1
|
Chen C, Liao Y, Zhu M, Wang L, Yu X, Li M, Peng G. Dual-nuclease single-cell lineage tracing by Cas9 and Cas12a. Cell Rep 2025; 44:115105. [PMID: 39721023 DOI: 10.1016/j.celrep.2024.115105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/30/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Single-cell lineage tracing based on CRISPR-Cas9 gene editing enables the simultaneous linkage of cell states and lineage history at a high resolution. Despite its immense potential in resolving the cell fate determination and genealogy within an organism, existing implementations of this technology suffer from limitations in recording capabilities and considerable barcode dropout. Here, we introduce DuTracer, a versatile tool that utilizes two orthogonal gene editing systems to record cell lineage history at single-cell resolution in an inducible manner. DuTracer shows the ability to enhance lineage recording with minimized target dropouts and potentially deeper tree depths. Applying DuTracer in mouse embryoid bodies and neuromesodermal organoids illustrates the lineage relationship of different cell types and proposes potential lineage-biased molecular drivers, showcased by identifying transcription factor Foxb1 as a modulator in the cell fate determination of neuromesodermal progenitors. Collectively, DuTracer facilitates the precise and regulatory interrogation of cellular lineages of complex biological processes.
Collapse
Affiliation(s)
- Cheng Chen
- Center for Cell Lineage Technology and Engineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yuanxin Liao
- Center for Cell Lineage Technology and Engineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing, China
| | - Miao Zhu
- Center for Cell Lineage Technology and Engineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing, China
| | - Li Wang
- Center for Cell Lineage Technology and Engineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xinran Yu
- Center for Cell Lineage Technology and Engineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Meishi Li
- Center for Cell Lineage Technology and Engineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Guangdun Peng
- Center for Cell Lineage Technology and Engineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| |
Collapse
|
2
|
Trinh LT, Finnel RR, Osipovich AB, Musselman JR, Sampson LL, Wright CVE, Magnuson MA. Positive autoregulation of Sox17 is necessary for gallbladder and extrahepatic bile duct formation. Development 2025; 152:dev203033. [PMID: 39745200 PMCID: PMC11829758 DOI: 10.1242/dev.203033] [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: 05/08/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025]
Abstract
Expression of SRY-box transcription factor 17 (Sox17) in the endodermal region caudal to the hepatic diverticulum during late gastrulation is necessary for hepato-pancreato-biliary system formation. Analysis of an allelic series of promoter-proximal mutations near the transcription start site (TSS) 2 of Sox17 in mouse has revealed that gallbladder (GB) and extrahepatic bile duct (EHBD) development is exquisitely sensitive to Sox17 expression levels. Deletion of a SOX17-binding cis-regulatory element in the TSS2 promoter impairs GB and EHBD development by reducing outgrowth of the nascent biliary bud. These findings reveal the existence of a SOX17-dependent autoregulatory loop that drives Sox17 expression above a critical threshold concentration necessary for GB and EHBD development to occur, and that minor impairments in Sox17 gene expression are sufficient to impair the expression of SOX17-regulated genes in the nascent GB and EHBD system, impairing or preventing development.
Collapse
Affiliation(s)
- Linh T. Trinh
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ryan R. Finnel
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Anna B. Osipovich
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Leesa L. Sampson
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Christopher V. E. Wright
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Mark A. Magnuson
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
3
|
Kinkpe L, Khan R, Suhail SM, Ahmad I, Khan FA, Ayari-Akkari A, Siddiqui S. Polymorphism and association study of lactoferrin (LF) gene with milk yield, milk composition, and somatic cell count in Beetal goats. Trop Anim Health Prod 2023; 55:415. [PMID: 37996555 DOI: 10.1007/s11250-023-03834-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
The sequence analysis of PCR product exhibited four novel SNPs in the promoter region of the LF gene at loci g.98T>C, g.143T>A, g.189AC>A, and g.346A>G. Each SNP yielded three genotypes; the genotypes TT (SNP1), AA (SNP3), and GG (SNP4) decreased SCC and increase milk quality traits such as density, protein, and milk yield (P < 0.01). The genotype CC (SNP2) and CA (SNP4) significantly (P < 0.01) decreased the milk quality parameters, while genotypes TC (SNP2) and GG (SNP4) showed significantly (P < 0.01) less SCC and increase lactose % in milk. Furthermore, screening of the LF promoter sequence explored the gain of four TF binding sites at locus g.98T˃C and three TF binding sites at g.346A˃G. However, the loss of four and two TF binding sites was seen at locus g.143T˃A and g.189C˃A, respectively. We can conclude from the present study that the GG, TT, and AA genotype might be utilized as genetic markers in marker-assisted selection for the breed improvement program of Beetal goats.
Collapse
Affiliation(s)
- Lionel Kinkpe
- Department of Livestock Management, Breeding and Genetics, The University of Agriculture, Peshawar, 25130, Pakistan
| | - Rajwali Khan
- Department of Livestock Management, Breeding and Genetics, The University of Agriculture, Peshawar, 25130, Pakistan.
| | - Syed Muhammad Suhail
- Department of Livestock Management, Breeding and Genetics, The University of Agriculture, Peshawar, 25130, Pakistan
| | - Ijaz Ahmad
- Department of Livestock Management, Breeding and Genetics, The University of Agriculture, Peshawar, 25130, Pakistan
| | - Farhan Anwar Khan
- College of Veterinary Sciences, The University of Agriculture, Peshawar, Pakistan
| | - Amel Ayari-Akkari
- Biology Department, College of Science, King Khalid University, Abha, P.O Box 960, Saudi Arabia
| | - Sazada Siddiqui
- Biology Department, College of Science, King Khalid University, Abha, P.O Box 960, Saudi Arabia
| |
Collapse
|
4
|
Expression of Oocyte Vitellogenesis Receptor Was Regulated by C/EBPα in Developing Follicle of Wanxi White Goose. Animals (Basel) 2022; 12:ani12070874. [PMID: 35405862 PMCID: PMC8997188 DOI: 10.3390/ani12070874] [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: 01/16/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/29/2022] Open
Abstract
Yolk precursor was synthesized under regulation of hormone secretion, while the mechanism of its incorporation into follicle is still unknown. The reproductive hormones, oocyte vitellogenesis receptor (OVR) expression at pre-, early-, peak- and ceased-laying period, and localization of Wanxi White goose were determined in this study. The results showed that the concentration of LH was lowest in serum at peak laying period compared to the other periods (p < 0.01). Moreover, the concentration of E2 was highest (p < 0.01) in serum at early laying period than that of other periods. Moreover, the gene expression level of OVR was highest at ceased laying period compared to other periods (p = 0.014) and was higher in developing follicles than other follicles (p < 0.01). The OVR was distributed in the granular cell layer and decreased with the maturation of follicles. Five transcription factors were predicted in the promoter of OVR, then were screened and verified by overexpression in granulosa cells. C/EBPα and MF3 significantly stimulated the expression of OVR. The combined overexpression of C/EBPα and OVR significantly stimulated the transportation of lipid from culture medium to cytoplasm. In conclusion, C/EBPα is the key transcription factor promoting OVR expression in goose follicle granulosa cells.
Collapse
|
5
|
Lu MM, Li S, Yang H, Morrisey EE. Foxp4: a novel member of the Foxp subfamily of winged-helix genes co-expressed with Foxp1 and Foxp2 in pulmonary and gut tissues. Mech Dev 2016; 119 Suppl 1:S197-202. [PMID: 14516685 DOI: 10.1016/s0925-4773(03)00116-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In this study, we describe the isolation and characterization of Foxp4, a new member of the Foxp subfamily of winged-helix transcription factors. The full-length mouse Foxp4 cDNA encodes a 685-amino-acid protein that is similar to Foxp1 and Foxp2. Foxp4 gene expression is observed primarily in pulmonary, neural, and gut tissues during embryonic development. To compare the protein expression patterns of Foxp4 to Foxp1 and Foxp2, specific polyclonal antisera to each of these proteins was used in immunohistochemical analysis of mouse embryonic tissues. All three proteins are expressed in lung epithelium with Foxp1 and Foxp4 expressed in both proximal and distal airway epithelium while Foxp2 is expressed primarily in distal epithelium. Foxp1 protein expression is also observed in the mesenchyme and vascular endothelial cells of the lung. At embryonic day 12.5, Foxp1 and Foxp2 are expressed in both the mucosal and epithelial layers of the intestine. However, Foxp2 is expressed only in the outer mucosal layer of the intestine and stomach later in development. Finally, Foxp4 is expressed exclusively in the epithelial cells of the developing intestine, where, in late development, it is expressed in a gradient along the longitudinal axis of the villi.
