1
|
Goyal S, Tibrewal S, Ratna R, Vanita V. Genetic and environmental factors contributing to anophthalmia and microphthalmia: Current understanding and future directions. World J Clin Pediatr 2025; 14:101982. [DOI: 10.5409/wjcp.v14.i2.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/18/2025] Open
Abstract
Anophthalmia is defined as a complete absence of one eye or both the eyes, while microphthalmia represents the presence of a small eye within the orbit. The estimated birth prevalence for anophthalmia is approximately 3 per 100000 live births, and for microphthalmia, it is around 14 per 100000 live births. However, combined evidence suggests that the prevalence of these malformations could be as high as 30 per 100000 individuals. Microphthalmia is reported to occur in 3.2% to 11.2% of blind children. Anophthalmia and microphthalmia (A/M) are part of a phenotypic spectrum alongside ocular coloboma, hypothesized to share a common genetic basis. Both A/M can occur in isolation or as part of a syndrome. Their complex etiology involves chromosomal aberrations, monogenic inheritance pattern, and the contribution of environmental factors such as gestational-acquired infections, maternal vitamin A deficiency (VAD), exposure to X-rays, solvent misuse, and thalidomide exposure. A/M exhibit significant clinical and genetic heterogeneity with over 90 genes identified so far. Familial cases of A/M have a complex genetic basis, including all Mendelian modes of inheritance, i.e., autosomal dominant, recessive, and X-linked. Most cases arise sporadically due to de novo mutations. Examining gene expression during eye development and the effects of various environmental variables will help us better understand the phenotypic heterogeneity found in A/M, leading to more effective diagnosis and management strategies. The present review focuses on key genetic factors, developmental abnormalities, and environmental modifiers linked with A/M. It also emphasizes at potential research areas including multiomic methods and disease modeling with induced pluripotent stem cell technologies, which aim to create innovative treatment options.
Collapse
Affiliation(s)
- Shiwali Goyal
- Department of Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Rockville, MD 20852, United States
| | - Shailja Tibrewal
- Department of Pediatric Ophthalmology, Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
- Department of Ocular Genetics (Center for Unknown and Rare Eye Diseases), Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
| | - Ria Ratna
- Department of Ocular Genetics (Center for Unknown and Rare Eye Diseases), Dr. Shroff’s Charity Eye Hospital, New Delhi 110002, Delhi, India
| | - Vanita Vanita
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| |
Collapse
|
2
|
Kim H, Heo HH, Um SJ. Loss of Asxl1 disrupts telencephalic midline integrity through dysregulation of SIX3 target genes. Biochem Biophys Res Commun 2025; 770:152039. [PMID: 40393104 DOI: 10.1016/j.bbrc.2025.152039] [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: 05/13/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025]
Abstract
Mutations in the epigenetic regulator Additional sex combs like 1 (Asxl1) have been implicated in neurodevelopmental syndromes; however, its role in embryonic brain development remains poorly understood. Here, we report that Asxl1 knockout mice exhibit severe telencephalic midline defects, including agenesis of the corpus callosum, absence of the septum, and formation of a single cerebral ventricle. These phenotypes closely resemble those of Six3-deficient brains, suggesting a functional link between Asxl1 and Six3. Co-immunoprecipitation and domain mapping revealed that ASXL1 directly interacts with SIX3 via its N-terminal domain (residues 371-655). Integrated analysis of RNA-seq and CUT&RUN datasets identified 806 direct Six3 target genes, among which 66 showed concordant expression changes in Asxl1-deficient neural stem cells. Gene ontology analysis revealed enrichment in pathways related to epigenetic regulation and forebrain development. Further motif and peak enrichment analyses identified eight forebrain-associated genes-Cacna1g, Col22a1, Cox6a2, Csmd3, Dock5, Palmd, Slc2a10, and Vit-that were significantly upregulated in the absence of Asxl1, as confirmed by RT-qPCR. These results indicate that ASXL1 cooperates with SIX3 to regulate a shared set of neurodevelopmental genes, thereby maintaining telencephalic midline integrity. Our findings provide new insights into the molecular basis of holoprosencephaly and related congenital brain malformations.
Collapse
Affiliation(s)
- Hyeju Kim
- Department of Integrative Bioscience and Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, South Korea
| | - Hyeon Ho Heo
- Department of Integrative Bioscience and Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, South Korea
| | - Soo-Jong Um
- Department of Integrative Bioscience and Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, South Korea.
