CHARGE syndrome, a genetic disorder characterized by coloboma, heart defects, choanal atresia, growth retardation, genital abnormalities, and ear abnormalities. Mutations in the gene encoding Chromodomain helicase DNA binding protein 7 (CHD7) cause the majority of CHARGE syndrome cases. However, the pathogenetic mechanisms that connect loss of CHD7 to the ocular complications observed in CHARGE syndrome have not been identified. In this review, we provide a general overview of ocular development and congenital disorders affecting the eye. This is followed by a comprehensive description of CHARGE syndrome, including discussion of the spectrum of ocular defects that have been described in this disorder. In addition, we discuss the current knowledge of CHD7 function and focus on its contributions to the development of ocular structures. Finally, we discuss outstanding gaps in our knowledge of the role of CHD7 in eye formation, and propose avenues of investigation to further our understanding of how CHD7 activity regulates ocular and retinal development.
Collapse
Affiliation(s)
| | - Ann C. Morris
- Department of Biology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
12
|
Jofré DM, Hoffman DK, Cervino AS, Hahn GM, Grundy M, Yun S, Amrit FRG, Stolz DB, Godoy LF, Salvatore E, Rossi FA, Ghazi A, Cirio MC, Yanowitz JL, Hochbaum D. The CHARGE syndrome ortholog CHD-7 regulates TGF-β pathways in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2022; 119:e2109508119. [PMID: 35394881 PMCID: PMC9169646 DOI: 10.1073/pnas.2109508119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 02/24/2022] [Indexed: 11/18/2022] Open
Abstract
CHARGE syndrome is a complex developmental disorder caused by mutations in the chromodomain helicase DNA-binding protein-7 (CHD7) and characterized by retarded growth and malformations in the heart and nervous system. Despite the public health relevance of this disorder, relevant cellular pathways and targets of CHD7 that relate to disease pathology are still poorly understood. Here we report that chd-7, the nematode ortholog of Chd7, is required for dauer morphogenesis, lifespan determination, stress response, and body size determination. Consistent with our discoveries, we found chd-7 to be allelic to scd-3, a previously identified dauer suppressor from the DAF-7/ tumor growth factor-β (TGF-β) pathway. Epistatic analysis places CHD-7 at the level of the DAF-3/DAF-5 complex, but we found that CHD-7 also directly impacts the expression of multiple components of this pathway. Transcriptomic analysis revealed that chd-7 mutants fail to repress daf-9 for execution of the dauer program. In addition, CHD-7 regulates the DBL-1/BMP pathway components and shares roles in male tail development and cuticle synthesis. To explore a potential conserved function for chd-7 in vertebrates, we used Xenopus laevis embryos, an established model to study craniofacial development. Morpholino-mediated knockdown of Chd7 led to a reduction in col2a1 messenger RNA (mRNA) levels, a collagen whose expression depends on TGF-β signaling. Both embryonic lethality and craniofacial defects in Chd7-depleted tadpoles were partially rescued by overexpression of col2a1 mRNA. We suggest that Chd7 has conserved roles in regulation of the TGF-β signaling pathway and pathogenic Chd7 could lead to a defective extracellular matrix deposition.
Collapse
Affiliation(s)
- Diego M. Jofré
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | | | - Ailen S. Cervino
- Instituto de Fisiología, Biología Molecular y Neurociencias, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | - Gabriella M. Hahn
- Interdisciplinary Biomedical Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | | | - Sijung Yun
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20814
| | - Francis R. G. Amrit
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Donna B. Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA 15213
| | - Luciana F. Godoy
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | - Esteban Salvatore
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | - Fabiana A. Rossi
- Instituto de Investigaciones en Medicina Traslacional, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Austral, B1630 Pilar, Argentina
| | - Arjumand Ghazi
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Department of Cell Biology & Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - M. Cecilia Cirio
- Instituto de Fisiología, Biología Molecular y Neurociencias, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| | - Judith L. Yanowitz
- Magee-Womens Research Institute, Pittsburgh, PA 15213
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213
| | - Daniel Hochbaum
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1053 Buenos Aires, Argentina
| |
Collapse
|
13
|
Cerrizuela S, Vega-Lopez GA, Méndez-Maldonado K, Velasco I, Aybar MJ. The crucial role of model systems in understanding the complexity of cell signaling in human neurocristopathies. WIREs Mech Dis 2022; 14:e1537. [PMID: 35023327 DOI: 10.1002/wsbm.1537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 11/07/2022]
Abstract
Animal models are useful to study the molecular, cellular, and morphogenetic mechanisms underlying normal and pathological development. Cell-based study models have emerged as an alternative approach to study many aspects of human embryonic development and disease. The neural crest (NC) is a transient, multipotent, and migratory embryonic cell population that generates a diverse group of cell types that arises during vertebrate development. The abnormal formation or development of the NC results in neurocristopathies (NCPs), which are characterized by a broad spectrum of functional and morphological alterations. The impaired molecular mechanisms that give rise to these multiphenotypic diseases are not entirely clear yet. This fact, added to the high incidence of these disorders in the newborn population, has led to the development of systematic approaches for their understanding. In this article, we have systematically reviewed the ways in which experimentation with different animal and cell model systems has improved our knowledge of NCPs, and how these advances might contribute to the development of better diagnostic and therapeutic tools for the treatment of these pathologies. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics Congenital Diseases > Stem Cells and Development Congenital Diseases > Molecular and Cellular Physiology Neurological Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Santiago Cerrizuela
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina
| | - Guillermo A Vega-Lopez
- Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Karla Méndez-Maldonado
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.,Departamento de Fisiología y Farmacología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.,Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, UNAM en el Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Ciudad de México, Mexico
| | - Manuel J Aybar
- Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| |
Collapse
|
14
|
Peusner KD, Bell NM, Hirsch JC, Beraneck M, Popratiloff A. Understanding the Pathophysiology of Congenital Vestibular Disorders: Current Challenges and Future Directions. Front Neurol 2021; 12:708395. [PMID: 34589045 PMCID: PMC8475631 DOI: 10.3389/fneur.2021.708395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
In congenital vestibular disorders (CVDs), children develop an abnormal inner ear before birth and face postnatal challenges to maintain posture, balance, walking, eye-hand coordination, eye tracking, or reading. Only limited information on inner ear pathology is acquired from clinical imaging of the temporal bone or studying histological slides of the temporal bone. A more comprehensive and precise assessment and determination of the underlying mechanisms necessitate analyses of the disorders at the cellular level, which can be achieved using animal models. Two main criteria for a suitable animal model are first, a pathology that mirrors the human disorder, and second, a reproducible experimental outcome leading to statistical power. With over 40 genes that affect inner ear development, the phenotypic abnormalities resulting from congenital vestibular disorders (CVDs) are highly variable. Nonetheless, there is a large subset of CVDs that form a common phenotype of a sac-like inner ear with the semicircular canals missing or dysplastic, and discrete abnormalities in the vestibular sensory organs. We have focused the review on this subset, but to advance research on CVDs we have added other CVDs not forming a sac-like inner ear. We have included examples of animal models used to study these CVDs. Presently, little is known about the central pathology resulting from CVDs at the cellular level in the central vestibular neural network, except for preliminary studies on a chick model that show significant loss of second-order, vestibular reflex projection neurons.
Collapse
Affiliation(s)
- Kenna D Peusner
- Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Nina M Bell
- Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - June C Hirsch
- Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Mathieu Beraneck
- Université de Paris, Integrative Neuroscience and Cognition Center, CNRS UMR 8002, Paris, France
| | - Anastas Popratiloff
- The George Washington University Nanofabrication and Imaging Center, Washington, DC, United States
| |
Collapse
|
15
|
Jamadagni P, Breuer M, Schmeisser K, Cardinal T, Kassa B, Parker JA, Pilon N, Samarut E, Patten SA. Chromatin remodeller CHD7 is required for GABAergic neuron development by promoting PAQR3 expression. EMBO Rep 2021; 22:e50958. [PMID: 33900016 PMCID: PMC8183419 DOI: 10.15252/embr.202050958] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 03/07/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Mutations in the chromatin remodeller‐coding gene CHD7 cause CHARGE syndrome (CS). CS features include moderate to severe neurological and behavioural problems, clinically characterized by intellectual disability, attention‐deficit/hyperactivity disorder and autism spectrum disorder. To investigate the poorly characterized neurobiological role of CHD7, we here generate a zebrafish chd7−/− model. chd7−/− mutants have less GABAergic neurons and exhibit a hyperactivity behavioural phenotype. The GABAergic neuron defect is at least in part due to downregulation of the CHD7 direct target gene paqr3b, and subsequent upregulation of MAPK/ERK signalling, which is also dysregulated in CHD7 mutant human cells. Through a phenotype‐based screen in chd7−/− zebrafish and Caenorhabditis elegans, we show that the small molecule ephedrine restores normal levels of MAPK/ERK signalling and improves both GABAergic defects and behavioural anomalies. We conclude that chd7 promotes paqr3b expression and that this is required for normal GABAergic network development. This work provides insight into the neuropathogenesis associated with CHD7 deficiency and identifies a promising compound for further preclinical studies.