Collapse
Affiliation(s)
- Min Min Lu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
6
|
Chen X, Yao Y, Guan J, Chen X, Zhang F. Up-regulation of FoxN4 expression in adult spinal cord after injury. J Mol Neurosci 2013; 52:403-9. [PMID: 24217796 PMCID: PMC3924027 DOI: 10.1007/s12031-013-0166-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/28/2013] [Indexed: 11/26/2022]
Abstract
FoxN4 (forkhead box N4), which is a transcription factor involved in developing spinal cord and spinal neurogenesis, implied important roles in the central nervous system (CNS). However, its expression and function in the adult CNS lesion are still unclear. In this study, we established a spinal cord injury (SCI) model in adult rats and investigated the expression of FoxN4 in the spinal cord. Western blot analysis revealed that FoxN4 was present in normal spinal cord. It gradually increased, peaked at day 3 after SCI, and then decreased during the following days. Immunohistochemistry further confirmed that FoxN4 was expressed at low levels in gray and white matters in normal condition and increased after SCI. Double immunofluorescence staining showed that FoxN4 is located on neurons and astrocytes, and FoxN4 expression was increased progressively in reactive astrocytes within the vicinity of the lesion, predominately in the white matter. In addition, almost all FoxN4-positive cells also expressed nestin or PCNA. Our data suggested that FoxN4 might play important roles in CNS pathophysiology after SCI.
Collapse
Affiliation(s)
- Xiangdong Chen
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Yu Yao
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Junjie Guan
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Xiaoqing Chen
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| | - Feng Zhang
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, 226001 Jiangsu People’s Republic of China
| |
Collapse
|
7
|
Wolf A, Ryu S. Specification of posterior hypothalamic neurons requires coordinated activities of Fezf2, Otp, Sim1a and Foxb1.2. Development 2013; 140:1762-73. [PMID: 23533176 DOI: 10.1242/dev.085357] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The hypothalamus is a key integrative center in the brain that consists of diverse cell types required for a variety of functions including homeostasis, reproduction, stress response, social and cognitive behavior. Despite our knowledge of several transcription factors crucial for hypothalamic development, it is not known how the wide diversity of neuron types in the hypothalamus is produced. In particular, almost nothing is known about the mechanisms that specify neurons in the posteriormost part of the hypothalamus, the mammillary area. Here, we investigated the specification of two distinct neuron types in the mammillary area that produce the hypothalamic hormones Vasoactive intestinal peptide (Vip) and Urotensin 1 (Uts1). We show that Vip- and Uts1-positive neurons develop in distinct domains in the mammillary area defined by the differential expression of the transcription factors Fezf2, Otp, Sim1a and Foxb1.2. Coordinated activities of these factors are crucial for the establishment of the mammillary area subdomains and the specification of Vip- and Uts1-positive neurons. In addition, Fezf2 is important for early development of the posterior hypothalamus. Thus, our study provides the first molecular anatomical map of the posterior hypothalamus in zebrafish and identifies, for the first time, molecular requirements underlying the specification of distinct posterior hypothalamic neuron types.
Collapse
Affiliation(s)
- Andrea Wolf
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Heidelberg, Germany
| | | |
Collapse
|
8
|
Dhillon SS, Gingerich S, Virtanen C, Belsham DD. Gene array analysis of embryonic- versus adult-derived hypothalamic NPY-expressing cell lines. Mol Cell Endocrinol 2012; 358:116-26. [PMID: 22476083 DOI: 10.1016/j.mce.2012.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 01/24/2012] [Accepted: 03/20/2012] [Indexed: 10/28/2022]
Abstract
Few studies have utilized microarray analysis to understand the genome wide changes involved in the development of the hypothalamus despite its overall importance to basic physiology. Gene expression profiling of immortalized, clonal hypothalamic neurons, embryonic-derived mHypoE-46 and adult-derived mHypoA-2/12, reveals that the expression of 1225 probes was significantly changed between the two neuronal models. Further comparison of the gene expression profiles identified two categories of genes that were confirmed with qRT-PCR: (i) genes implicated in the Wnt signaling pathway; and (ii) transcription factors previously implicated in the development of the central nervous system. Yet, functional analysis of the two cell lines, including hormonal responses and secretion, indicate that they are comparable despite their developmental origin. This study provides a comprehensive analysis of embryonic- and adult-derived hypothalamic neuronal cell models that both express neuropeptide Y, and identifies novel genes as candidates for mediating the development of specific hypothalamic neurons.
Collapse
Affiliation(s)
- Sandeep S Dhillon
- Department of Physiology, University Health Network, Toronto, ON, Canada M5S 1A8
| | | | | | | |
Collapse
|
9
|
Uchibe K, Shimizu H, Yokoyama S, Kuboki T, Asahara H. Identification of novel transcription-regulating genes expressed during murine molar development. Dev Dyn 2012; 241:1217-26. [PMID: 22639370 DOI: 10.1002/dvdy.23808] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The mechanism of tooth development is a complex process regulated by numerous genes including transcription factors, growth factors, and other intra- and extracellular molecules. Especially, transcription factors play a central role in gene expression, regulating a wide spectrum of biological processes including organogenesis. Substantial evidence has been demonstrated by a number of studies using genetically engineered animal models. However, detailed molecular mechanisms of tooth development have not been completely elucidated, partially because numerous genes that play essential roles in tooth development remain unidentified. RESULTS In this study, we conducted an expression-based screening using gene expression database and in situ hybridization assays. Based on the gene expression database "EMBRYS," 207 out of 1,520 genes were expressed in the maxillary and/or mandibular processes and thus were selected for further analysis by section in situ hybridization. Among these candidates, 28 genes were newly identified as potential factors associated with tooth development by in situ hybridization assays with frontal sections of embryonic day 13.5 and 14.5 mouse embryos. The expression patterns were also examined at embryonic day 16.5 and 18.5. CONCLUSIONS These results will contribute to elucidating the mechanisms of tooth development and to improving the technology for regeneration of tooth.
Collapse
Affiliation(s)
- Kenta Uchibe
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Setagaya, Japan
| | | | | | | | | |
Collapse
|
10
|
FOXJ2 expression in rat spinal cord after injury and its role in inflammation. J Mol Neurosci 2012; 47:158-65. [PMID: 22246994 DOI: 10.1007/s12031-011-9704-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 12/30/2011] [Indexed: 02/06/2023]
Abstract
Foxj2 (forkhead box J2), a novel member of the forkhead/HNF3 family, binds DNA with a dual sequence specificity. It may play a role in maintenance and survival of developing and adult neurons. However, its expression and function in the central nervous system lesion are still unclear. In this study, we performed a spinal cord injury (SCI) model in adult Sprague-Dawley rats and investigated the dynamic changes of Foxj2 expression in the spinal cord. Western blot analysis revealed that Foxj2 was present in normal spinal cord. It gradually increased, reached a peak at day 5 after SCI, and then declined during the following days. Double immunofluorescence staining revealed wide expression of Foxj2, which is detected in neurons and astrocytes. After injury, Foxj2 expression was increased predominantly in astrocytes, which highly expressed proliferating cell nuclear antigen, a marker for proliferating cells. And knockdown of Foxj2 in cultured primary astrocytes by siRNA showed that Foxj2 played an important role in lipopolysaccharide-induced inflammatory responses. These results suggested that Foxj2 may be involved in the pathophysiology of SCI, and further research is needed to have a good understanding of its function and mechanism.