| |
Collapse
|
3
|
Lan YZ, Wu Z, Chen WJ, Yu XN, Wu HT, Liu J. Sine oculis homeobox homolog family function in gastrointestinal cancer: Progression and comprehensive analysis. World J Clin Oncol 2025; 16:97163. [PMID: 39867730 PMCID: PMC11528897 DOI: 10.5306/wjco.v16.i1.97163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/20/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024] Open
Abstract
The sine oculis homeobox homolog (SIX) family, a group of transcription factors characterized by a conserved DNA-binding homology domain, plays a critical role in orchestrating embryonic development and organogenesis across various organisms, including humans. Comprising six distinct members, from SIX1 to SIX6, each member contributes uniquely to the development and differentiation of diverse tissues and organs, underscoring the versatility of the SIX family. Dysregulation or mutations in SIX genes have been implicated in a spectrum of developmental disorders, as well as in tumor initiation and progression, highlighting their pivotal role in maintaining normal developmental trajectories and cellular functions. Efforts to target the transcriptional complex of the SIX gene family have emerged as a promising strategy to inhibit tumor development. While the development of inhibitors targeting this gene family is still in its early stages, the significant potential of such interventions holds promise for future therapeutic advances. Therefore, this review aimed to comprehensively explore the advancements in understanding the SIX family within gastrointestinal cancers, focusing on its critical role in normal organ development and its implications in gastrointestinal cancers, including gastric, pancreatic, colorectal cancer, and hepatocellular carcinomas. In conclusion, this review deepened the understanding of the functional roles of the SIX family and explored the potential of utilizing this gene family for the diagnosis, prognosis, and treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Yang-Zheng Lan
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Zheng Wu
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Wen-Jia Chen
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Xin-Ning Yu
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Hua-Tao Wu
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jing Liu
- Department of The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
4
|
Shrestha P, Jaganathan A, Huilgol D, Ballon C, Hwangbo Y, Mills AA. Chd5 Regulates the Transcription Factor Six3 to Promote Neuronal Differentiation. Stem Cells 2023; 41:242-251. [PMID: 36636025 PMCID: PMC10020979 DOI: 10.1093/stmcls/sxad002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/16/2022] [Indexed: 01/14/2023]
Abstract
Chromodomain helicase DNA-binding protein 5 (Chd5) is an ATP-dependent chromatin remodeler that promotes neuronal differentiation. However, the mechanism behind the action of Chd5 during neurogenesis is not clearly understood. Here we use transcriptional profiling of cells obtained from Chd5 deficient mice at early and late stages of neuronal differentiation to show that Chd5 regulates neurogenesis by directing stepwise transcriptional changes. During early stages of neurogenesis, Chd5 promotes expression of the proneural transcription factor Six3 to repress Wnt5a, a non-canonical Wnt ligand essential for the maturation of neurons. This previously unappreciated ability of Chd5 to transcriptionally repress neuronal maturation factors is critical for both lineage specification and maturation. Thus, Chd5 facilitates early transcriptional changes in neural stem cells, thereby initiating transcriptional programs essential for neuronal fate specification.
Collapse
Affiliation(s)
- Padmina Shrestha
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Department of Molecular and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | | | - Dhananjay Huilgol
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Carlos Ballon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Yon Hwangbo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Alea A Mills
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| |
Collapse
|
5
|
Barratt KS, Drover KA, Thomas ZM, Arkell RM. Patterning of the antero-ventral mammalian brain: Lessons from holoprosencephaly comparative biology in man and mouse. WIREs Mech Dis 2022; 14:e1552. [PMID: 35137563 DOI: 10.1002/wsbm.1552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/30/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Adult form and function are dependent upon the activity of specialized signaling centers that act early in development at the embryonic midline. These centers instruct the surrounding cells to adopt a positional fate and to form the patterned structures of the phylotypic embryo. Abnormalities in these processes have devastating consequences for the individual, as exemplified by holoprosencephaly in which anterior midline development fails, leading to structural defects of the brain and/or face. In the 25 years since the first association between human holoprosencephaly and the sonic hedgehog gene, a combination of human and animal genetic studies have enhanced our understanding of the genetic and embryonic causation of this congenital defect. Comparative biology has extended the holoprosencephaly network via the inclusion of gene mutations from multiple signaling pathways known to be required for anterior midline formation. It has also clarified aspects of holoprosencephaly causation, showing that it arises when a deleterious variant is present within a permissive genome, and that environmental factors, as well as embryonic stochasticity, influence the phenotypic outcome of the variant. More than two decades of research can now be distilled into a framework of embryonic and genetic causation. This framework means we are poised to move beyond our current understanding of variants in signaling pathway molecules. The challenges now at the forefront of holoprosencephaly research include deciphering how the mutation of genes involved in basic cell processes can also cause holoprosencephaly, determining the important constituents of the holoprosencephaly permissive genome, and identifying environmental compounds that promote holoprosencephaly. This article is categorized under: Congenital Diseases > Stem Cells and Development Congenital Diseases > Genetics/Genomics/Epigenetics Congenital Diseases > Molecular and Cellular Physiology Congenital Diseases > Environmental Factors.