Collapse
Affiliation(s)
| | - Maximilian Breuer
- INRS- Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Kathrin Schmeisser
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Tatiana Cardinal
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montréal, QC, Canada
| | - Betelhem Kassa
- INRS- Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - J Alex Parker
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Modelis inc., Montréal, QC, Canada
| | - Nicolas Pilon
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montréal, QC, Canada.,Département des sciences biologiques, Université du Québec à Montréal (UQAM), Montréal, QC, Canada.,Département de pédiatrie, Université de Montréal, Montréal, QC, Canada
| | - Eric Samarut
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Modelis inc., Montréal, QC, Canada
| | - Shunmoogum A Patten
- INRS- Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montréal, QC, Canada
| |
Collapse
|
16
|
De Luca C, Picone S, Cassina M, Marziali S, Morlino S, Camerota L, Tamburrini G, Castori M, Paolillo P, Salviati L, Brancati F. Craniosynostosis is a feature of CHD7-related CHARGE syndrome. Am J Med Genet A 2021; 185:2160-2163. [PMID: 33844462 DOI: 10.1002/ajmg.a.62208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 11/08/2022]
Abstract
CHARGE syndrome is a rare genetic multiple-malformation disorder characterized by wide phenotypic variability. It is often caused by heterozygous variants in CHD7 and, more rarely, SEMA3E. Although craniofacial alterations are frequent in this condition, to date craniosynostosis is not considered part of the clinical spectrum. Here, we report bi-coronal craniosynostosis in a newborn affected by CHARGE syndrome caused by the de novo heterozygous c.6157C>T, p.(Arg2053*) CHD7 variant. We found two additional subjects in the literature with different craniosynostoses and distinct CHD7 alterations. The inclusion of CHD7-related CHARGE syndrome in the group of rare causes of syndromic craniosynostoses is proposed.
Collapse
Affiliation(s)
- Chiara De Luca
- Human Genetics, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simonetta Picone
- Neonatology and Neonatal Intensive Care, Policlinico Casilino Hospital, Rome, Italy
| | - Matteo Cassina
- Clinical Genetics, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Simone Marziali
- Division of Neuroradiology, Policlinico Casilino Hospital, Rome, Italy
| | - Silvia Morlino
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Letizia Camerota
- Human Genetics, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Gianpiero Tamburrini
- Pediatric Neurosurgery, Institute of Neurosurgery, Fondazione Policlinico Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Piermichele Paolillo
- Neonatology and Neonatal Intensive Care, Policlinico Casilino Hospital, Rome, Italy
| | - Leonardo Salviati
- Clinical Genetics, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Francesco Brancati
- Human Genetics, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Division of Human Functional Genomics, San Raffaele Pisana IRCCS, Rome, Italy
| |
Collapse
|
17
|
Lettieri A, Oleari R, Paganoni AJJ, Gervasini C, Massa V, Fantin A, Cariboni A. Semaphorin Regulation by the Chromatin Remodeler CHD7: An Emerging Genetic Interaction Shaping Neural Cells and Neural Crest in Development and Cancer. Front Cell Dev Biol 2021; 9:638674. [PMID: 33869187 PMCID: PMC8047133 DOI: 10.3389/fcell.2021.638674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
CHD7 is a chromatin remodeler protein that controls gene expression via the formation of multi-protein complexes with specific transcription factors. During development, CHD7 controls several differentiation programs, mainly by acting on neural progenitors and neural crest (NC) cells. Thus, its roles range from the central nervous system to the peripheral nervous system and the organs colonized by NC cells, including the heart. Accordingly, mutated CHD7 is linked to CHARGE syndrome, which is characterized by several neuronal dysfunctions and by malformations of NC-derived/populated organs. Altered CHD7 has also been associated with different neoplastic transformations. Interestingly, recent evidence revealed that semaphorins, a class of molecules involved in developmental and pathological processes similar to those controlled by CHD7, are regulated by CHD7 in a context-specific manner. In this article, we will review the recent insights that support the existence of genetic interactions between these pathways, both during developmental processes and cancer progression.
Collapse
Affiliation(s)
- Antonella Lettieri
- CRC Aldo Ravelli for Neurotechnology and Experimental Brain Therapeutics, Università degli Studi di Milano, Milan, Italy.,Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alyssa J J Paganoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Cristina Gervasini
- CRC Aldo Ravelli for Neurotechnology and Experimental Brain Therapeutics, Università degli Studi di Milano, Milan, Italy.,Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valentina Massa
- CRC Aldo Ravelli for Neurotechnology and Experimental Brain Therapeutics, Università degli Studi di Milano, Milan, Italy.,Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alessandro Fantin
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
18
|
Raeisossadati R, Ferrari MFR, Kihara AH, AlDiri I, Gross JM. Epigenetic regulation of retinal development. Epigenetics Chromatin 2021; 14:11. [PMID: 33563331 PMCID: PMC7871400 DOI: 10.1186/s13072-021-00384-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/28/2021] [Indexed: 01/10/2023] Open
Abstract
In the developing vertebrate retina, retinal progenitor cells (RPCs) proliferate and give rise to terminally differentiated neurons with exquisite spatio-temporal precision. Lineage commitment, fate determination and terminal differentiation are controlled by intricate crosstalk between the genome and epigenome. Indeed, epigenetic regulation plays pivotal roles in numerous cell fate specification and differentiation events in the retina. Moreover, aberrant chromatin structure can contribute to developmental disorders and retinal pathologies. In this review, we highlight recent advances in our understanding of epigenetic regulation in the retina. We also provide insight into several aspects of epigenetic-related regulation that should be investigated in future studies of retinal development and disease. Importantly, focusing on these mechanisms could contribute to the development of novel treatment strategies targeting a variety of retinal disorders.
Collapse
Affiliation(s)
- Reza Raeisossadati
- Departamento de Genética E Biologia Evolutiva, Instituto de Biociencias, Universidade de Sao Paulo, Rua Do Matao, 277, Cidade Universitaria, Sao Paulo, SP, 05508-090, Brazil.,Departments of Ophthalmology and Developmental Biology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Merari F R Ferrari
- Departamento de Genética E Biologia Evolutiva, Instituto de Biociencias, Universidade de Sao Paulo, Rua Do Matao, 277, Cidade Universitaria, Sao Paulo, SP, 05508-090, Brazil
| | | | - Issam AlDiri
- Departments of Ophthalmology and Developmental Biology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey M Gross
- Departments of Ophthalmology and Developmental Biology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
19
|
Liu S, Narumi R, Ikeda N, Morita O, Tasaki J. Chemical-induced craniofacial anomalies caused by disruption of neural crest cell development in a zebrafish model. Dev Dyn 2020; 249:794-815. [PMID: 32314458 PMCID: PMC7384000 DOI: 10.1002/dvdy.179] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/11/2022] Open
Abstract
Background Craniofacial anomalies are among the most frequent birth defects worldwide, and are thought to be caused by gene‐environment interactions. Genetically manipulated zebrafish simulate human diseases and provide great advantages for investigating the etiology and pathology of craniofacial anomalies. Although substantial advances have been made in understanding genetic factors causing craniofacial disorders, limited information about the etiology by which environmental factors, such as teratogens, induce craniofacial anomalies is available in zebrafish. Results Zebrafish embryos displayed craniofacial malformations after teratogen treatments. Further observations revealed characteristic disruption of chondrocyte number, shape and stacking. These findings suggested aberrant development of cranial neural crest (CNC) cells, which was confirmed by gene expression analysis of the CNC. Notably, these observations suggested conserved etiological pathways between zebrafish and mammals including human. Furthermore, several of these chemicals caused malformations of the eyes, otic vesicle, and/or heart, representing a phenocopy of neurocristopathy, and these chemicals altered the expression levels of the responsible genes. Conclusions Our results demonstrate that chemical‐induced craniofacial malformation is caused by aberrant development of neural crest. This study indicates that zebrafish provide a platform for investigating contributions of environmental factors as causative agents of craniofacial anomalies and neurocristopathy.