Collapse
|
11
|
Weymouth N, Shi Z, Rockey DC. Smooth muscle α actin is specifically required for the maintenance of lactation. Dev Biol 2011; 363:1-14. [PMID: 22123032 DOI: 10.1016/j.ydbio.2011.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 11/03/2011] [Accepted: 11/04/2011] [Indexed: 11/30/2022]
Abstract
Smooth muscle α-actin (Acta2) is one of six highly conserved mammalian actin isoforms that appear to exhibit functional redundancy. Nonetheless, we have postulated a specific functional role for the smooth muscle specific isoform. Here, we show that Acta2 deficient mice have a remarkable mammary phenotype such that dams lacking Acta2 are unable to nurse their offspring effectively. The phenotype was rescued in cross fostering experiments with wild type mice, excluding a developmental defect in Acta2 null pups. The mechanism for the underlying phenotype is due to myoepithelial dysfunction postpartum resulting in precocious involution. Further, we demonstrate a specific defect in myoepithelial cell contractility in Acta2 null mammary glands, despite normal expression of cytoplasmic actins. We conclude that Acta2 specifically mediates myoepithelial cell contraction during lactation and that this actin isoform therefore exhibits functional specificity.
Collapse
Affiliation(s)
- Nate Weymouth
- University of Texas Southwestern Medical Center, Dallas, TX 75390-8887, USA
| | | | | |
Collapse
|
12
|
Takebayashi-Suzuki K, Kitayama A, Terasaka-Iioka C, Ueno N, Suzuki A. The forkhead transcription factor FoxB1 regulates the dorsal-ventral and anterior-posterior patterning of the ectoderm during early Xenopus embryogenesis. Dev Biol 2011; 360:11-29. [PMID: 21958745 DOI: 10.1016/j.ydbio.2011.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 08/04/2011] [Accepted: 09/05/2011] [Indexed: 12/18/2022]
Abstract
The formation of the dorsal-ventral (DV) and anterior-posterior (AP) axes, fundamental to the body plan of animals, is regulated by several groups of polypeptide growth factors including the TGF-β, FGF, and Wnt families. In order to ensure the establishment of the body plan, the processes of DV and AP axis formation must be linked and coordinately regulated. However, the molecular mechanisms responsible for these interactions remain unclear. Here, we demonstrate that the forkhead box transcription factor FoxB1, which is upregulated by the neuralizing factor Oct-25, plays an important role in the formation of the DV and AP axes. Overexpression of FoxB1 promoted neural induction and inhibited BMP-dependent epidermal differentiation in ectodermal explants, thereby regulating the DV patterning of the ectoderm. In addition, FoxB1 was also found to promote the formation of posterior neural tissue in both ectodermal explants and whole embryos, suggesting its involvement in embryonic AP patterning. Using knockdown analysis, we found that FoxB1 is required for the formation of posterior neural tissues, acting in concert with the Wnt and FGF pathways. Consistent with this, FoxB1 suppressed the formation of anterior structures via a process requiring the function of XWnt-8 and eFGF. Interestingly, while downregulation of FoxB1 had little effect on neural induction, we found that it functionally interacted with its upstream factor Oct-25 and plays a supportive role in the induction and/or maintenance of neural tissue. Our results suggest that FoxB1 is part of a mechanism that fine-tunes, and leads to the coordinated formation of, the DV and AP axes during early development.
Collapse
Affiliation(s)
- Kimiko Takebayashi-Suzuki
- Institute for Amphibian Biology, Hiroshima University Graduate School of Science, Kagamiyama 1-3-1, Higashi-Hiroshima, Japan
| | | | | | | | | |
Collapse
|
13
|
Kuroda KO, Tachikawa K, Yoshida S, Tsuneoka Y, Numan M. Neuromolecular basis of parental behavior in laboratory mice and rats: with special emphasis on technical issues of using mouse genetics. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:1205-31. [PMID: 21338647 DOI: 10.1016/j.pnpbp.2011.02.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 02/14/2011] [Accepted: 02/14/2011] [Indexed: 01/07/2023]
Abstract
To support the well-being of the parent-infant relationship, the neuromolecular mechanisms of parental behaviors should be clarified. From neuroanatomical analyses in laboratory rats, the medial preoptic area (MPOA) has been shown to be of critical importance in parental retrieving behavior. More recently, various gene-targeted mouse strains have been found to be defective in different aspects of parental behaviors, contributing to the identification of molecules and signaling pathways required for the behavior. Therefore, the neuromolecular basis of "mother love" is now a fully approachable research field in modern molecular neuroscience. In this review, we will provide a summary of the required brain areas and gene for parental behavior in laboratory mice (Mus musculus) and rats (Rattus norvegicus). Basic protocols and technical considerations on studying the mechanism of parental behavior using genetically-engineered mouse strains will also be presented.
Collapse
Affiliation(s)
- Kumi O Kuroda
- Unit for Affiliative Social Behavior, RIKEN Brain Science Institute, Saitama 351-0198, Japan.
| | | | | | | | | |
Collapse
|
14
|
Lalonde R, Strazielle C. Brain regions and genes affecting limb-clasping responses. ACTA ACUST UNITED AC 2011; 67:252-9. [DOI: 10.1016/j.brainresrev.2011.02.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 02/14/2011] [Accepted: 02/20/2011] [Indexed: 10/18/2022]
|
15
|
Szabó NE, Zhao T, Çankaya M, Stoykova A, Zhou X, Alvarez-Bolado G. Interaction between axons and specific populations of surrounding cells is indispensable for collateral formation in the mammillary system. PLoS One 2011; 6:e20315. [PMID: 21625468 PMCID: PMC3098884 DOI: 10.1371/journal.pone.0020315] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 04/29/2011] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND An essential phenomenon during brain development is the extension of long collateral branches by axons. How the local cellular environment contributes to the initial sprouting of these branches in specific points of an axonal shaft remains unclear. METHODOLOGY/PRINCIPAL FINDINGS The principal mammillary tract (pm) is a landmark axonal bundle connecting ventral diencephalon to brainstem (through the mammillotegmental tract, mtg). Late in development, the axons of the principal mammillary tract sprout collateral branches at a very specific point forming a large bundle whose target is the thalamus. Inspection of this model showed a number of distinct, identified cell populations originated in the dorsal and the ventral diencephalon and migrating during development to arrange themselves into several discrete groups around the branching point. Further analysis of this system in several mouse lines carrying mutant alleles of genes expressed in defined subpopulations (including Pax6, Foxb1, Lrp6 and Gbx2) together with the use of an unambiguous genetic marker of mammillary axons revealed: 1) a specific group of Pax6-expressing cells in close apposition with the prospective branching point is indispensable to elicit axonal branching in this system; and 2) cooperation of transcription factors Foxb1 and Pax6 to differentially regulate navigation and fasciculation of distinct branches of the principal mammillary tract. CONCLUSIONS/SIGNIFICANCE Our results define for the first time a model system where interaction of the axonal shaft with a specific group of surrounding cells is essential to promote branching. Additionally, we provide insight on the cooperative transcriptional regulation necessary to promote and organize an intricate axonal tree.