Collapse
Affiliation(s)
- Kristen S Barratt
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Kyle A Drover
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Zoe M Thomas
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Ruth M Arkell
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
6
|
Meurer L, Ferdman L, Belcher B, Camarata T. The SIX Family of Transcription Factors: Common Themes Integrating Developmental and Cancer Biology. Front Cell Dev Biol 2021; 9:707854. [PMID: 34490256 PMCID: PMC8417317 DOI: 10.3389/fcell.2021.707854] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 01/19/2023] Open
Abstract
The sine oculis (SIX) family of transcription factors are key regulators of developmental processes during embryogenesis. Members of this family control gene expression to promote self-renewal of progenitor cell populations and govern mechanisms of cell differentiation. When the function of SIX genes becomes disrupted, distinct congenital defects develops both in animal models and humans. In addition to the embryonic setting, members of the SIX family have been found to be critical regulators of tumorigenesis, promoting cell proliferation, epithelial-to-mesenchymal transition, and metastasis. Research in both the fields of developmental biology and cancer research have provided an extensive understanding of SIX family transcription factor functions. Here we review recent progress in elucidating the role of SIX family genes in congenital disease as well as in the promotion of cancer. Common themes arise when comparing SIX transcription factor function during embryonic and cancer development. We highlight the complementary nature of these two fields and how knowledge in one area can open new aspects of experimentation in the other.
Collapse
Affiliation(s)
- Logan Meurer
- Department of Basic Sciences, NYIT College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR, United States
| | - Leonard Ferdman
- Department of Basic Sciences, NYIT College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR, United States
| | - Beau Belcher
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR, United States
| | - Troy Camarata
- Department of Basic Sciences, NYIT College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR, United States
| |
Collapse
|
7
|
de Castro VF, Mattos D, de Carvalho FM, Cavalcanti DP, Duenas-Roque MM, Llerena J, Cosentino VR, Honjo RS, Leite JCL, Sanseverino MT, de Souza MPA, Bernardi P, Bolognese AM, Santana da Silva LC, Barbero P, Correia PS, Bueno LSM, Savastano CP, Orioli IM. New SHH and Known SIX3 Variants in a Series of Latin American Patients with Holoprosencephaly. Mol Syndromol 2021; 12:219-233. [PMID: 34421500 DOI: 10.1159/000515044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 02/04/2021] [Indexed: 11/19/2022] Open
Abstract
Holoprosencephaly (HPE) is the failure of the embryonic forebrain to develop into 2 hemispheres promoting midline cerebral and facial defects. The wide phenotypic variability and causal heterogeneity make genetic counseling difficult. Heterozygous variants with incomplete penetrance and variable expressivity in the SHH, SIX3, ZIC2, and TGIF1 genes explain ∼25% of the known causes of nonchromosomal HPE. We studied these 4 genes and clinically described 27 Latin American families presenting with nonchromosomal HPE. Three new SHH variants and a third known SIX3 likely pathogenic variant found by Sanger sequencing explained 15% of our cases. Genotype-phenotype correlation in these 4 families and published families with identical or similar driver gene, mutated domain, conservation of residue in other species, and the type of variant explain the pathogenicity but not the phenotypic variability. Nine patients, including 2 with SHH pathogenic variants, presented benign variants of the SHH, SIX3, ZIC2, and TGIF1 genes with potential alteration of splicing, a causal proposition in need of further studies. Finding more families with the same SIX3 variant may allow further identification of genetic or environmental modifiers explaining its variable phenotypic expression.
Collapse
Affiliation(s)
- Viviane Freitas de Castro
- ECLAMC at Departamento de Genética, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Genética Médica Populacional INAGEMP, Porto Alegre, Brazil
| | - Daniel Mattos
- ECLAMC at Departamento de Genética, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Genética Médica Populacional INAGEMP, Porto Alegre, Brazil
| | - Flavia Martinez de Carvalho
- Instituto Nacional de Genética Médica Populacional INAGEMP, Porto Alegre, Brazil.,ECLAMC at Laboratorio Epidemiol. Malformações Congênitas, IOC/FIOCRUZ, Rio de Janeiro, Brazil
| | | | - Milagros M Duenas-Roque
- ECLAMC at Servicio de Genética, Hospital Nacional Edgardo Rebagliati Martins/EsSalud, Lima, Peru
| | - Juan Llerena
- Instituto Nacional de Genética Médica Populacional INAGEMP, Porto Alegre, Brazil.,ECLAMC at Centro de Genética Médica, IFF/FIOCRUZ, Rio de Janeiro, Brazil
| | | | | | | | | | | | - Pricila Bernardi
- Núcleo de Genética Clínica, Departamento de Clínica Médica/UFSC, Florianópolis, Brazil
| | - Ana Maria Bolognese
- Departamento de Ortodontia, Faculdade de Odontologia/UFRJ, Rio de Janeiro, Brazil
| | - Luiz Carlos Santana da Silva
- Instituto Nacional de Genética Médica Populacional INAGEMP, Porto Alegre, Brazil.,Laboratório de Erros Inatos de Metabolismo, Instituto de Ciências Biológicas/UFP, Belém, Brazil
| | - Pablo Barbero
- RENAC, Centro Nacional de Genética Médica Dr. Eduardo E. Castilla/MS, Buenos Aires, Argentina
| | | | | | | | - Iêda Maria Orioli
- ECLAMC at Departamento de Genética, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Genética Médica Populacional INAGEMP, Porto Alegre, Brazil
| |
Collapse
|
8
|
Song X, Chen H, Shang Z, Du H, Li Z, Wen Y, Liu G, Qi D, You Y, Yang Z, Zhang Z, Xu Z. Homeobox Gene Six3 is Required for the Differentiation of D2-Type Medium Spiny Neurons. Neurosci Bull 2021; 37:985-998. [PMID: 34014554 PMCID: PMC8275777 DOI: 10.1007/s12264-021-00698-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 01/09/2021] [Indexed: 12/31/2022] Open
Abstract
Medium spiny neurons (MSNs) in the striatum, which can be divided into D1 and D2 MSNs, originate from the lateral ganglionic eminence (LGE). Previously, we reported that Six3 is a downstream target of Sp8/Sp9 in the transcriptional regulatory cascade of D2 MSN development and that conditionally knocking out Six3 leads to a severe loss of D2 MSNs. Here, we showed that Six3 mainly functions in D2 MSN precursor cells and gradually loses its function as D2 MSNs mature. Conditional deletion of Six3 had little effect on cell proliferation but blocked the differentiation of D2 MSN precursor cells. In addition, conditional overexpression of Six3 promoted the differentiation of precursor cells in the LGE. We measured an increase of apoptosis in the postnatal striatum of conditional Six3-knockout mice. This suggests that, in the absence of Six3, abnormally differentiated D2 MSNs are eliminated by programmed cell death. These results further identify Six3 as an important regulatory element during D2 MSN differentiation.