Collapse
Affiliation(s)
- Shujie Liu
- R&D, Safety Science Research, Kao Corporation, Tochigi, Japan
| | - Rika Narumi
- R&D, Safety Science Research, Kao Corporation, Tochigi, Japan
| | - Naohiro Ikeda
- R&D, Safety Science Research, Kao Corporation, Tochigi, Japan
| | - Osamu Morita
- R&D, Safety Science Research, Kao Corporation, Tochigi, Japan
| | - Junichi Tasaki
- R&D, Safety Science Research, Kao Corporation, Tochigi, Japan
| |
Collapse
|
20
|
Al Dhaheri N, Wu N, Zhao S, Wu Z, Blank RD, Zhang J, Raggio C, Halanski M, Shen J, Noonan K, Qiu G, Nemeth B, Sund S, Dunwoodie SL, Chapman G, Glurich I, Steiner RD, Wohler E, Martin R, Sobreira NL, Giampietro PF. KIAA1217: A novel candidate gene associated with isolated and syndromic vertebral malformations. Am J Med Genet A 2020; 182:1664-1672. [PMID: 32369272 DOI: 10.1002/ajmg.a.61607] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 04/05/2020] [Accepted: 04/05/2020] [Indexed: 12/30/2022]
Abstract
Vertebral malformations (VMs) are caused by alterations in somitogenesis and may occur in association with other congenital anomalies. The genetic etiology of most VMs remains unknown and their identification may facilitate the development of novel therapeutic and prevention strategies. Exome sequencing was performed on both the discovery cohort of nine unrelated probands from the USA with VMs and the replication cohort from China (Deciphering Disorders Involving Scoliosis & COmorbidities study). The discovery cohort was analyzed using the PhenoDB analysis tool. Heterozygous and homozygous, rare and functional variants were selected and evaluated for their ClinVar, HGMD, OMIM, GWAS, mouse model phenotypes, and other annotations to identify the best candidates. Genes with candidate variants in three or more probands were selected. The replication cohort was analyzed by another in-house developed pipeline. We identified rare heterozygous variants in KIAA1217 in four out of nine probands in the discovery cohort and in five out of 35 probands in the replication cohort. Collectively, we identified 11 KIAA1217 rare variants in 10 probands, three of which have not been described in gnomAD and one of which is a nonsense variant. We propose that genetic variations of KIAA1217 may contribute to the etiology of VMs.
Collapse
Affiliation(s)
- Noura Al Dhaheri
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, College of Medicine and Health Sciences, UAE University, Al-Ain, UAE
| | - Nan Wu
- Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Sen Zhao
- Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Wu
- Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | - Jianguo Zhang
- Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Cathy Raggio
- Hospital for Special Surgery, New York, New York, USA
| | | | - Jianxiong Shen
- Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ken Noonan
- University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Guixing Qiu
- Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Blaise Nemeth
- University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sarah Sund
- University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sally L Dunwoodie
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,University of New South Wales, Sydney, New South Wales, Australia
| | - Gavin Chapman
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,University of New South Wales, Sydney, New South Wales, Australia
| | - Ingrid Glurich
- Marshfield Clinic Research Institute, Marshfield, Wisconsin, USA
| | - Robert D Steiner
- University of Wisconsin-Madison, Madison, Wisconsin, USA.,Marshfield Clinic Research Institute, Marshfield, Wisconsin, USA
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Renan Martin
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nara Lygia Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
21
|
Schwenty-Lara J, Nehl D, Borchers A. The histone methyltransferase KMT2D, mutated in Kabuki syndrome patients, is required for neural crest cell formation and migration. Hum Mol Genet 2020; 29:305-319. [PMID: 31813957 PMCID: PMC7003132 DOI: 10.1093/hmg/ddz284] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 12/30/2022] Open
Abstract
Kabuki syndrome is an autosomal dominant developmental disorder with high similarities to CHARGE syndrome. It is characterized by a typical facial gestalt in combination with short stature, intellectual disability, skeletal findings and additional features like cardiac and urogenital malformations, cleft palate, hearing loss and ophthalmological anomalies. The major cause of Kabuki syndrome are mutations in KMT2D, a gene encoding a histone H3 lysine 4 (H3K4) methyltransferase belonging to the group of chromatin modifiers. Here we provide evidence that Kabuki syndrome is a neurocrestopathy, by showing that Kmt2d loss-of-function inhibits specific steps of neural crest (NC) development. Using the Xenopus model system, we find that Kmt2d loss-of-function recapitulates major features of Kabuki syndrome including severe craniofacial malformations. A detailed marker analysis revealed defects in NC formation as well as migration. Transplantation experiments confirm that Kmt2d function is required in NC cells. Furthermore, analyzing in vivo and in vitro NC migration behavior demonstrates that Kmt2d is necessary for cell dispersion but not protrusion formation of migrating NC cells. Importantly, Kmt2d knockdown correlates with a decrease in H3K4 monomethylation and H3K27 acetylation supporting a role of Kmt2d in the transcriptional activation of target genes. Consistently, using a candidate approach, we find that Kmt2d loss-of-function inhibits Xenopus Sema3F expression, and overexpression of Sema3F can partially rescue Kmt2d loss-of-function defects. Taken together, our data reveal novel functions of Kmt2d in multiple steps of NC development and support the hypothesis that major features of Kabuki syndrome are caused by defects in NC development.
Collapse
Affiliation(s)
- Janina Schwenty-Lara
- Department of Biology, Molecular Embryology, Philipps-Universität Marburg, Marburg 35043, Germany
| | - Denise Nehl
- Department of Biology, Molecular Embryology, Philipps-Universität Marburg, Marburg 35043, Germany
| | - Annette Borchers
- Department of Biology, Molecular Embryology, Philipps-Universität Marburg, Marburg 35043, Germany
- DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg 35043, Germany
| |
Collapse
|
22
|
Asad Z, Sachidanandan C. Chemical screens in a zebrafish model of CHARGE syndrome identifies small molecules that ameliorate disease-like phenotypes in embryo. Eur J Med Genet 2019; 63:103661. [PMID: 31051269 DOI: 10.1016/j.ejmg.2019.04.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/09/2019] [Accepted: 04/28/2019] [Indexed: 01/03/2023]
Abstract
CHARGE syndrome is an autosomal dominant congenital disorder caused primarily by mutations in the CHD7 gene. Using a small molecule screen in a zebrafish model of CHARGE syndrome, we identified 4 compounds that rescue embryos from disease-like phenotypes. Our screen yielded DAPT, a Notch signaling inhibitor that could ameliorate the craniofacial, cranial neuronal and myelination defects in chd7 morphant zebrafish embryos. We discovered that Procainamide, an inhibitor of DNA methyltransferase 1, was able to recover the pattern of expression of isl2a, a cranial neuronal marker while also reducing the effect on craniofacial cartilage and myelination. M344, an inhibitor of Histone deacetylases had a strong recovery effect on craniofacial cartilage defects and could also modestly revert the myelination defects in zebrafish embryos. CHIC-35, a SIRT1 inhibitor partially restored the expression of isl2a in cranial neurons while causing a partial reversion of myelination and craniofacial cartilage defects. Our results suggest that a modular approach to phenotypic rescue in multi-organ syndromes might be a more successful approach to treat these disorders. Our findings also open up the possibility of using these compounds for other disorders with shared phenotypes.
Collapse
Affiliation(s)
- Zainab Asad
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, 110025, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Chetana Sachidanandan
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, 110025, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
23
|
Machado RAC, Schneider H, DeOcesano-Pereira C, Lichtenstein F, Andrade F, Fujita A, Trombetta-Lima M, Weller M, Bowman-Colin C, Sogayar MC. CHD7 promotes glioblastoma cell motility and invasiveness through transcriptional modulation of an invasion signature. Sci Rep 2019; 9:3952. [PMID: 30850678 PMCID: PMC6408455 DOI: 10.1038/s41598-019-39564-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/10/2019] [Indexed: 01/21/2023] Open
Abstract
Chromatin remodeler proteins exert an important function in promoting dynamic modifications in the chromatin architecture, performing a central role in regulating gene transcription. Deregulation of these molecular machines may lead to striking perturbations in normal cell function. The CHD7 gene is a member of the chromodomain helicase DNA-binding family and, when mutated, has been shown to be the cause of the CHARGE syndrome, a severe developmental human disorder. Moreover, CHD7 has been described to be essential for neural stem cells and it is also highly expressed or mutated in a number of human cancers. However, its potential role in glioblastoma has not yet been tested. Here, we show that CHD7 is up-regulated in human glioma tissues and we demonstrate that CHD7 knockout (KO) in LN-229 glioblastoma cells suppresses anchorage-independent growth and spheroid invasion in vitro. Additionally, CHD7 KO impairs tumor growth and increases overall survival in an orthotopic mouse xenograft model. Conversely, ectopic overexpression of CHD7 in LN-428 and A172 glioblastoma cell lines increases cell motility and invasiveness in vitro and promotes LN-428 tumor growth in vivo. Finally, RNA-seq analysis revealed that CHD7 modulates a specific transcriptional signature of invasion-related target genes. Further studies should explore clinical-translational implications for glioblastoma treatment.