Collapse
Affiliation(s)
- Nora-Emöke Szabó
- Brain Development Group, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | - Tianyu Zhao
- Brain Development Group, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | - Murat Çankaya
- Department of Biology, Faculty of Sciences and Art, Erzincan University, Erzincan, Turkey
| | - Anastassia Stoykova
- Department of Molecular Cell Biology, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | - Xunlei Zhou
- Brain Development Group, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | - Gonzalo Alvarez-Bolado
- Brain Development Group, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
16
|
Wong L, Weadick CJ, Kuo C, Chang BSW, Tropepe V. Duplicate dmbx1 genes regulate progenitor cell cycle and differentiation during zebrafish midbrain and retinal development. BMC DEVELOPMENTAL BIOLOGY 2010; 10:100. [PMID: 20860823 PMCID: PMC2954992 DOI: 10.1186/1471-213x-10-100] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 09/22/2010] [Indexed: 01/03/2023]
Abstract
Background The Dmbx1 gene is important for the development of the midbrain and hindbrain, and mouse gene targeting experiments reveal that this gene is required for mediating postnatal and adult feeding behaviours. A single Dmbx1 gene exists in terrestrial vertebrate genomes, while teleost genomes have at least two paralogs. We compared the loss of function of the zebrafish dmbx1a and dmbx1b genes in order to gain insight into the molecular mechanism by which dmbx1 regulates neurogenesis, and to begin to understand why these duplicate genes have been retained in the zebrafish genome. Results Using gene knockdown experiments we examined the function of the dmbx1 gene paralogs in zebrafish, dmbx1a and dmbx1b in regulating neurogenesis in the developing retina and midbrain. Dose-dependent loss of dmbx1a and dmbx1b function causes a significant reduction in growth of the midbrain and retina that is evident between 48-72 hpf. We show that this phenotype is not due to patterning defects or persistent cell death, but rather a deficit in progenitor cell cycle exit and differentiation. Analyses of the morphant retina or anterior hindbrain indicate that paralogous function is partially diverged since loss of dmbx1a is more severe than loss of dmbx1b. Molecular evolutionary analyses of the Dmbx1 genes suggest that while this gene family is conservative in its evolution, there was a dramatic change in selective constraint after the duplication event that gave rise to the dmbx1a and dmbx1b gene families in teleost fish, suggestive of positive selection. Interestingly, in contrast to zebrafish dmbx1a, over expression of the mouse Dmbx1 gene does not functionally compensate for the zebrafish dmbx1a knockdown phenotype, while over expression of the dmbx1b gene only partially compensates for the dmbx1a knockdown phenotype. Conclusion Our data suggest that both zebrafish dmbx1a and dmbx1b genes are retained in the fish genome due to their requirement during midbrain and retinal neurogenesis, although their function is partially diverged. At the cellular level, Dmbx1 regulates cell cycle exit and differentiation of progenitor cells. The unexpected observation of putative post-duplication positive selection of teleost Dmbx1 genes, especially dmbx1a, and the differences in functionality between the mouse and zebrafish genes suggests that the teleost Dmbx1 genes may have evolved a diverged function in the regulation of neurogenesis.
Collapse
Affiliation(s)
- Loksum Wong
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
17
|
A genomic atlas of mouse hypothalamic development. Nat Neurosci 2010; 13:767-75. [PMID: 20436479 DOI: 10.1038/nn.2545] [Citation(s) in RCA: 288] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 03/11/2010] [Indexed: 01/09/2023]
Abstract
The hypothalamus is a central regulator of many behaviors that are essential for survival, such as temperature regulation, food intake and circadian rhythms. However, the molecular pathways that mediate hypothalamic development are largely unknown. To identify genes expressed in developing mouse hypothalamus, we performed microarray analysis at 12 different developmental time points. We then conducted developmental in situ hybridization for 1,045 genes that were dynamically expressed over the course of hypothalamic neurogenesis. We identified markers that stably labeled each major hypothalamic nucleus over the entire course of neurogenesis and constructed a detailed molecular atlas of the developing hypothalamus. As a proof of concept of the utility of these data, we used these markers to analyze the phenotype of mice in which Sonic Hedgehog (Shh) was selectively deleted from hypothalamic neuroepithelium and found that Shh is essential for anterior hypothalamic patterning. Our results serve as a resource for functional investigations of hypothalamic development, connectivity, physiology and dysfunction.
Collapse
|
18
|
Morikawa Y, Hisaoka T, Senba E. Characterization of Foxp2-expressing cells in the developing spinal cord. Neuroscience 2009; 162:1150-62. [PMID: 19463901 DOI: 10.1016/j.neuroscience.2009.05.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 05/10/2009] [Accepted: 05/12/2009] [Indexed: 02/03/2023]
Abstract
Two members of winged-helix/forkhead transcription factors, Foxp1 and Foxp2, are expressed in the developing and adult CNS, including the striatum, cerebral cortex, and thalamus. In a previous study, we have demonstrated that Foxp1 is expressed in a subpopulation of V1 interneurons in addition to motor neurons of the spinal cord during mouse embryogenesis. However, the detailed expression pattern of Foxp2 and its relationship with Foxp1 in the developing spinal cord remains to be elucidated. To shed light on the potential roles of Foxp1 and Foxp2 in the developing spinal cord, we characterized Foxp2-expressing cells during mouse embryogenesis. At embryonic day (E) 11.0, Foxp2-expressing cells were first observed in the ventral spinal cord, which were Pax6(-), p27(+), and neuron-specific class III beta-tubulin(+) postmitotic neurons. Between E13.5 and E15.5, high expression of Foxp2 was observed in both medial and lateral parts of the ventral spinal cord. Double-immunofluorescence staining for Foxp2 with some homeodomain transcription factors revealed that Foxp2-expressing neurons were Pax2(+), En1(+), Evx1(-), Chx10(-), Gata3(-), and Lhx3(-) V1 interneurons in the intermediate zone throughout the ventral spinal cord, indicating that Foxp2-expressing neurons were also V1 interneurons with the same phenotypes as Foxp1-expressing interneurons. In addition, neither Foxp1 nor Foxp2 was expressed in ventral calbindin(+) Renshaw cells. However, Foxp2 did not colocalize with Foxp1 in interneurons of the ventral spinal cord. These findings suggest that Foxp1 and Foxp2 are expressed in the distinct subsets of V1 interneurons that belong to non-Renshaw cells in the ventral spinal cord during embryogenesis. Thus, Foxp1 and Foxp2 may be involved in the determination of the cell type identities during late embryogenesis: the classes of neurotransmitters and the functional subtypes of non-Renshaw cells, such as Ia and Ib inhibitory interneurons.
Collapse
Affiliation(s)
- Y Morikawa
- Department of Anatomy and Neurobiology, Wakayama Medical University, 811-1, Kimiidera, Wakayama 641-8509, Japan.
| | | | | |
Collapse
|
19
|
Zhao T, Szabó N, Ma J, Luo L, Zhou X, Alvarez-Bolado G. Genetic mapping of Foxb1-cell lineage shows migration from caudal diencephalon to telencephalon and lateral hypothalamus. Eur J Neurosci 2009; 28:1941-55. [PMID: 19046377 PMCID: PMC2777254 DOI: 10.1111/j.1460-9568.2008.06503.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The hypothalamus is a brain region with vital functions, and alterations in its development can cause human disease. However, we still do not have a complete description of how this complex structure is put together during embryonic and early postnatal stages. Radially oriented, outside-in migration of cells is prevalent in the developing hypothalamus. In spite of this, cell contingents from outside the hypothalamus as well as tangential hypothalamic migrations also have an important role. Here we study migrations in the hypothalamic primordium by genetically labeling the Foxb1 diencephalic lineage. Foxb1 is a transcription factor gene expressed in the neuroepithelium of the developing neural tube with a rostral expression boundary between caudal and rostral diencephalon, and therefore appropriate for marking migrations from caudal levels into the hypothalamus. We have found a large, longitudinally oriented migration stream apparently originating in the thalamic region and following an axonal bundle to end in the anterior portion of the lateral hypothalamic area. Additionally, we have mapped a specific expansion of the neuroepithelium into the rostral diencephalon. The expanded neuroepithelium generates abundant neurons for the medial hypothalamus at the tuberal level. Finally, we have uncovered novel diencephalon-to-telencephalon migrations into septum, piriform cortex and amygdala.