Collapse
Affiliation(s)
- Xiaolei Song
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Haotian Chen
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zicong Shang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Heng Du
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhenmeiyu Li
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yan Wen
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Guoping Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Dashi Qi
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yan You
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhengang Yang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhuangzhi Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Zhejun Xu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Diaz C, Puelles L. Developmental Genes and Malformations in the Hypothalamus. Front Neuroanat 2020; 14:607111. [PMID: 33324176 PMCID: PMC7726113 DOI: 10.3389/fnana.2020.607111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
The hypothalamus is a heterogeneous rostral forebrain region that regulates physiological processes essential for survival, energy metabolism, and reproduction, mainly mediated by the pituitary gland. In the updated prosomeric model, the hypothalamus represents the rostralmost forebrain, composed of two segmental regions (terminal and peduncular hypothalamus), which extend respectively into the non-evaginated preoptic telencephalon and the evaginated pallio-subpallial telencephalon. Complex genetic cascades of transcription factors and signaling molecules rule their development. Alterations of some of these molecular mechanisms acting during forebrain development are associated with more or less severe hypothalamic and pituitary dysfunctions, which may be associated with brain malformations such as holoprosencephaly or septo-optic dysplasia. Studies on transgenic mice with mutated genes encoding critical transcription factors implicated in hypothalamic-pituitary development are contributing to understanding the high clinical complexity of these pathologies. In this review article, we will analyze first the complex molecular genoarchitecture of the hypothalamus resulting from the activity of previous morphogenetic signaling centers and secondly some malformations related to alterations in genes implicated in the development of the hypothalamus.
Collapse
Affiliation(s)
- Carmen Diaz
- Department of Medical Sciences, School of Medicine and Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, Albacete, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| |
Collapse
|
10
|
Yu Z, Feng J, Wang W, Deng Z, Zhang Y, Xiao L, Wang Z, Liu C, Liu Q, Chen S, Wu M. The EGFR-ZNF263 signaling axis silences SIX3 in glioblastoma epigenetically. Oncogene 2020; 39:3163-3178. [PMID: 32051553 PMCID: PMC7142014 DOI: 10.1038/s41388-020-1206-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/28/2020] [Accepted: 02/03/2020] [Indexed: 12/15/2022]
Abstract
The homeotic protein SIX3 is a transcription factor vital for neurogenesis and has a bivalent promoter. We previously showed that SIX3 can be transcriptionally silenced by DNA hypermethylation, functions as a tumor suppressor gene, and inhibits human glioblastoma transcriptionally. Here, we show that the activation of epidermal growth factor (EGFR) induces DNA methylation of SIX3 promoter through the MAPK pathway. ERK, when activated, binds with ZNF263, consequently abrogating the ubiquitination of ZNF263 and leading to its stabilization. ZNF263 binds to the core promoter region of SIX3 and recruits the KAP1/HATS/DNMT corepressor complex to induce transcriptional silencing of SIX3 through H3K27me3 and methylation of SIX3 promoter. Activation of the EGFR-ZNF263 signaling axis in phenotypically normal astrocytes or glioblastoma cells triggers or enhances tumorigenic activities, while elevated expression of the EGFR-ZNF263 signaling components in glioblastoma tissues is associated with poor prognosis of the patients. Together, our findings demonstrate that epigenetic silencing of SIX3 is controlled by a sophisticated and highly ordered oncogenic signaling pathway and therefore provide new insights into initiation and progression of glioblastoma.