Collapse
Affiliation(s)
- Raquel A C Machado
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo 05508-000 SP, Brazil
- Cell and Molecular Therapy Center (NUCEL), Internal Medicine Department, School of Medicine, University of São Paulo, São Paulo 05360-130 SP, Brazil
| | - Hannah Schneider
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Carlos DeOcesano-Pereira
- Cell and Molecular Therapy Center (NUCEL), Internal Medicine Department, School of Medicine, University of São Paulo, São Paulo 05360-130 SP, Brazil
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP, Brazil
| | - Flavio Lichtenstein
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP, Brazil
| | - Fernando Andrade
- Department of Computer Science, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, Brazil
| | - André Fujita
- Department of Computer Science, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, Brazil
| | - Marina Trombetta-Lima
- Cell and Molecular Therapy Center (NUCEL), Internal Medicine Department, School of Medicine, University of São Paulo, São Paulo 05360-130 SP, Brazil
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Christian Bowman-Colin
- Cell and Molecular Therapy Center (NUCEL), Internal Medicine Department, School of Medicine, University of São Paulo, São Paulo 05360-130 SP, Brazil.
- Dana Farber Cancer Institute, Harvard Medical School, 1 Jimmi Fund Way - SM808, Boston, MA, USA.
| | - Mari Cleide Sogayar
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo 05508-000 SP, Brazil.
- Cell and Molecular Therapy Center (NUCEL), Internal Medicine Department, School of Medicine, University of São Paulo, São Paulo 05360-130 SP, Brazil.
| |
Collapse
|
24
|
Moore-Morris T, van Vliet PP, Andelfinger G, Puceat M. Role of Epigenetics in Cardiac Development and Congenital Diseases. Physiol Rev 2019; 98:2453-2475. [PMID: 30156497 DOI: 10.1152/physrev.00048.2017] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The heart is the first organ to be functional in the fetus. Heart formation is a complex morphogenetic process regulated by both genetic and epigenetic mechanisms. Congenital heart diseases (CHD) are the most prominent congenital diseases. Genetics is not sufficient to explain these diseases or the impact of them on patients. Epigenetics is more and more emerging as a basis for cardiac malformations. This review brings the essential knowledge on cardiac biology of development. It further provides a broad background on epigenetics with a focus on three-dimensional conformation of chromatin. Then, we summarize the current knowledge of the impact of epigenetics on cardiac cell fate decision. We further provide an update on the epigenetic anomalies in the genesis of CHD.
Collapse
Affiliation(s)
- Thomas Moore-Morris
- Université Aix-Marseille, INSERM UMR- 1251, Marseille , France ; Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec , Canada ; Université de Montréal, Montreal, Quebec , Canada ; and Laboratoire International Associé INSERM, Marseille France-CHU Ste Justine, Quebec, Canada
| | - Patrick Piet van Vliet
- Université Aix-Marseille, INSERM UMR- 1251, Marseille , France ; Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec , Canada ; Université de Montréal, Montreal, Quebec , Canada ; and Laboratoire International Associé INSERM, Marseille France-CHU Ste Justine, Quebec, Canada
| | - Gregor Andelfinger
- Université Aix-Marseille, INSERM UMR- 1251, Marseille , France ; Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec , Canada ; Université de Montréal, Montreal, Quebec , Canada ; and Laboratoire International Associé INSERM, Marseille France-CHU Ste Justine, Quebec, Canada
| | - Michel Puceat
- Université Aix-Marseille, INSERM UMR- 1251, Marseille , France ; Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Montreal, Quebec , Canada ; Université de Montréal, Montreal, Quebec , Canada ; and Laboratoire International Associé INSERM, Marseille France-CHU Ste Justine, Quebec, Canada
| |
Collapse
|
25
|
Multiple muscular abnormalities in a fetal cadaver with CHARGE syndrome. Surg Radiol Anat 2018; 41:601-605. [PMID: 30465074 DOI: 10.1007/s00276-018-2137-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
Abstract
The CHARGE syndrome characterized by coloboma, heart defects, atresia of the choanae, retarded growth, genitourinary hypoplasia, and ear anomalies is one of the rare syndromes. Although certain clinical issues (scapular winging, sloping shoulder, Sprengel's deformity, kyphosis and scoliosis) which could be related to abnormalities in musculoskeletal structures of the neck and shoulder have been identified in CHARGE syndrome, data on details of muscle anomalies seem to be quite limited in the literature. In this case report, bilateral multiple muscular abnormalities (agenesis of the trapezius, presence of the rhombo-atloid muscle, and presence of the bipartite rhomboid minor with superficial and deep parts) was presented in a fetus cadaver with atypical CHARGE syndrome to attract the attention of clinicians for definitive diagnoses and surgical reconstruction of the shoulders deformity such as scapular winging and Sprengel's deformity. By considering the previous studies, we propose that the absence of the trapezius, as well as the other muscle abnormalities around the neck and shoulder, should be revised as being a component of CHARGE syndrome.
Collapse
|
26
|
Kasah S, Oddy C, Basson MA. Autism-linked CHD gene expression patterns during development predict multi-organ disease phenotypes. J Anat 2018; 233:755-769. [PMID: 30277262 DOI: 10.1111/joa.12889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2018] [Indexed: 12/24/2022] Open
Abstract
Recent large-scale exome sequencing studies have identified mutations in several members of the CHD (Chromodomain Helicase DNA-binding protein) gene family in neurodevelopmental disorders. Mutations in the CHD2 gene have been linked to developmental delay, intellectual disability, autism and seizures, CHD8 mutations to autism and intellectual disability, whereas haploinsufficiency of CHD7 is associated with executive dysfunction and intellectual disability. In addition to these neurodevelopmental features, a wide range of other developmental defects are associated with mutants of these genes, especially with regards to CHD7 haploinsufficiency, which is the primary cause of CHARGE syndrome. Whilst the developmental expression of CHD7 has been reported previously, limited information on the expression of CHD2 and CHD8 during development is available. Here, we compare the expression patterns of all three genes during mouse development directly. We find high, widespread expression of these genes at early stages of development that gradually becomes restricted during later developmental stages. Chd2 and Chd8 are widely expressed in the developing central nervous system (CNS) at all stages of development, with moderate expression remaining in the neocortex, hippocampus, olfactory bulb and cerebellum of the postnatal brain. Similarly, Chd7 expression is seen throughout the CNS during late embryogenesis and early postnatal development, with strong enrichment in the cerebellum, but displays low expression in the cortex and neurogenic niches in early life. In addition to expression in the brain, novel sites of Chd2 and Chd8 expression are reported. These findings suggest additional roles for these genes in organogenesis and predict that mutation of these genes may predispose individuals to a range of other, non-neurological developmental defects.
Collapse
Affiliation(s)
- Sahrunizam Kasah
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Christopher Oddy
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| |
Collapse
|
27
|
How Surrogate and Chemical Genetics in Model Organisms Can Suggest Therapies for Human Genetic Diseases. Genetics 2018; 208:833-851. [PMID: 29487144 PMCID: PMC5844338 DOI: 10.1534/genetics.117.300124] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/26/2017] [Indexed: 12/12/2022] Open
Abstract
Genetic diseases are both inherited and acquired. Many genetic diseases fall under the paradigm of orphan diseases, a disease found in < 1 in 2000 persons. With rapid and cost-effective genome sequencing becoming the norm, many causal mutations for genetic diseases are being rapidly determined. In this regard, model organisms are playing an important role in validating if specific mutations identified in patients drive the observed phenotype. An emerging challenge for model organism researchers is the application of genetic and chemical genetic platforms to discover drug targets and drugs/drug-like molecules for potential treatment options for patients with genetic disease. This review provides an overview of how model organisms have contributed to our understanding of genetic disease, with a focus on the roles of yeast and zebrafish in gene discovery and the identification of compounds that could potentially treat human genetic diseases.
Collapse
|
28
|
de Geus CM, Bergman JEH, van Ravenswaaij-Arts CMA, Meiners LC. Imaging of Clival Hypoplasia in CHARGE Syndrome and Hypothesis for Development: A Case-Control Study. AJNR Am J Neuroradiol 2018; 39:1938-1942. [PMID: 30237300 PMCID: PMC7410728 DOI: 10.3174/ajnr.a5810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/19/2018] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE We present the largest case series to date on basiocciput abnormalities in CHARGE syndrome (Coloboma of the eye, Heart defects, Atresia of the choanae, Retardation of growth and/or development, Genital and/or urinary abnormalities, and Ear abnormalities and/or deafness). We aimed to show that basiocciput abnormalities are common and may aid in diagnosis. We furthermore explored whether clivus size correlates with the type of chromodomain-helicase-DNA binding protein 7 gene (CHD7) mutation, which causes CHARGE syndrome, and with clinical criteria according to Blake et al and Verloes. MATERIALS AND METHODS We retrospectively analyzed the clivus of 23 patients with CHARGE syndrome with CHD7 mutations on MR imaging or CT. We recorded the size of the clivus, the Welcher angle, basilar invagination, and Chiari I malformations. We compared the clival size and Welcher angle of patients with CHARGE syndrome with those of 72 age-matched controls. Additionally, we tested for correlations between clivus size and mutation type or clinical criteria. RESULTS Eighty-seven percent of the patients with CHARGE syndrome had an abnormal clivus; 61% had a clivus >2.5 SD smaller than that of age-matched controls. An abnormally large Welcher angle was observed in 35%. Basiocciput hypoplasia was found in 70%, and basilar invagination, in 29%. None of the patients had a Chiari I malformation. At the group level, patients with CHARGE syndrome had a smaller clivus and larger Welcher angle than controls. No significant correlation between clivus size and mutation type or clinical criteria was found. CONCLUSIONS Most patients with CHARGE syndrome have an abnormal clivus. This suggests that clivus abnormalities may be used as an additional diagnostic tool. Our results provide evidence that CHD7, which is expressed in the presomitic mesoderm during somitogenesis, plays an important role in the formation of the clivus.