Collapse
Affiliation(s)
- Tianyu Zhao
- Department of Genes and Behavior, Brain Development Group, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Sato Y, Baba T, Zubair M, Miyabayashi K, Toyama Y, Maekawa M, Owaki A, Mizusaki H, Sawamura T, Toshimori K, Morohashi KI, Katoh-Fukui Y. Importance of forkhead transcription factor Fkhl18 for development of testicular vasculature. Mol Reprod Dev 2008; 75:1361-71. [PMID: 18288644 DOI: 10.1002/mrd.20888] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Forkhead transcription factors are characterized by a winged helix DNA binding domain, and the members of this family are classified into 20 subclasses by phylogenetic analyses. Fkhl18 is structurally unique, and is classified into FoxS subfamily. We found Fkhl18 expression in periendothelial cells of the developing mouse fetal testis. In an attempt to clarify its function, we generated mice with Fkhl18 gene disruption. Although KO mice developed normally and were fertile in both sexes, we frequently noticed unusual blood accumulation in the fetal testis. Electron microscopic analysis demonstrated frequent gaps, measuring 100-400 nm, in endothelial cells of blood vessels. These gaps probably represented ectopic apoptosis of testicular periendothelial cells, identified by caspase-3 expression, in KO fetuses. No apoptosis of endothelial cells was noted. Fkhl18 suppressed the transcriptional activity of FoxO3a and FoxO4. Considering that Fas ligand gene expression is activated by Foxs, the elevated activity of Foxs in the absence of Fkhl18 probably explains the marked apoptosis of periendothelial cells in Fkhl18 KO mice.
Collapse
Affiliation(s)
- Yuko Sato
- Division of Sex Differentiation, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The basis of social evolution in mammals is the mother-offspring relationship. It is also the primary and most important instance of indirect genetic effects, where genetic variation in one individual affects phenotypic variation among others. This relationship is so important in mammals that often the major factor determining the life or death of newborns is the environment provided by their mother. Variations in these environments can be due to variations in maternal genotypes. In our work with the intercross of two mouse inbred strains, LG/J and SM/J, we uncovered a very severe variation in maternal performance. These females failed to nurture their offspring and showed abnormal maternal behaviors leading to loss of their litter. Rather than this being due to a single gene variant as in knockout mice, we uncovered a complex genetic basis for this trait. The effects of genes on maternal performance are entirely context dependent in our cross. They depend on the alleles present at the same or other epistatically interacting loci. Genomic locations identified in this study include locations of candidate genes whose knockouts displayed similar aberrant maternal behavior. Behaviors significantly associated with maternal performance in this study include suckling, nest building, placentophagia, pup grooming, and retrieval of pups after disturbance.
Collapse
Affiliation(s)
- Andréa C Peripato
- Department of Biology/Genetics, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP 05508-900, Brazil
| | | |
Collapse
|
22
|
Harris MJ, Juriloff DM. Mouse mutants with neural tube closure defects and their role in understanding human neural tube defects. ACTA ACUST UNITED AC 2007; 79:187-210. [PMID: 17177317 DOI: 10.1002/bdra.20333] [Citation(s) in RCA: 241] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The number of mouse mutants and strains with neural tube closure defects (NTDs) now exceeds 190, including 155 involving known genes, 33 with unidentified genes, and eight "multifactorial" strains. METHODS The emerging patterns of mouse NTDs are considered in relation to the unknown genetics of the common human NTDs, anencephaly, and spina bifida aperta. RESULTS Of the 150 mouse mutants that survive past midgestation, 20% have risk of either exencephaly and spina bifida aperta or both, parallel to the majority of human NTDs, whereas 70% have only exencephaly, 5% have only spina bifida, and 5% have craniorachischisis. The primary defect in most mouse NTDs is failure of neural fold elevation. Most null mutations (>90%) produce syndromes of multiple affected structures with high penetrance in homozygotes, whereas the "multifactorial" strains and several null-mutant heterozygotes and mutants with partial gene function (hypomorphs) have low-penetrance nonsyndromic NTDs, like the majority of human NTDs. The normal functions of the mutated genes are diverse, with clusters in pathways of actin function, apoptosis, and chromatin methylation and structure. The female excess observed in human anencephaly is found in all mouse exencephaly mutants for which gender has been studied. Maternal agents, including folate, methionine, inositol, or alternative commercial diets, have specific preventative effects in eight mutants and strains. CONCLUSIONS If the human homologs of the mouse NTD mutants contribute to risk of common human NTDs, it seems likely to be in multifactorial combinations of hypomorphs and low-penetrance heterozygotes, as exemplified by mouse digenic mutants and the oligogenic SELH/Bc strain.
Collapse
Affiliation(s)
- Muriel J Harris
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| | | |
Collapse
|
23
|
Foxb1-drivenCre expression in somites and the neuroepithelium of diencephalon, brainstem, and spinal cord. Genesis 2007; 45:781-7. [DOI: 10.1002/dvg.20356] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
24
|
Wijchers PJEC, Burbach JPH, Smidt MP. In control of biology: of mice, men and Foxes. Biochem J 2006; 397:233-46. [PMID: 16792526 PMCID: PMC1513289 DOI: 10.1042/bj20060387] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 05/04/2006] [Accepted: 05/05/2006] [Indexed: 12/11/2022]
Abstract
Forkhead proteins comprise a highly conserved family of transcription factors, named after the original forkhead gene in Drosophila. To date, over 100 forkhead genes have been identified in a large variety of species, all sharing the evolutionary conserved 'forkhead' DNA-binding domain, and the cloning and characterization of forkhead genes have continued in recent years. Forkhead transcription factors regulate the expression of countless genes downstream of important signalling pathways in most, if not all, tissues and cell types. Recent work has provided novel insights into the mechanisms that contribute to their functional diversity, including functional protein domains and interactions of forkheads with other transcription factors. Studies using loss- and gain-of-function models have elucidated the role of forkhead factors in developmental biology and cellular functions such as metabolism, cell division and cell survival. The importance of forkhead transcription factors is underlined by the developmental defects observed in mutant model organisms, and multiple human disorders and cancers which can be attributed to mutations within members of the forkhead gene family. This review provides a comprehensive overview of current knowledge on forkhead transcription factors, from structural organization and regulatory mechanisms to cellular and developmental functions in mice and humans. Finally, we will discuss how novel insights gained from involvement of 'Foxes' in the mechanisms underlying human pathology may create new opportunities for treatment strategies.
Collapse
Key Words
- cell cycle
- development
- forkhead
- fox
- immunoregulation
- transcription factor
- cbp, creb (camp-response-element-binding protein)-binding protein
- ccnb, cyclin b
- cdk, cyclin-dependent kinase
- cki, cdk inhibitor
- dyrk1a, dual-specificity tyrosine-phosphorylated and -regulated kinase 1a
- er, oestrogen receptor
- fha, forkhead-associated domain
- fm, foxh1 motif
- fox, forkhead box
- gadd45a, growth arrest and dna-damage-inducible protein 45α
- hdac, histone deacetylase
- iκb, inhibitory κb
- ikkβ, iκb kinase β
- mh domain, mothers against decapentaplegic homology domain
- nf-κb, nuclear factor κb
- nls, nuclear localization signal
- pkb, protein kinase b
- plk-1, polo-like kinase 1
- scf, skp2/cullin/f-box
- sgk, serum- and glucocorticoid-induced protein kinase
- smad, similar to mothers against decapentaplegic
- sid, smad-interaction domain
- sim, smad-interaction motif
- tgfβ, transforming growth factor β
Collapse
Affiliation(s)
- Patrick J E C Wijchers
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands.
| | | | | |
Collapse
|
25
|
Liljander M, Sällström MA, Andersson S, Wernhoff P, Andersson A, Holmdahl R, Mattsson R. Identification of genetic regions of importance for reproductive performance in female mice. Genetics 2006; 173:901-9. [PMID: 16547111 PMCID: PMC1526510 DOI: 10.1534/genetics.105.054049] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Both environmental and genetic factors can dramatically affect reproductive performance in mice. In this study we have focused on the identification of genetic regions, quantitative trait loci (QTL), which affect the breeding capacity of female mice. We have identified polymorphic microsatellite markers for the mouse strains used and performed a genomewide scan on 237 females from a gene-segregating backcross between a high breeder and a relatively poor breeder. The high-breeder mouse strain we used is the inbred NFR/N mouse (MHC haplotype H-2q), which has extraordinary good breeding properties. The moderate breeder chosen for F(1) and N2 progeny was B10.Q, which is a genetically well-characterized MHC-congenic mouse of the H-2q haplotype. Each of the 237 females of the N2 generation was allowed to mate twice with MHC-congenic B10.RIII (H-2r) males and twice with B10.Q males. A predetermined number of phenotypes related to reproductive performance were recorded, and these included litter size, neonatal growth, and pregnancy rate. Loci controlling litter size were detected on chromosomes 1 (Fecq3) and 9 (Fecq4). The neonatal growth phenotype was affected by Fecq3 and a locus on chromosome 9 (Neogq1). On chromosome 11 two loci affecting the pregnancy rate (Pregq1 and Pregq2) were identified. Furthermore, on chromosomes 13 and 17 we found loci (Pregq3 and Pregq4) influencing the outcome of allogeneic pregnancy (allogeneic by means of MHC disparity between mother and fetuses). A locus on chromosome 1 affecting maternal body weight was also identified and has been denoted Bwq7. It is well known that reproductive performance is polygenically controlled, and the identification of the major loci in this complex process opens the possibility of investigating the natural genetic control of reproduction.