Collapse
Affiliation(s)
- Zhibin Yu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
- Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jianbo Feng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Wei Wang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, China
| | - Zhiyong Deng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Yan Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Lan Xiao
- Cancer Research Institute, School of Basic Medical Science, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Zeyou Wang
- Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Changhong Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- Cancer Research Institute, School of Basic Medical Science, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China
| | - Qing Liu
- Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shuai Chen
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.
- Cancer Research Institute, School of Basic Medical Science, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, Ministry of Health, Changsha, 410078, Hunan, China.
| |
Collapse
|
11
|
Sagai T, Amano T, Maeno A, Ajima R, Shiroishi T. SHH signaling mediated by a prechordal and brain enhancer controls forebrain organization. Proc Natl Acad Sci U S A 2019; 116:23636-23642. [PMID: 31685615 PMCID: PMC6876251 DOI: 10.1073/pnas.1901732116] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Sonic hedgehog (SHH) signaling plays a pivotal role in 2 different phases during brain development. Early SHH signaling derived from the prechordal plate (PrCP) triggers secondary Shh induction in the forebrain, which overlies the PrCP, and the induced SHH signaling, in turn, directs late neuronal differentiation of the forebrain. Consequently, Shh regulation in the PrCP is crucial for initiation of forebrain development. However, no enhancer that regulates prechordal Shh expression has yet been found. Here, we identified a prechordal enhancer, named SBE7, in the vicinity of a cluster of known forebrain enhancers for Shh This enhancer also directs Shh expression in the ventral midline of the forebrain, which receives the prechordal SHH signal. Thus, the identified enhancer acts not only for the initiation of Shh regulation in the PrCP but also for subsequent Shh induction in the forebrain. Indeed, removal of the enhancer from the mouse genome markedly down-regulated the expression of Shh in the rostral domains of the axial mesoderm and in the ventral midline of the forebrain and hypothalamus in the mouse embryo, and caused a craniofacial abnormality similar to human holoprosencephaly (HPE). These findings demonstrate that SHH signaling mediated by the newly identified enhancer is essential for development and growth of the ventral midline of the forebrain and hypothalamus. Understanding of the Shh regulation governed by this prechordal and brain enhancer provides an insight into the mechanism underlying craniofacial morphogenesis and the etiology of HPE.
Collapse
Affiliation(s)
- Tomoko Sagai
- Mammalian Genetics Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima 411-8540, Japan
- Information Resource Research Center, Association for Propagation of the Knowledge of Genetics, Mishima 411-8540, Japan
| | - Takanori Amano
- Mammalian Genetics Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima 411-8540, Japan
- Next Generation Human Disease Model Team, RIKEN BioResource Research Center, Tsukuba 305-0074, Japan
- Department of Genetics, SOKENDAI, Mishima 411-8540, Japan
| | - Akiteru Maeno
- Mammalian Genetics Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima 411-8540, Japan
- Plant Cytogenetics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Rieko Ajima
- Department of Genetics, SOKENDAI, Mishima 411-8540, Japan
- Mammalian Development Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima 411-8540, Japan
- Mouse Research Supporting Unit, National Institute of Genetics, Mishima 411-8540, Japan
| | - Toshihiko Shiroishi
- Mammalian Genetics Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima 411-8540, Japan;
- Department of Genetics, SOKENDAI, Mishima 411-8540, Japan
- RIKEN BioResource Research Center, Tsukuba 305-0074, Japan
| |
Collapse
|
12
|
The Homeodomain Transcription Factors Vax1 and Six6 Are Required for SCN Development and Function. Mol Neurobiol 2019; 57:1217-1232. [PMID: 31705443 DOI: 10.1007/s12035-019-01781-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
The brain's primary circadian pacemaker, the suprachiasmatic nucleus (SCN), is required to translate day-length and circadian rhythms into neuronal, hormonal, and behavioral rhythms. Here, we identify the homeodomain transcription factor ventral anterior homeobox 1 (Vax1) as required for SCN development, vasoactive intestinal peptide expression, and SCN output. Previous work has shown that VAX1 is required for gonadotropin-releasing hormone (GnRH/LHRH) neuron development, a neuronal population controlling reproductive status. Surprisingly, the ectopic expression of a Gnrh-Cre allele (Gnrhcre) in the SCN confirmed the requirement of both VAX1 (Vax1flox/flox:Gnrhcre, Vax1Gnrh-cre) and sine oculis homeobox protein 6 (Six6flox/flox:Gnrhcre, Six6Gnrh-cre) in SCN function in adulthood. To dissociate the role of Vax1 and Six6 in GnRH neuron and SCN function, we used another Gnrh-cre allele that targets GnRH neurons, but not the SCN (Lhrhcre). Both Six6Lhrh-cre and Vax1Lhrh-cre were infertile, and in contrast to Vax1Gnrh-cre and Six6Gnrh-cre mice, Six6Lhrh-cre and Vax1Lhrh-cre had normal circadian behavior. Unexpectedly, ~ 1/4 of the Six6Gnrh-cre mice were unable to entrain to light, showing that ectopic expression of Gnrhcre impaired function of the retino-hypothalamic tract that relays light information to the brain. This study identifies VAX1, and confirms SIX6, as transcription factors required for SCN development and function and demonstrates the importance of understanding how ectopic CRE expression can impact the results.