Collapse
Affiliation(s)
- C M de Geus
- From the Departments of Genetics (C.M.d.G., J.E.H.B., C.M.A.v.R.)
| | - J E H Bergman
- From the Departments of Genetics (C.M.d.G., J.E.H.B., C.M.A.v.R.)
| | | | - L C Meiners
- Radiology (L.C.M.), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
29
|
Sakai C, Ijaz S, Hoffman EJ. Zebrafish Models of Neurodevelopmental Disorders: Past, Present, and Future. Front Mol Neurosci 2018; 11:294. [PMID: 30210288 PMCID: PMC6123572 DOI: 10.3389/fnmol.2018.00294] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/03/2018] [Indexed: 12/21/2022] Open
Abstract
Zebrafish are increasingly being utilized as a model system to investigate the function of the growing list of risk genes associated with neurodevelopmental disorders. This is due in large part to the unique features of zebrafish that make them an optimal system for this purpose, including rapid, external development of transparent embryos, which enable the direct visualization of the developing nervous system during early stages, large progenies, which provide considerable tractability for performing high-throughput pharmacological screens to identify small molecule suppressors of simple behavioral phenotypes, and ease of genetic manipulation, which has been greatly facilitated by the advent of CRISPR/Cas9 gene editing technologies. This review article focuses on studies that have harnessed these advantages of the zebrafish system for the functional analysis of genes that are strongly associated with the following neurodevelopmental disorders: autism spectrum disorders (ASD), epilepsy, intellectual disability (ID) and schizophrenia. We focus primarily on studies describing early morphological and behavioral phenotypes during embryonic and larval stages resulting from loss of risk gene function. We highlight insights into basic mechanisms of risk gene function gained from these studies as well as limitations of studies to date. Finally, we discuss advances in in vivo neural circuit imaging in zebrafish, which promise to transform research using the zebrafish model by illuminating novel circuit-level mechanisms with relevance to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Catalina Sakai
- Child Study Center, Program on Neurogenetics, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Sundas Ijaz
- Child Study Center, Program on Neurogenetics, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Ellen J Hoffman
- Child Study Center, Program on Neurogenetics, Yale School of Medicine, Yale University, New Haven, CT, United States.,Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
30
|
Tordjman S, Cohen D, Anderson G, Botbol M, Canitano R, Coulon N, Roubertoux P. Repint of “Reframing autism as a behavioral syndrome and not a specific mental disorder: Implications of genetic and phenotypic heterogeneity”. Neurosci Biobehav Rev 2018; 89:132-150. [DOI: 10.1016/j.neubiorev.2018.01.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/18/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022]
|
31
|
Neurocristopathies: New insights 150 years after the neural crest discovery. Dev Biol 2018; 444 Suppl 1:S110-S143. [PMID: 29802835 DOI: 10.1016/j.ydbio.2018.05.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/16/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022]
Abstract
The neural crest (NC) is a transient, multipotent and migratory cell population that generates an astonishingly diverse array of cell types during vertebrate development. These cells, which originate from the ectoderm in a region lateral to the neural plate in the neural fold, give rise to neurons, glia, melanocytes, chondrocytes, smooth muscle cells, odontoblasts and neuroendocrine cells, among others. Neurocristopathies (NCP) are a class of pathologies occurring in vertebrates, especially in humans that result from the abnormal specification, migration, differentiation or death of neural crest cells during embryonic development. Various pigment, skin, thyroid and hearing disorders, craniofacial and heart abnormalities, malfunctions of the digestive tract and tumors can also be considered as neurocristopathies. In this review we revisit the current classification and propose a new way to classify NCP based on the embryonic origin of the affected tissues, on recent findings regarding the molecular mechanisms that drive NC formation, and on the increased complexity of current molecular embryology techniques.
Collapse
|
32
|
Zebrafish Models of Rare Hereditary Pediatric Diseases. Diseases 2018; 6:diseases6020043. [PMID: 29789451 PMCID: PMC6023479 DOI: 10.3390/diseases6020043] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/17/2018] [Accepted: 05/19/2018] [Indexed: 12/12/2022] Open
Abstract
Recent advances in sequencing technologies have made it significantly easier to find the genetic roots of rare hereditary pediatric diseases. These novel methods are not panaceas, however, and they often give ambiguous results, highlighting multiple possible causative mutations in affected patients. Furthermore, even when the mapping results are unambiguous, the affected gene might be of unknown function. In these cases, understanding how a particular genotype can result in a phenotype also needs carefully designed experimental work. Model organism genetics can offer a straightforward experimental setup for hypothesis testing. Containing orthologs for over 80% of the genes involved in human diseases, zebrafish (Danio rerio) has emerged as one of the top disease models over the past decade. A plethora of genetic tools makes it easy to create mutations in almost any gene of the zebrafish genome and these mutant strains can be used in high-throughput preclinical screens for active molecules. As this small vertebrate species offers several other advantages as well, its popularity in biomedical research is bound to increase, with “aquarium to bedside” drug development pipelines taking a more prevalent role in the near future.
Collapse
|
33
|
Cloney K, Steele SL, Stoyek MR, Croll RP, Smith FM, Prykhozhij SV, Brown MM, Midgen C, Blake K, Berman JN. Etiology and functional validation of gastrointestinal motility dysfunction in a zebrafish model of CHARGE syndrome. FEBS J 2018; 285:2125-2140. [PMID: 29660852 DOI: 10.1111/febs.14473] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 03/17/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022]
Abstract
CHARGE syndrome is linked to autosomal-dominant mutations in the CHD7 gene and results in a number of physiological and structural abnormalities, including heart defects, hearing and vision loss, and gastrointestinal (GI) problems. Of these challenges, GI problems have a profound impact throughout an individual's life, resulting in increased morbidity and mortality. A homolog of CHD7 has been identified in the zebrafish, the loss of which recapitulates many of the features of the human disease. Using a morpholino chd7 knockdown model complemented by a chd7 null mutant zebrafish line, we examined GI structure, innervation, and motility in larval zebrafish. Loss of chd7 resulted in physically smaller GI tracts with normal epithelial and muscular histology, but decreased and disorganized vagal projections, particularly in the foregut. chd7 morphant larvae had significantly less ability to empty their GI tract of gavaged fluorescent beads, and this condition was only minimally improved by the prokinetic agents, domperidone and erythromycin, in keeping with mixed responses to these agents in patients with CHARGE syndrome. The conserved genetics and transparency of the zebrafish have provided new insights into the consequences of chd7 gene dysfunction on the GI system and cranial nerve patterning. These findings highlight the opportunity of the zebrafish to serve as a preclinical model for studying compounds that may improve GI motility in individuals with CHARGE syndrome.
Collapse
Affiliation(s)
- Kellie Cloney
- Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Shelby L Steele
- Department of Pediatrics, Dalhousie University, Halifax, Canada
| | - Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Roger P Croll
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Frank M Smith
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | | | - Mary M Brown
- Departments of Pediatrics and Obstetrics and Gynaecology, Dalhousie University, Halifax, Canada
| | - Craig Midgen
- Department of Pathology, Dalhousie University, Halifax, Canada
| | - Kim Blake
- Faculty of Medicine, Dalhousie University, Halifax, Canada.,Department of Pediatrics, Dalhousie University, Halifax, Canada
| | - Jason N Berman
- Department of Pediatrics, Dalhousie University, Halifax, Canada.,Department of Pathology, Dalhousie University, Halifax, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
| |
Collapse
|
34
|
Liu ZZ, Wang ZL, Choi TI, Huang WT, Wang HT, Han YY, Zhu LY, Kim HT, Choi JH, Lee JS, Kim HG, Zhao J, Chen Y, Lu Z, Tian XL, Pan BX, Li BM, Kim CH, Xu HA. Chd7 Is Critical for Early T-Cell Development and Thymus Organogenesis in Zebrafish. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1043-1058. [PMID: 29353058 DOI: 10.1016/j.ajpath.2017.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
Abstract
Coloboma, heart defect, atresia choanae, retarded growth and development, genital hypoplasia, ear anomalies/deafness (CHARGE) syndrome is a congenital disorder affecting multiple organs and mainly caused by mutations in CHD7, a gene encoding a chromatin-remodeling protein. Immunodeficiency and reduced T cells have been noted in CHARGE syndrome. However, the mechanisms underlying T lymphopenia are largely unexplored. Herein, we observed dramatic decrease of T cells in both chd7knockdown and knockout zebrafish embryos. Unexpectedly, hematopoietic stem and progenitor cells and, particularly, lymphoid progenitor cells were increased peripherally in nonthymic areas in chd7-deficient embryos, unlikely to contribute to the T-cell decrease. Further analysis demonstrated that both the organogenesis and homing function of the thymus were seriously impaired. Chd7 might regulate thymus organogenesis through modulating the development of both neural crest cell-derived mesenchyme and pharyngeal endoderm-derived thymic epithelial cells. The expression of foxn1, a central regulator of thymic epithelium, was remarkably down-regulated in the pharyngeal region in chd7-deficient embryos. Moreover, the T-cell reduction in chd7-deficient embryos was partially rescued by overexpressing foxn1, suggesting that restoring thymic epithelium may be a potential therapeutic strategy for treating immunodeficiency in CHARGE syndrome. Collectively, the results indicated that chd7 was critical for thymic development and T-lymphopenia in CHARGE syndrome may be mainly attributed to the defects of thymic organogenesis. The current finding may benefit the diagnosis and therapy of T lymphopenia and immunodeficiency in CHARGE syndrome.