Collapse
|
26
|
Wijchers PJEC, Hoekman MFM, Burbach JPH, Smidt MP. Identification of forkhead transcription factors in cortical and dopaminergic areas of the adult murine brain. Brain Res 2006; 1068:23-33. [PMID: 16376864 DOI: 10.1016/j.brainres.2005.11.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 11/02/2005] [Accepted: 11/10/2005] [Indexed: 11/30/2022]
Abstract
The murine forkhead family of transcription factors consists of over 30 members, the vast majority of which is important in embryonic development. Implicated in processes such as proliferation, differentiation and survival, forkhead factors show highly restricted expression patterns. In search for forkhead genes expressed in specific neural systems, we identified multiple family members. We performed a detailed expression analysis for Foxj2, Foxk1 and the murine orthologue of the human ILF1 gene, which show a remarkable preference for complex cortical structures. In addition, a comprehensive examination of forkhead gene expression in dopamine neurons of the ventral tegmental area and substantia nigra pars compacta, revealed Ilf1 as a novel transcriptional regulator in midbrain dopamine neurons. These forkhead transcription factors may play a role in maintenance and survival of developing and adult neurons.
Collapse
Affiliation(s)
- Patrick J E C Wijchers
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | | | | | | |
Collapse
|
27
|
Wijchers PJEC, Hoekman MFM, Burbach JPH, Smidt MP. Cloning and analysis of the murine Foxi2 transcription factor. ACTA ACUST UNITED AC 2005; 1731:133-8. [PMID: 16289364 DOI: 10.1016/j.bbaexp.2005.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2005] [Revised: 08/31/2005] [Accepted: 09/13/2005] [Indexed: 02/06/2023]
Abstract
Forkhead transcription factors comprise a large family of key regulators of embryonic development. Here, we describe the cloning and analysis of the murine Foxi2 gene, coding for a putative 311 amino acid protein resembling Foxi subfamily members in mice and other species. Expression analysis during the final stages of embryonic development revealed that Foxi2 expression is mainly confined to subsets of cells in epithelial structures and particular ducts, in addition to the developing forebrain and neural retina. Since FoxI factors are thought to be implicated in the regulation of cell fate, the highly restricted expression pattern of Foxi2 suggestive of a possible role in controlling cellular identity.
Collapse
Affiliation(s)
- Patrick J E C Wijchers
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | | | | | | |
Collapse
|
28
|
Heglind M, Cederberg A, Aquino J, Lucas G, Ernfors P, Enerbäck S. Lack of the central nervous system- and neural crest-expressed forkhead gene Foxs1 affects motor function and body weight. Mol Cell Biol 2005; 25:5616-25. [PMID: 15964817 PMCID: PMC1157007 DOI: 10.1128/mcb.25.13.5616-5625.2005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To gain insight into the expression pattern and functional importance of the forkhead transcription factor Foxs1, we constructed a Foxs1-beta-galactosidase reporter gene "knock-in" (Foxs1beta-gal/beta-gal) mouse, in which the wild-type (wt) Foxs1 allele has been inactivated and replaced by a beta-galactosidase reporter gene. Staining for beta-galactosidase activity reveals an expression pattern encompassing neural crest-derived cells, e.g., cranial and dorsal root ganglia as well as several other cell populations in the central nervous system (CNS), most prominently the internal granule layer of cerebellum. Other sites of expression include the lachrymal gland, outer nuclear layer of retina, enteric ganglion neurons, and a subset of thalamic and hypothalamic nuclei. In the CNS, blood vessel-associated smooth muscle cells and pericytes stain positive for Foxs1. Foxs1beta-gal/beta-gal mice perform significantly better (P < 0.01) on a rotating rod than do wt littermates. We have also noted a lower body weight gain (P < 0.05) in Foxs1beta-gal/lbeta-gal males on a high-fat diet, and we speculate that dorsomedial hypothalamic neurons, expressing Foxs1, could play a role in regulating body weight via regulation of sympathetic outflow. In support of this, we observed increased levels of uncoupling protein 1 mRNA in Foxs1beta-gal/beta-gal mice. This points toward a role for Foxs1 in the integration and processing of neuronal signals of importance for energy turnover and motor function.
Collapse
Affiliation(s)
- Mikael Heglind
- Medical Genetics, Department of Medical Biochemistry, Göteborg University, Medicinareg. 9A, Box 440, SE 405 30 Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
29
|
Moret F, Christiaen L, Deyts C, Blin M, Vernier P, Joly JS. Regulatory gene expressions in the ascidian ventral sensory vesicle: evolutionary relationships with the vertebrate hypothalamus. Dev Biol 2005; 277:567-79. [PMID: 15617694 DOI: 10.1016/j.ydbio.2004.11.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2004] [Revised: 10/13/2004] [Accepted: 11/03/2004] [Indexed: 11/28/2022]
Abstract
In extant chordates, the overall patterning along the anteroposterior and dorsoventral axes of the neural tube is remarkably conserved. It has thus been proposed that four domains corresponding to the vertebrate presumptive forebrain, midbrain-hindbrain transition, hindbrain, and spinal cord were already present in the common chordate ancestor. To obtain insights on the evolution of the patterning of the anterior neural tube, we performed a study aimed at characterizing the expression of regulatory genes in the sensory vesicle of Ciona intestinalis, the anteriormost part of the central nervous system (CNS) related to the vertebrate forebrain, at tailbud stages. Selected genes encoded primarily for homologues of transcription factors involved in vertebrate forebrain patterning. Seven of these genes were expressed in the ventral sensory vesicle. A prominent feature of these ascidian genes is their restricted and complementary domains of expression at tailbud stages. These patterning markers thus refine the map of the developing sensory vesicle. Furthermore, they allow us to propose that a large part of the ventral and lateral sensory vesicle consists in a patterning domain corresponding to the vertebrate presumptive hypothalamus.
Collapse
Affiliation(s)
- Frédéric Moret
- Development, Evolution and Plasticity of the Nervous System, Institut de Neurobiologie Alfred Fessard, Centre National de la Recherche Scientifique, UPR2197, 1 ave de la terrasse, F-91198 Gif-sur-Yvette, France
| | | | | | | | | | | |
Collapse
|
30
|
Ohtoshi A, Behringer RR. Neonatal lethality, dwarfism, and abnormal brain development in Dmbx1 mutant mice. Mol Cell Biol 2004; 24:7548-58. [PMID: 15314164 PMCID: PMC507007 DOI: 10.1128/mcb.24.17.7548-7558.2004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dmbx1 encodes a paired-like homeodomain protein that is expressed in developing neural tissues during mouse embryogenesis. To elucidate the in vivo role of Dmbx1, we generated two Dmbx1 mutant alleles. Dmbx1- lacks the homeobox and Dmbx1z is an insertion of a lacZ reporter gene. Dmbx1z appears to be a faithful reporter of Dmbx1 expression during embryogenesis and after birth. Dmbx1-lacZ expression was detected in the superior colliculus, cerebellar nuclei, and subpopulations of the medulla oblongata and spinal cord. Some Dmbx1 homozygous mutant mice died during the neonatal period, while others survived to adulthood; however, their growth was impaired. Both heterozygous and homozygous mutant offspring from Dmbx1 homozygous mutant females exhibited a low survival rate and poor growth. However, even wild-type pups fostered onto Dmbx1 homozygous mutant females grew poorly, suggesting a Dmbx1-dependent nursing defect. Dmbx1 mutant mice had an aberrant Dmbx1-lacZ expression pattern in the nervous system, indicating that they had abnormal brain development. These results demonstrate that Dmbx1 is required for postnatal survival, growth, and brain development.