Collapse
|
13
|
Grinblat Y, Lipinski RJ. A forebrain undivided: Unleashing model organisms to solve the mysteries of holoprosencephaly. Dev Dyn 2019; 248:626-633. [PMID: 30993762 DOI: 10.1002/dvdy.41] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/10/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022] Open
Abstract
Evolutionary conservation and experimental tractability have made animal model systems invaluable tools in our quest to understand human embryogenesis, both normal and abnormal. Standard genetic approaches, particularly useful in understanding monogenic diseases, are no longer sufficient as research attention shifts toward multifactorial outcomes. Here, we examine this progression through the lens of holoprosencephaly (HPE), a common human malformation involving incomplete forebrain division, and a classic example of an etiologically complex outcome. We relate the basic underpinning of HPE pathogenesis to critical cell-cell interactions and signaling molecules discovered through embryological and genetic approaches in multiple model organisms, and discuss the role of the mouse model in functional examination of HPE-linked genes. We then outline the most critical remaining gaps to understanding human HPE, including the conundrum of incomplete penetrance/expressivity and the role of gene-environment interactions. To tackle these challenges, we outline a strategy that leverages new and emerging technologies in multiple model systems to solve the puzzle of HPE.
Collapse
Affiliation(s)
- Yevgenya Grinblat
- Department of Integrative Biology, University of Wisconsin, Madison, Wisconsin.,Department of Neuroscience, University of Wisconsin, Madison, Wisconsin.,McPherson Eye Research Institute, University of Wisconsin, Madison, Wisconsin
| | - Robert J Lipinski
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin.,Molecular and Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
14
|
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: 118] [Impact Index Per Article: 16.9] [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
|
15
|
Roessler E, Hu P, Muenke M. Holoprosencephaly in the genomics era. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2018; 178:165-174. [PMID: 29770992 DOI: 10.1002/ajmg.c.31615] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 01/08/2023]
Abstract
Holoprosencephaly (HPE) is the direct consequence of specific genetic and/or environmental insults interrupting the midline specification of the nascent forebrain. Such disturbances can lead to a broad range of phenotypic consequences for the brain and face in humans. This malformation sequence is remarkably common in utero (1 in 250 human fetuses), but 97% typically do not survive to birth. The precise molecular pathogenesis of HPE in these early human embryos remains largely unknown. Here, we outline our current understanding of the principal driving factors leading to HPE pathologies and elaborate our multifactorial integrated genomics approach. Overall, our understanding of the pathogenesis continues to become simpler, rather than more complicated. Genomic technologies now provide unprecedented insight into disease-associated variation, including the overall extent of genetic interactions (coding and noncoding) predicted to explain divergent phenotypes.
Collapse
Affiliation(s)
- Erich Roessler
- Medical Genetics Branch, National Human, Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Ping Hu
- Medical Genetics Branch, National Human, Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Maximilian Muenke
- Medical Genetics Branch, National Human, Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
16
|
Hong M, Krauss RS. Modeling the complex etiology of holoprosencephaly in mice. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2018; 178:140-150. [PMID: 29749693 DOI: 10.1002/ajmg.c.31611] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 03/28/2018] [Accepted: 04/02/2018] [Indexed: 12/14/2022]
Abstract
Holoprosencephaly (HPE) is a common developmental defect caused by failure to define the midline of the forebrain and/or midface. HPE is associated with heterozygous mutations in Nodal and Sonic hedgehog (SHH) pathway components, but clinical presentation is highly variable, and many mutation carriers are unaffected. It is therefore thought that such mutations interact with more common modifiers, genetic and/or environmental, to produce severe patterning defects. Modifiers are difficult to identify, as their effects are context-dependent and occur within the complex genetic and environmental landscapes that characterize human populations. This has made a full understanding of HPE etiology challenging. We discuss here the use of mice, a genetically tractable model sensitive to teratogens, as a system to address this challenge. Mice carrying mutations in human HPE genes often display wide variations in phenotypic penetrance and expressivity when placed on different genetic backgrounds, demonstrating the existence of silent HPE modifier genes. Studies with mouse lines carrying SHH pathway mutations on appropriate genetic backgrounds have led to identification of both genetic and environmental modifiers that synergize with the mutations to produce a spectrum of HPE phenotypes. These models favor a scenario in which multiple modifying influences-both genetic and environmental, sensitizing and protective-interact with bona fide HPE mutations to grade phenotypic outcomes. Despite the complex interplay of HPE risk factors, mouse models have helped establish some clear concepts in HPE etiology. A combination of mouse and human cohort studies should improve our understanding of this fascinating and medically important issue.