Collapse
Affiliation(s)
- Zhi-Zhi Liu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Zi-Long Wang
- Institute of Life Science, Nanchang University, Nanchang, China; Queen Mary School, Nanchang University, Nanchang, China
| | - Tae-Ik Choi
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Wen-Ting Huang
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Han-Tsing Wang
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Ying-Ying Han
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Lou-Yin Zhu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Hyun-Taek Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jung-Hwa Choi
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jin-Soo Lee
- National Cancer Center, Goyang, Republic of Korea
| | - Hyung-Goo Kim
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia; Children's Hospital of Jiang Xi, Nanchang, China; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia
| | - Jian Zhao
- Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Yue Chen
- Children's Hospital of Jiang Xi, Nanchang, China
| | - Zhuo Lu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Xiao-Li Tian
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Bing-Xing Pan
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Bao-Ming Li
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea.
| | - Hong A Xu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China.
| |
Collapse
|
35
|
van Ravenswaaij-Arts C, Martin DM. New insights and advances in CHARGE syndrome: Diagnosis, etiologies, treatments, and research discoveries. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2017; 175:397-406. [PMID: 29171162 DOI: 10.1002/ajmg.c.31592] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 01/17/2023]
Abstract
CHARGE syndrome is a multiple congenital anomaly condition caused, in a majority of individuals, by loss of function pathogenic variants in the gene CHD7. In this special issue of the American Journal of Medical Genetics part C, authors of eleven manuscripts describe specific organ system features of CHARGE syndrome, with a focus on recent developments in diagnosis, etiologies, and treatments. Since 2004, when CHD7 was identified as the major causative gene in CHARGE, several animal models (mice, zebrafish, flies, and frog) and cell-based systems have been developed to explore the underlying pathophysiology of this condition. In this article, we summarize those advances, highlight opportunities for new discoveries, and encourage readers to explore specific organ systems in more detail in each individual article. We hope the excitement around innovative research and development in CHARGE syndrome will encourage others to join this effort, and will stimulate other investigators and professionals to engage with individuals diagnosed as having CHARGE syndrome, their families, and their care providers.
Collapse
Affiliation(s)
- Conny van Ravenswaaij-Arts
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Donna M Martin
- Departments of Human Genetics, The University of Michigan Medical School, Ann Arbor, Michigan.,Departments of Pediatrics, The University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
36
|
Corsten-Janssen N, Scambler PJ. Clinical and molecular effects of CHD7 in the heart. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2017; 175:487-495. [PMID: 29088513 DOI: 10.1002/ajmg.c.31590] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/28/2017] [Accepted: 10/01/2017] [Indexed: 12/28/2022]
Abstract
Heart defects caused by loss-of-function mutations in CHD7 are a frequent cause of morbidity and mortality in CHARGE syndrome. Here we review the clinical and molecular aspects of CHD7 that are related to the cardiovascular manifestations of the syndrome. The types of heart defects found in patients with CHD7 mutations are variable, with an overrepresentation of atrioventricular septal defect and outflow tract defect including aortic arch anomalies compared to nonsyndromic heart defects. Chd7 haploinsufficiency in mouse is a good model for studying the heart effects seen in CHARGE syndrome, and mouse models reveal a role for Chd7 in multiple lineages during heart development. Formation of the great vessels requires Chd7 expression in the pharyngeal surface ectoderm, and this expression likely has an non-autonomous effect on neural crest cells. In the cardiogenic mesoderm, Chd7 is required for atrioventricular cushion development and septation of the outflow tract. Emerging knowledge about the function of CHD7 in the heart indicates that it may act in concert with transcription factors such as TBX1 and SMADs to regulate genes such as p53 and the cardiac transcription factor NKX2.5.
Collapse
Affiliation(s)
- Nicole Corsten-Janssen
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter J Scambler
- UCL Great Ormond Street Institute of Child Health, Section Developmental Biology of Birth Defects, London, UK
| |
Collapse
|
37
|
Pauli S, Bajpai R, Borchers A. CHARGEd with neural crest defects. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2017; 175:478-486. [PMID: 29082625 DOI: 10.1002/ajmg.c.31584] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 08/23/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022]
Abstract
Neural crest cells are highly migratory pluripotent cells that give rise to diverse derivatives including cartilage, bone, smooth muscle, pigment, and endocrine cells as well as neurons and glia. Abnormalities in neural crest-derived tissues contribute to the etiology of CHARGE syndrome, a complex malformation disorder that encompasses clinical symptoms like coloboma, heart defects, atresia of the choanae, retarded growth and development, genital hypoplasia, ear anomalies, and deafness. Mutations in the chromodomain helicase DNA-binding protein 7 (CHD7) gene are causative of CHARGE syndrome and loss-of-function data in different model systems have firmly established a role of CHD7 in neural crest development. Here, we will summarize our current understanding of the function of CHD7 in neural crest development and discuss possible links of CHARGE syndrome to other developmental disorders.
Collapse
Affiliation(s)
- Silke Pauli
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Ruchi Bajpai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Annette Borchers
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
38
|
Feng W, Shao C, Liu HK. Versatile Roles of the Chromatin Remodeler CHD7 during Brain Development and Disease. Front Mol Neurosci 2017; 10:309. [PMID: 29033785 PMCID: PMC5625114 DOI: 10.3389/fnmol.2017.00309] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/14/2017] [Indexed: 11/13/2022] Open
Abstract
CHD7 (Chromo-Helicase-DNA binding protein 7) protein is an ATP-dependent chromatin remodeler. Heterozygous mutation of the CHD7 gene causes a severe congenital disease known as CHARGE syndrome. Most CHARGE syndrome patients have brain structural anomalies, implicating an important role of CHD7 during brain development. In this review, we summarize studies dissecting developmental functions of CHD7 in the brain and discuss pathogenic mechanisms behind neurodevelopmental defects caused by mutation of CHD7. As we discussed, CHD7 protein exhibits a remarkably specific and dynamic expression pattern in the brain. Studies in human and animal models have revealed that CHD7 is involved in multiple developmental lineages and processes in the brain. Mechanistically, CHD7 is essential for neural differentiation due to its transcriptional regulation in progenitor cells.
Collapse
Affiliation(s)
- Weijun Feng
- Division of Molecular Neurogenetics, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Chunxuan Shao
- Division of Molecular Neurogenetics, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Hai-Kun Liu
- Division of Molecular Neurogenetics, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
39
|
Robert C, Pasquier L, Cohen D, Fradin M, Canitano R, Damaj L, Odent S, Tordjman S. Role of Genetics in the Etiology of Autistic Spectrum Disorder: Towards a Hierarchical Diagnostic Strategy. Int J Mol Sci 2017; 18:E618. [PMID: 28287497 PMCID: PMC5372633 DOI: 10.3390/ijms18030618] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/17/2017] [Accepted: 02/20/2017] [Indexed: 12/27/2022] Open
Abstract
Progress in epidemiological, molecular and clinical genetics with the development of new techniques has improved knowledge on genetic syndromes associated with autism spectrum disorder (ASD). The objective of this article is to show the diversity of genetic disorders associated with ASD (based on an extensive review of single-gene disorders, copy number variants, and other chromosomal disorders), and consequently to propose a hierarchical diagnostic strategy with a stepwise evaluation, helping general practitioners/pediatricians and child psychiatrists to collaborate with geneticists and neuropediatricians, in order to search for genetic disorders associated with ASD. The first step is a clinical investigation involving: (i) a child psychiatric and psychological evaluation confirming autism diagnosis from different observational sources and assessing autism severity; (ii) a neuropediatric evaluation examining neurological symptoms and developmental milestones; and (iii) a genetic evaluation searching for dysmorphic features and malformations. The second step involves laboratory and if necessary neuroimaging and EEG studies oriented by clinical results based on clinical genetic and neuropediatric examinations. The identification of genetic disorders associated with ASD has practical implications for diagnostic strategies, early detection or prevention of co-morbidity, specific treatment and follow up, and genetic counseling.