Collapse
Affiliation(s)
- Akihira Ohtoshi
- Department of Molecular Genetics, University of Texas, M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | |
Collapse
|
31
|
Pierce EA, Liu Q, Igoucheva O, Omarrudin R, Ma H, Diamond SL, Yoon K. Oligonucleotide-directed single-base DNA alterations in mouse embryonic stem cells. Gene Ther 2003; 10:24-33. [PMID: 12525834 DOI: 10.1038/sj.gt.3301857] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We have investigated the use of single-stranded oligodeoxy-nucleotides (ssODN) to produce specific single-base alterations in episomal and chromosomal DNA in mouse embryonic stem (ES) cells. Two different reporter genes, EGFP and LacZ, each with a single point mutation that inactivates reporter activity, were used. ssODN homologous to the target sequence, except for a single mismatch at the mutant base, were used to correct the mutant reporter genes. When tested in CHO-K1 cells, the ssODN showed correction rates of 0.5-1.0%, consistent with prior reports. ssODN in the antisense orientation provided higher rates of gene conversion than those in the sense orientation for both reporter genes. Nuclear extracts from mouse ES cells exhibited nearly the same correction activity as extracts from CHO-K1 cells. ssODN corrected the mutant bases of both episomal and chromosomal mutant reporter genes in mouse ES cells. Although the efficiency of gene correction observed in ES cells is low, approximately 10(-4), these results demonstrate that ssODN can produce single-base alterations in the genomic DNA of mouse ES cells. As conversion efficiency is improved by the continued development of oligonucleotide structure and DNA delivery methods, ssODN could be used to produce ES cells with specific mutations in any gene in a single step. The targeted ES cells could in turn be used to create accurate mouse models of inherited diseases.
Collapse
Affiliation(s)
- E A Pierce
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Amphioxus is the closest relative to vertebrates but lacks key vertebrate characters, like rhombomeres, neural crest cells, and the cartilaginous endoskeleton. This reflects major differences in the developmental patterning of neural and mesodermal structures between basal chordates and vertebrates. Here, we analyse the expression pattern of an amphioxus FoxB ortholog and an amphioxus single-minded ortholog to gain insight into the evolution of vertebrate neural segmentation. AmphiFoxB expression shows cryptic segmentation of the cerebral vesicle and hindbrain, suggesting that neuromeric segmentation of the chordate neural tube arose before the origin of the vertebrates. In the forebrain, AmphiFoxB expression combined with AmphiSim and other amphioxus gene expression patterns shows that the cerebral vesicle is divided into several distinct domains: we propose homology between these domains and the subdivided diencephalon and midbrain of vertebrates. In the Hox-expressing region of the amphioxus neural tube that is homologous to the vertebrate hindbrain, AmphiFoxB shows the presence of repeated blocks of cells along the anterior-posterior axis, each aligned with a somite. This and other data lead us to propose a model for the evolution of vertebrate rhombomeric segmentation, in which rhombomere evolution involved the transfer of mechanisms regulating neural segmentation from vertical induction by underlying segmented mesoderm to horizontal induction by graded retinoic acid signalling. A consequence of this would have been that segmentation of vertebrate head mesoderm would no longer have been required, paving the way for the evolution of the unsegmented head mesoderm seen in living vertebrates.
Collapse
Affiliation(s)
- Françoise Mazet
- School of Animal and Microbial Sciences, The University of Reading, P.O. Box 228, Whiteknights, Reading, RG6 6AJ, United Kingdom
| | | |
Collapse
|
33
|
Carlsson P, Mahlapuu M. Forkhead transcription factors: key players in development and metabolism. Dev Biol 2002; 250:1-23. [PMID: 12297093 DOI: 10.1006/dbio.2002.0780] [Citation(s) in RCA: 673] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Peter Carlsson
- Department of Molecular Biology, Göteborg University, Box 462, SE-405 30 Göteborg, Sweden.
| | | |
Collapse
|
34
|
Peripato AC, De Brito RA, Vaughn TT, Pletscher LS, Matioli SR, Cheverud JM. Quantitative trait loci for maternal performance for offspring survival in mice. Genetics 2002; 162:1341-53. [PMID: 12454078 PMCID: PMC1462336 DOI: 10.1093/genetics/162.3.1341] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Maternal performance refers to the effect that the environment provided by mothers has on their offspring's phenotypes, such as offspring survival and growth. Variations in maternal behavior and physiology are responsible for variations in maternal performance, which in turn affects offspring survival. In our study we found females that failed to nurture their offspring and showed abnormal maternal behaviors. The genetic architecture of maternal performance for offspring survival was investigated in 241 females of an F(2) intercross of the SM/J and LG/J inbred mouse strains. Using interval-mapping methods we found two quantitative trait loci (QTL) affecting maternal performance at D2Mit17 + 6 cM and D7Mit21 + 2 cM on chromosomes 2 and 7, respectively. In a two-way genome-wide epistasis scan we found 15 epistatic interactions involving 23 QTL distributed across all chromosomes except 12, 16, and 17. These loci form several small sets of interacting QTL, suggesting a complex set of mechanisms operating to determine maternal performance for offspring survival. Taken all together and correcting for the large number of significant factors, QTL and their interactions explain almost 35% of the phenotypic variation for maternal performance for offspring survival in this cross. This study allowed the identification of many possible candidate genes, as well as the relative size of gene effects and patterns of gene action affecting maternal performance in mice. Detailed behavior observation of mothers from later generations suggests that offspring survival in the first week is related to maternal success in building nests, grooming their pups, providing milk, and/or manifesting aggressive behavior against intruders.
Collapse
Affiliation(s)
- Andréa C Peripato
- Department of Biology/Genetics, IB, Universidade de São Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
35
|
Arden KC, Biggs WH. Regulation of the FoxO family of transcription factors by phosphatidylinositol-3 kinase-activated signaling. Arch Biochem Biophys 2002; 403:292-8. [PMID: 12139979 DOI: 10.1016/s0003-9861(02)00207-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Karen C Arden
- The Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093-0660, USA.
| | | |
Collapse
|
36
|
Abstract
The development of the neuroendocrine hypothalamus has been studied using a variety of neuroanatomical and molecular techniques. Here, the major findings that mold our understanding of hypothalamic development are reviewed. The rat hypothalamus is generated predominantly from the third ventricular neuroepithelium in a "lateral early to medial late" pattern dictated perhaps by the medially receding third ventricle. Neuroendocrine neurons seem to exhibit a delayed migrational strategy, showing relatively early birthdates, although they are located in the latest-generated, periventricular nuclei. Several homeobox genes seem to play a role in hypothalamic development, and gene knockout experiments implicate a number of genes of importance in the generation of the neuroendocrine cell type.