Collapse
Affiliation(s)
- Mingi Hong
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
17
|
Ferran JL, Puelles L. Lessons from Amphioxus Bauplan About Origin of Cranial Nerves of Vertebrates That Innervates Extrinsic Eye Muscles. Anat Rec (Hoboken) 2018; 302:452-462. [PMID: 29659196 DOI: 10.1002/ar.23824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 12/23/2022]
Abstract
Amphioxus is the living chordate closest to the ancestral form of vertebrates, and in a key position to reveal essential aspects of the evolution of the brain Bauplan of vertebrates. The dorsal neural cord of this species at the larval stage is characterized by a small cerebral vesicle at its anterior end and a large posterior region. The latter is comparable in some aspects to the hindbrain and spinal cord regions of vertebrates. The rostral end of the cerebral vesicle contains a median pigment spot and associated rows of photoreceptor and other nerve cells; this complex is known as "the frontal eye." However, this is not a complete eye in the sense that it has neither eye muscles nor lens (only a primitive retina-like tissue). Cranial nerves III, IV, and VI take part in the motor control of eye muscles in all vertebrates. Using a recent model that postulates distinct molecularly characterized hypothalamo-prethalamic and mesodiencephalic domains in the early cerebral vesicle of amphioxus, we analyze here possible scenarios for the origin from the common ancestor of cephalochordates and vertebrates of the cranial nerves related with extrinsic eye muscle innervations. Anat Rec, 302:452-462, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- José Luis Ferran
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia - IMIB, Group of Brain Regionalization and genes of development; Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia - IMIB, Group of Brain Regionalization and genes of development; Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| |
Collapse
|
18
|
Percival CJ, Green R, Roseman CC, Gatti DM, Morgan JL, Murray SA, Donahue LR, Mayeux JM, Pollard KM, Hua K, Pomp D, Marcucio R, Hallgrímsson B. Developmental constraint through negative pleiotropy in the zygomatic arch. EvoDevo 2018; 9:3. [PMID: 29423138 PMCID: PMC5787316 DOI: 10.1186/s13227-018-0092-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/08/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Previous analysis suggested that the relative contribution of individual bones to regional skull lengths differ between inbred mouse strains. If the negative correlation of adjacent bone lengths is associated with genetic variation in a heterogeneous population, it would be an example of negative pleiotropy, which occurs when a genetic factor leads to opposite effects in two phenotypes. Confirming negative pleiotropy and determining its basis may reveal important information about the maintenance of overall skull integration and developmental constraint on skull morphology. RESULTS We identified negative correlations between the lengths of the frontal and parietal bones in the midline cranial vault as well as the zygomatic bone and zygomatic process of the maxilla, which contribute to the zygomatic arch. Through gene association mapping of a large heterogeneous population of Diversity Outbred (DO) mice, we identified a quantitative trait locus on chromosome 17 driving the antagonistic contribution of these two zygomatic arch bones to total zygomatic arch length. Candidate genes in this region were identified and real-time PCR of the maxillary processes of DO founder strain embryos indicated differences in the RNA expression levels for two of the candidate genes, Camkmt and Six2. CONCLUSIONS A genomic region underlying negative pleiotropy of two zygomatic arch bones was identified, which provides a mechanism for antagonism in component bone lengths while constraining overall zygomatic arch length. This type of mechanism may have led to variation in the contribution of individual bones to the zygomatic arch noted across mammals. Given that similar genetic and developmental mechanisms may underlie negative correlations in other parts of the skull, these results provide an important step toward understanding the developmental basis of evolutionary variation and constraint in skull morphology.
Collapse
Affiliation(s)
| | - Rebecca Green
- Alberta Children’s Hospital Institute for Child and Maternal Health, University of Calgary, Calgary, AB Canada
- The McCaig Bone and Joint Institute, University of Calgary, Calgary, AB Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB Canada
| | - Charles C. Roseman
- Program in Ecology Evolution and Conservation Biology, University of Illinois, Urbana, IL USA
| | | | | | | | | | - Jessica M. Mayeux
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - K. Michael Pollard
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - Kunjie Hua
- Department of Genetics, University of North Carolina Medical School, Chapel Hill, NC USA
| | - Daniel Pomp
- Department of Genetics, University of North Carolina Medical School, Chapel Hill, NC USA
| | - Ralph Marcucio
- The Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, UCSF School of Medicine, San Francisco, CA USA
| | - Benedikt Hallgrímsson
- Alberta Children’s Hospital Institute for Child and Maternal Health, University of Calgary, Calgary, AB Canada
- The McCaig Bone and Joint Institute, University of Calgary, Calgary, AB Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB Canada
| |
Collapse
|
19
|
Hong M, Srivastava K, Kim S, Allen BL, Leahy DJ, Hu P, Roessler E, Krauss RS, Muenke M. BOC is a modifier gene in holoprosencephaly. Hum Mutat 2017; 38:1464-1470. [PMID: 28677295 DOI: 10.1002/humu.23286] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022]
Abstract
Holoprosencephaly (HPE), a common developmental defect of the forebrain and midface, has a complex etiology. Heterozygous, loss-of-function mutations in the sonic hedgehog (SHH) pathway are associated with HPE. However, mutation carriers display highly variable clinical presentation, leading to an "autosomal dominant with modifier" model, in which the penetrance and expressivity of a predisposing mutation is graded by genetic or environmental modifiers. Such modifiers have not been identified. Boc encodes a SHH coreceptor and is a silent HPE modifier gene in mice. Here, we report the identification of missense BOC variants in HPE patients. Consistent with these alleles functioning as HPE modifiers, individual variant BOC proteins had either loss- or gain-of-function properties in cell-based SHH signaling assays. Therefore, in addition to heterozygous loss-of-function mutations in specific SHH pathway genes and an ill-defined environmental component, our findings identify a third variable in HPE: low-frequency modifier genes, BOC being the first identified.