Collapse
Affiliation(s)
- Cyrille Robert
- Pôle Hospitalo-Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (PHUPEA), University of Rennes 1 and Centre Hospitalier Guillaume Régnier, 35200 Rennes, France.
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - Laurent Pasquier
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - David Cohen
- Hospital-University Department of Child and Adolescent Psychiatry, Pitié-Salpétrière Hospital, Paris 6 University, 75013 Paris, France.
| | - Mélanie Fradin
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - Roberto Canitano
- Division of Child and Adolescent Neuropsychiatry, University Hospital of Siena, 53100 Siena, Italy.
| | - Léna Damaj
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - Sylvie Odent
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - Sylvie Tordjman
- Pôle Hospitalo-Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (PHUPEA), University of Rennes 1 and Centre Hospitalier Guillaume Régnier, 35200 Rennes, France.
- Laboratory of Psychology of Perception, University Paris Descartes, 75270 Paris, France.
| |
Collapse
|
40
|
Tordjman S, Cohen D, Coulon N, Anderson GM, Botbol M, Canitano R, Roubertoux PL. Reframing autism as a behavioral syndrome and not a specific mental disorder: Implications of genetic and phenotypic heterogeneity. Neurosci Biobehav Rev 2017; 80:210. [PMID: 28153685 DOI: 10.1016/j.neubiorev.2017.01.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/18/2016] [Accepted: 01/23/2017] [Indexed: 12/13/2022]
Abstract
Clinical and molecular genetics have advanced current knowledge on genetic disorders associated with autism. A review of diverse genetic disorders associated with autism is presented and for the first time discussed extensively with regard to possible common underlying mechanisms leading to a similar cognitive-behavioral phenotype of autism. The possible role of interactions between genetic and environmental factors, including epigenetic mechanisms, is in particular examined. Finally, the pertinence of distinguishing non-syndromic autism (isolated autism) from syndromic autism (autism associated with genetic disorders) will be reconsidered. Given the high genetic and etiological heterogeneity of autism, autism can be viewed as a behavioral syndrome related to known genetic disorders (syndromic autism) or currently unknown disorders (apparent non-syndromic autism), rather than a specific categorical mental disorder. It highlights the need to study autism phenotype and developmental trajectory through a multidimensional, non-categorical approach with multivariate analyses within autism spectrum disorder but also across mental disorders, and to conduct systematically clinical genetic examination searching for genetic disorders in all individuals (children but also adults) with autism.
Collapse
Affiliation(s)
- S Tordjman
- Pôle Hospitalo-Universitaire de Psychiatrie de l'Enfant et de l'Adolescent, Université de Rennes 1 and Centre Hospitalier Guillaume Régnier, 154 rue de Châtillon, 35200 Rennes, France; Laboratoire Psychologie de la Perception, Université Paris Descartes and CNRS UMR 8158, Paris, France.
| | - D Cohen
- Department of Child and Adolescent Psychiatry, AP-HP, GH Pitié-Salpétrière, CNRS FRE 2987, Université Pierre et Marie Curie, Paris, France
| | - N Coulon
- Laboratoire Psychologie de la Perception, Université Paris Descartes and CNRS UMR 8158, Paris, France
| | - G M Anderson
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - M Botbol
- Departement Hospitalo-Universitaire de Psychiatrie de l'Enfant et de l'Adolescent, Université de Bretagne Occidentale, Brest, France
| | - R Canitano
- Division of Child and Adolescent Neuropsychiatry, University Hospital of Siena, Siena, Italy
| | - P L Roubertoux
- Aix Marseille Université, GMGF, Inserm, UMR_S 910, 13385, Marseille, France
| |
Collapse
|
41
|
Grant MG, Patterson VL, Grimes DT, Burdine RD. Modeling Syndromic Congenital Heart Defects in Zebrafish. Curr Top Dev Biol 2017; 124:1-40. [DOI: 10.1016/bs.ctdb.2016.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Blanco-Sánchez B, Clément A, Phillips JB, Westerfield M. Zebrafish models of human eye and inner ear diseases. Methods Cell Biol 2016; 138:415-467. [PMID: 28129854 DOI: 10.1016/bs.mcb.2016.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Eye and inner ear diseases are the most common sensory impairments that greatly impact quality of life. Zebrafish have been intensively employed to understand the fundamental mechanisms underlying eye and inner ear development. The zebrafish visual and vestibulo-acoustic systems are very similar to these in humans, and although not yet mature, they are functional by 5days post-fertilization (dpf). In this chapter, we show how the zebrafish has significantly contributed to the field of biomedical research and how researchers, by establishing disease models and meticulously characterizing their phenotypes, have taken the first steps toward therapies. We review here models for (1) eye diseases, (2) ear diseases, and (3) syndromes affecting eye and/or ear. The use of new genome editing technologies and high-throughput screening systems should increase considerably the speed at which knowledge from zebrafish disease models is acquired, opening avenues for better diagnostics, treatments, and therapies.
Collapse
Affiliation(s)
| | - A Clément
- University of Oregon, Eugene, OR, United States
| | | | | |
Collapse
|
43
|
Mandalos NP, Remboutsika E. Sox2: To crest or not to crest? Semin Cell Dev Biol 2016; 63:43-49. [PMID: 27592260 DOI: 10.1016/j.semcdb.2016.08.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/29/2016] [Accepted: 08/30/2016] [Indexed: 12/12/2022]
Abstract
Precise control of neural progenitor transformation into neural crest stem cells ensures proper craniofacial and head development. In the neural progenitor pool, SoxB factors play an essential role as cell fate determinants of neural development, whereas during neural crest stem cell formation, Sox2 plays a predominant role as a guardian of the developmental clock that ensures precision of cell flow in the developing head.
Collapse
Affiliation(s)
- Nikolaos Panagiotis Mandalos
- National University of Athens Medical School, Department of Pediatrics, 75 Mikras Asias Str., 115 27, Athens, Greece; Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming", 34 Fleming Str., 16672 Vari-Attica, Greece; Adjunct Faculty, The Lieber Institute for Brain Development, Basic Sciences Division, Johns Hopkins Medical Campus, 855 North Wolfe Str., Suite 300, 3rd Floor, Baltimore, MD 21205, USA
| | - Eumorphia Remboutsika
- National University of Athens Medical School, Department of Pediatrics, 75 Mikras Asias Str., 115 27, Athens, Greece; Stem Cell Biology Laboratory, Biomedical Sciences Research Centre "Alexander Fleming", 34 Fleming Str., 16672 Vari-Attica, Greece; Adjunct Faculty, The Lieber Institute for Brain Development, Basic Sciences Division, Johns Hopkins Medical Campus, 855 North Wolfe Str., Suite 300, 3rd Floor, Baltimore, MD 21205, USA.
| |
Collapse
|
44
|
Asad Z, Pandey A, Babu A, Sun Y, Shevade K, Kapoor S, Ullah I, Ranjan S, Scaria V, Bajpai R, Sachidanandan C. Rescue of neural crest-derived phenotypes in a zebrafish CHARGE model by Sox10 downregulation. Hum Mol Genet 2016; 25:3539-3554. [PMID: 27418670 PMCID: PMC5179949 DOI: 10.1093/hmg/ddw198] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 05/27/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022] Open
Abstract
CHD7 mutations are implicated in a majority of cases of the congenital disorder, CHARGE syndrome. CHARGE, an autosomal dominant syndrome, is known to affect multiple tissues including eye, heart, ear, craniofacial nerves and skeleton and genital organs. Using a morpholino-antisense-oligonucleotide-based zebrafish model for CHARGE syndrome, we uncover a complex spectrum of abnormalities in the neural crest and the crest-derived cell types. We report for the first time, defects in myelinating Schwann cells, enteric neurons and pigment cells in a CHARGE model. We also observe defects in the specification of peripheral neurons and the craniofacial skeleton as previously reported. Chd7 morphants have impaired migration of neural crest cells and deregulation of sox10 expression from the early stages. Knocking down Sox10 in the zebrafish CHARGE model rescued the defects in Schwann cells and craniofacial cartilage. Our zebrafish CHARGE model thus reveals important regulatory roles for Chd7 at multiple points of neural crest development viz., migration, fate choice and differentiation and we suggest that sox10 deregulation is an important driver of the neural crest-derived aspects of Chd7 dependent CHARGE syndrome.
Collapse
Affiliation(s)
- Zainab Asad
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Aditi Pandey
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Aswini Babu
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Yuhan Sun
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kaivalya Shevade
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shruti Kapoor
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Ikram Ullah
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Shashi Ranjan
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
| | - Vinod Scaria
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| | - Ruchi Bajpai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry and Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chetana Sachidanandan
- CSIR-Institute of Genomics & Integrative Biology, South Campus, New Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India and
| |
Collapse
|
45
|
Raghunathan R, Singh M, Dickinson ME, Larin KV. Optical coherence tomography for embryonic imaging: a review. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:50902. [PMID: 27228503 PMCID: PMC4881290 DOI: 10.1117/1.jbo.21.5.050902] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/25/2016] [Indexed: 05/18/2023]
Abstract
Embryogenesis is a highly complex and dynamic process, and its visualization is crucial for understanding basic physiological processes during development and for identifying and assessing possible defects, malformations, and diseases. While traditional imaging modalities, such as ultrasound biomicroscopy, micro-magnetic resonance imaging, and micro-computed tomography, have long been adapted for embryonic imaging, these techniques generally have limitations in their speed, spatial resolution, and contrast to capture processes such as cardiodynamics during embryogenesis. Optical coherence tomography (OCT) is a noninvasive imaging modality with micrometer-scale spatial resolution and imaging depth up to a few millimeters in tissue. OCT has bridged the gap between ultrahigh resolution imaging techniques with limited imaging depth like confocal microscopy and modalities, such as ultrasound sonography, which have deeper penetration but poorer spatial resolution. Moreover, the noninvasive nature of OCT has enabled live imaging of embryos without any external contrast agents. We review how OCT has been utilized to study developing embryos and also discuss advances in techniques used in conjunction with OCT to understand embryonic development.
Collapse
Affiliation(s)
- Raksha Raghunathan
- University of Houston, Department of Biomedical Engineering, 3517 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
| | - Manmohan Singh
- University of Houston, Department of Biomedical Engineering, 3517 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
| | - Mary E. Dickinson
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, One Baylor Plaza- BCM335, Houston, Texas 77030, United States
| | - Kirill V. Larin
- University of Houston, Department of Biomedical Engineering, 3517 Cullen Boulevard, Room 2027, Houston, Texas 77204-5060, United States
- Baylor College of Medicine, Department of Molecular Physiology and Biophysics, One Baylor Plaza- BCM335, Houston, Texas 77030, United States
| |
Collapse
|
46
|
Prominent scapulae mimicking an inherited myopathy expands the phenotype of CHD7-related disease. Eur J Hum Genet 2016; 24:1216-9. [PMID: 26813943 DOI: 10.1038/ejhg.2015.276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/19/2015] [Accepted: 12/08/2015] [Indexed: 11/09/2022] Open
Abstract
CHD7 variants are a well-established cause of CHARGE syndrome, a disabling multi-system malformation disorder that is often associated with deafness, visual impairment and intellectual disability. Less severe forms of CHD7-related disease are known to exist, but the full spectrum of phenotypes remains uncertain. We identified a de novo missense variant in CHD7 in a family presenting with musculoskeletal abnormalities as the main manifestation of CHD7-related disease, representing a new phenotype. The proband presented with prominent scapulae, mild shoulder girdle weakness and only subtle dysmorphic features. Investigation revealed hypoplasia of the trapezius and sternocleidomastoid muscles and semicircular canal defects, but he did not fulfill diagnostic criteria for CHARGE syndrome. Although the shoulders are often sloping and anteverted in CHARGE syndrome, the underlying neuromuscular cause has never been investigated. This report expands the phenotypes associated with CHD7 mutations to include a musculoskeletal presentation, with hypoplasia of the shoulder and neck muscles. CHD7 should be considered in patients presenting in childhood with stable scapular winging, particularly if accompanied by dysmorphic features and balance difficulties.
Collapse
|
47
|
Abstract
The formation of the face and skull involves a complex series of developmental events mediated by cells derived from the neural crest, endoderm, mesoderm, and ectoderm. Although vertebrates boast an enormous diversity of adult facial morphologies, the fundamental signaling pathways and cellular events that sculpt the nascent craniofacial skeleton in the embryo have proven to be highly conserved from fish to man. The zebrafish Danio rerio, a small freshwater cyprinid fish from eastern India, has served as a popular model of craniofacial development since the 1990s. Unique strengths of the zebrafish model include a simplified skeleton during larval stages, access to rapidly developing embryos for live imaging, and amenability to transgenesis and complex genetics. In this chapter, we describe the anatomy of the zebrafish craniofacial skeleton; its applications as models for the mammalian jaw, middle ear, palate, and cranial sutures; the superior imaging technology available in fish that has provided unprecedented insights into the dynamics of facial morphogenesis; the use of the zebrafish to decipher the genetic underpinnings of craniofacial biology; and finally a glimpse into the most promising future applications of zebrafish craniofacial research.
Collapse
|
48
|
Cheng J, Ma D, Wu Y, Luo C, Huang C, Hu P, Zhang J, Jiang T, Xu Z. Identification of one novel CHD7 mutation in a patient from China with atypical CHARGE syndrome. Gene 2015; 571:298-302. [PMID: 26187070 DOI: 10.1016/j.gene.2015.07.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/25/2015] [Accepted: 07/12/2015] [Indexed: 12/20/2022]
Abstract
CHARGE syndrome is an autosomal-dominant disorder involved in multiple organs. Loss-of-function mutations in CHD7, a member of the chromodomain helicase DNA-binding (CHD) protein family, are known to cause the CHARGE syndrome. The purposes of this paper were to affirm the diagnosis and to identify the molecular basis of one atypical CHARGE syndrome patient from China, where only one CHARGE case was reported before. We employed the Verloes criteria to make a preliminary clinical diagnosis, and performed mutation screening of CHD7 via Ion Torrent semiconductor sequencing. The patient was preliminary diagnosed as atypical CHARGE syndrome according to Verloes criteria with a novel heterozygous small deletion of CHD7 (CHD7: c.3462_3471delTCGCTTCCCT). As the second reported case of CHARGE syndrome in China, it was caused by one novel heterozygous mutation of the CHD7 gene. Our findings further reveal the relationship between CHD7 and CHARGE syndrome and provide a potential clinical diagnosis for CHARGE syndrome.
Collapse
Affiliation(s)
- Jian Cheng
- State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Dingyuan Ma
- State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yun Wu
- Department of Ultrasound Diagnosis, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Chunyu Luo
- State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Chengyi Huang
- Department of Interventional Radiology, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Ping Hu
- State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Jingjing Zhang
- State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Tao Jiang
- State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Zhengfeng Xu
- State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China.
| |
Collapse
|
49
|
Abstract
Epigenetic events including chromatin remodeling and histone modifications have recently emerged as important contributors to a variety of neurodevelopmental disorders. This review focuses on CHARGE syndrome, a multiple anomaly condition caused by mutations in the gene encoding CHD7, an ATP-dependent chromatin remodeling protein. CHD7 exhibits pleiotropic effects during embryonic development, consistent with highly variable clinical features in CHARGE syndrome. In this review, a historical description of CHARGE is provided, followed by establishment of diagnostic criteria, gene discovery, and development of animal models. Current understanding of epigenetic CHD7 functions and interacting proteins in cells and tissues is also presented, and final emphasis is placed on challenges and major questions to be answered with ongoing research efforts.
Collapse
Affiliation(s)
- Donna M Martin
- Department of Human Genetics at The University of Michigan Medical School, Ann Arbor, MI, 48109 ; Department of Pediatrics and Communicable Diseases at The University of Michigan Medical School, Ann Arbor, MI, 48109
| |
Collapse
|
50
|
Functionally compromised CHD7 alleles in patients with isolated GnRH deficiency. Proc Natl Acad Sci U S A 2014; 111:17953-8. [PMID: 25472840 DOI: 10.1073/pnas.1417438111] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inactivating mutations in chromodomain helicase DNA binding protein 7 (CHD7) cause CHARGE syndrome, a severe multiorgan system disorder of which Isolated gonadotropin-releasing hormone (GnRH) deficiency (IGD) is a minor feature. Recent reports have described predominantly missense CHD7 alleles in IGD patients, but it is unclear if these alleles are relevant to causality or overall genetic burden of Kallmann syndrome (KS) and normosmic form of IGD. To address this question, we sequenced CHD7 in 783 well-phenotyped IGD patients lacking full CHARGE features; we identified nonsynonymous rare sequence variants in 5.2% of the IGD cohort (73% missense and 27% splice variants). Functional analyses in zebrafish using a surrogate otolith assay of a representative set of these CHD7 alleles showed that rare sequence variants observed in controls showed no altered function. In contrast, 75% of the IGD-associated alleles were deleterious and resulted in both KS and normosmic IGD. In two families, pathogenic mutations in CHD7 coexisted with mutations in other known IGD genes. Taken together, our data suggest that rare deleterious CHD7 alleles contribute to the mutational burden of patients with both KS and normosmic forms of IGD in the absence of full CHARGE syndrome. These findings (i) implicate a unique role or preferential sensitivity for CHD7 in the ontogeny of GnRH neurons, (ii) reiterate the emerging genetic complexity of this family of IGD disorders, and (iii) demonstrate how the coordinated use of well-phenotyped cohorts, families, and functional studies can inform genetic architecture and provide insights into the developmental biology of cellular systems.
Collapse
|