Collapse
Affiliation(s)
- Mingi Hong
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kshitij Srivastava
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Sungjin Kim
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Daniel J Leahy
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas
| | - Ping Hu
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Erich Roessler
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Maximilian Muenke
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
20
|
Yu Z, Sun Y, She X, Wang Z, Chen S, Deng Z, Zhang Y, Liu Q, Liu Q, Zhao C, Li P, Liu C, Feng J, Fu H, Li G, Wu M. SIX3, a tumor suppressor, inhibits astrocytoma tumorigenesis by transcriptional repression of AURKA/B. J Hematol Oncol 2017; 10:115. [PMID: 28595628 PMCID: PMC5465582 DOI: 10.1186/s13045-017-0483-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/31/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND SIX homeobox 3 (SIX3) is a member of the sine oculis homeobox transcription factor family. It plays a vital role in the nervous system development. Our previous study showed that the SIX3 gene is hypermethylated, and its expression is decreased in astrocytoma, but the role of SIX3 remains unknown. METHODS Chromatin-immunoprecipitation (ChIP) and luciferase reporter assay were used to confirm the binding of SIX3 to the promoter regions of aurora kinase A (AURKA) and aurora kinase B (AURKB). Confocal imaging and co-immunoprecipitation (Co-IP) were used to detect the interaction between AURKA and AURKB. Flow cytometry was performed to assess the effect of SIX3 on cell cycle distribution. Colony formation, EdU incorporation, transwell, and intracranial xenograft assays were performed to demonstrate the effect of SIX3 on the malignant phenotype of astrocytoma cells. RESULTS SIX3 is identified as a novel negative transcriptional regulator of AURKA and AURKB, and it decreases the expression of AURKA and AURKB in a dose-dependent manner in astrocytoma cells. Importantly, interactions between AURKA and AURKB stabilize and protect AURKA/B from degradation, and overexpression of SIX3 does not affect these interactions; SIX3 also acts as a tumor suppressor, and it increases p53 activity and expression at the post-translational level by the negative regulation of AURKA or AURKB, reduces the events of numerical centrosomal aberrations and misaligned chromosomes, and significantly inhibits the proliferation, invasion, and tumorigenesis of astrocytoma in vitro and in vivo. Moreover, experiments using primary cultured astrocytoma cells indicate that astrocytoma patients with a low expression of SIX3 and mutant p53 are more sensitive to treatment with aurora kinase inhibitors. CONCLUSION SIX3 is a novel negative transcriptional regulator and acts as a tumor suppressor that directly represses the transcription of AURKA and AURKB in astrocytoma. For the first time, the functional interaction of AURKA and AURKB has been found, which aids in the protection of their stability, and partially explains their constant high expression and activity in cancers. SIX3 is a potential biomarker that could be used to predict the response of astrocytoma patients to aurora kinase inhibitors.
Collapse
Affiliation(s)
- Zhibin Yu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Yingnan Sun
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Xiaoling She
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zeyou Wang
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shuai Chen
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Zhiyong Deng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Yan Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Qiang Liu
- The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Qing Liu
- The Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chunhua Zhao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Peiyao Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Changhong Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Jianbo Feng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Haijuan Fu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Guiyuan Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
21
|
Kumamoto T, Hanashima C. Evolutionary conservation and conversion of Foxg1 function in brain development. Dev Growth Differ 2017; 59:258-269. [PMID: 28581027 DOI: 10.1111/dgd.12367] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 05/01/2017] [Accepted: 05/01/2017] [Indexed: 12/31/2022]
Abstract
Among the forkhead box protein family, Foxg1 is a unique transcription factor that plays pleiotropic and non-redundant roles in vertebrate brain development. The emergence of the telencephalon at the rostral end of the neural tube and its subsequent expansion that is mediated by Foxg1 was a key reason for the vertebrate brain to acquire higher order information processing, where Foxg1 is repetitively used in the sequential events of telencephalic development to control multi-steps of brain circuit formation ranging from cell cycle control to neuronal differentiation in a clade- and species-specific manner. The objective of this review is to discuss how the evolutionary changes in cis- and trans-regulatory network that is mediated by a single transcription factor has contributed to determining the fundamental vertebrate brain structure and its divergent roles in instructing species-specific neuronal circuitry and functional specialization.
Collapse
Affiliation(s)
- Takuma Kumamoto
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, 75012, Paris, France
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan.,Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan.,Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe, 650-0047, Japan
| |
Collapse
|