Collapse
Affiliation(s)
- Eleni A Markakis
- Department of Neurobiology, Yale University School of Medicine, P.O. Box 208001, New Haven, CT 06510-8001, USA.
| |
Collapse
|
37
|
Lee CS, Perreault N, Brestelli JE, Kaestner KH. Neurogenin 3 is essential for the proper specification of gastric enteroendocrine cells and the maintenance of gastric epithelial cell identity. Genes Dev 2002; 16:1488-97. [PMID: 12080087 PMCID: PMC186338 DOI: 10.1101/gad.985002] [Citation(s) in RCA: 237] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The notch signaling pathway is essential for the endocrine cell fate in various tissues including the enteroendocrine system of the gastrointestinal tract. Enteroendocrine cells are one of the four major cell types found in the gastric epithelium of the glandular stomach. To understand the molecular basis of enteroendocrine cell development, we have used gene targeting in mouse embryonic stem cells to derive an EGFP-marked null allele of the bHLH transcription factor, neurogenin 3 (ngn3). In ngn3(-/-) mice, glucagon secreting A-cells, somatostatin secreting D-cells, and gastrin secreting G-cells are absent from the epithelium of the glandular stomach, whereas the number of serotonin-expressing enterochromaffin (EC) cells is decreased dramatically. In addition, ngn3(-/-) mice display intestinal metaplasia of the gastric epithelium. Thus, ngn3 is required for the differentiation of enteroendocrine cells in the stomach and the maintenance of gastric epithelial cell identity.
Collapse
Affiliation(s)
- Catherine S Lee
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia 19104, USA
| | | | | | | |
Collapse
|
38
|
Chiao E, Fisher P, Crisponi L, Deiana M, Dragatsis I, Schlessinger D, Pilia G, Efstratiadis A. Overgrowth of a mouse model of the Simpson-Golabi-Behmel syndrome is independent of IGF signaling. Dev Biol 2002; 243:185-206. [PMID: 11846487 DOI: 10.1006/dbio.2001.0554] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The type 1 Simpson-Golabi-Behmel overgrowth syndrome (SGBS1) is caused by loss-of-function mutations of the X-linked GPC3 gene encoding glypican-3, a cell-surface heparan sulfate proteoglycan that apparently plays a negative role in growth control by an unknown mechanism. Mice carrying a Gpc3 gene knockout exhibited several phenotypic features that resemble clinical hallmarks of SGBS1, including somatic overgrowth, renal dysplasia, accessory spleens, polydactyly, and placentomegaly. In Gpc3/DeltaH19 double mutants (lacking GPC3 and also carrying a deletion around the H19 gene region that causes bialellic expression of the closely linked Igf2 gene by imprint relaxation), the Gpc3-null phenotype was exacerbated, while additional SGBS1 features (omphalocele and skeletal defects) were manifested. However, results from a detailed comparative analysis of growth patterns in double mutants lacking GPC3 and also IGF2, IGF1, or the type 1 IGF receptor (IGF1R) provided conclusive genetic evidence inconsistent with the hypothesis that GPC3 acts as a growth suppressor by sequestering or downregulating an IGF ligand. Nevertheless, our data are compatible with a model positing that there is downstream convergence of the independent signaling pathways in which either IGFs or (indirectly) GPC3 participate.
Collapse
Affiliation(s)
- Eric Chiao
- Department of Genetics and Development, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Burbach JP, Luckman SM, Murphy D, Gainer H. Gene regulation in the magnocellular hypothalamo-neurohypophysial system. Physiol Rev 2001; 81:1197-267. [PMID: 11427695 DOI: 10.1152/physrev.2001.81.3.1197] [Citation(s) in RCA: 244] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The hypothalamo-neurohypophysial system (HNS) is the major peptidergic neurosecretory system through which the brain controls peripheral physiology. The hormones vasopressin and oxytocin released from the HNS at the neurohypophysis serve homeostatic functions of water balance and reproduction. From a physiological viewpoint, the core question on the HNS has always been, "How is the rate of hormone production controlled?" Despite a clear description of the physiology, anatomy, cell biology, and biochemistry of the HNS gained over the last 100 years, this question has remained largely unanswered. However, recently, significant progress has been made through studies of gene identity and gene expression in the magnocellular neurons (MCNs) that constitute the HNS. These are keys to mechanisms and events that exist in the HNS. This review is an inventory of what we know about genes expressed in the HNS, about the regulation of their expression in response to physiological stimuli, and about their function. Genes relevant to the central question include receptors and signal transduction components that receive and process the message that the organism is in demand of a neurohypophysial hormone. The key players in gene regulatory events, the transcription factors, deserve special attention. They do not only control rates of hormone production at the level of the gene, but also determine the molecular make-up of the cell essential for appropriate development and physiological functioning. Finally, the HNS neurons are equipped with a machinery to produce and secrete hormones in a regulated manner. With the availability of several gene transfer approaches applicable to the HNS, it is anticipated that new insights will be obtained on how the HNS is able to respond to the physiological demands for its hormones.
Collapse
Affiliation(s)
- J P Burbach
- Rudolf Magnus Institute for Neurosciences, Section of Molecular Neuroscience, Department of Medical Pharmacology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
40
|
Crisponi L, Deiana M, Loi A, Chiappe F, Uda M, Amati P, Bisceglia L, Zelante L, Nagaraja R, Porcu S, Ristaldi MS, Marzella R, Rocchi M, Nicolino M, Lienhardt-Roussie A, Nivelon A, Verloes A, Schlessinger D, Gasparini P, Bonneau D, Cao A, Pilia G. The putative forkhead transcription factor FOXL2 is mutated in blepharophimosis/ptosis/epicanthus inversus syndrome. Nat Genet 2001; 27:159-66. [PMID: 11175783 DOI: 10.1038/84781] [Citation(s) in RCA: 646] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In type I blepharophimosis/ptosis/epicanthus inversus syndrome (BPES), eyelid abnormalities are associated with ovarian failure. Type II BPES shows only the eyelid defects, but both types map to chromosome 3q23. We have positionally cloned a novel, putative winged helix/forkhead transcription factor gene, FOXL2, that is mutated to produce truncated proteins in type I families and larger proteins in type II. Consistent with an involvement in those tissues, FOXL2 is selectively expressed in the mesenchyme of developing mouse eyelids and in adult ovarian follicles; in adult humans, it appears predominantly in the ovary. FOXL2 represents a candidate gene for the polled/intersex syndrome XX sex-reversal goat.
Collapse
Affiliation(s)
- L Crisponi
- Istituto di Ricerca sulle Talassemie ed Anemie Mediterranee CNR, and Dipartimento di Scienze Biomediche e Biotecnologie, Ospedale Regionale per le Microcitemie, Università degli Studi di Cagliari, Cagliari, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kloetzli JM, Fontaine-Glover IA, Brown ER, Kuo M, Labosky PA. The winged helix gene, Foxb1, controls development of mammary glands and regions of the CNS that regulate the milk-ejection reflex. Genesis 2001; 29:60-71. [PMID: 11170346 DOI: 10.1002/1526-968x(200102)29:2<60::aid-gene1006>3.0.co;2-l] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The ability to lactate is a process restricted to mammals and is necessary for the survival of nonhuman mammals. Female mice carrying a null mutation in the winged helix transcription factor Foxb1 (previously Mf3/Fkh5/TWH) have lactation defects on inbred genetic backgrounds. To determine the cellular basis of the Foxb1 lactation defect we have inserted a tau-lacZ lineage marker into the locus to follow the fate of Foxb1 expressing cells. This approach has revealed that Foxb1 is expressed in epithelial cells of developing and adult mammary glands as well as previously described regions of the central nervous system. Mammary glands from C57BL/6 Foxb1-/- mice have incomplete lobuloalveolar development. In addition, the tau-lacZ lineage label was used to determine that the mammillothalamic tract was lost in all Foxb1-/- mice. Finally, morphological defects in the inferior colliculi of the midbrain in Foxb1-/- mice correlate with the inability to lactate, suggesting that the midbrain defect, but not the loss of the mammillothalamic tract, may be responsible for the lactation defect.
Collapse
Affiliation(s)
- J M Kloetzli
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia 19104-6058, USA
| | | | | | | | | |
Collapse
|
42
|
Morini M, Astigiano S, Mora M, Ricotta C, Ferrari N, Mantero S, Levi G, Rossini M, Barbieri O. Hyperplasia and impaired involution in the mammary gland of transgenic mice expressing human FGF4. Oncogene 2000; 19:6007-14. [PMID: 11146552 DOI: 10.1038/sj.onc.1204011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fgf4, a member of the fibroblast growth factor family, is frequently amplified in a variety of human cancers, however, its expression in neoplastic tissues is rarely detectable. This makes uncertain its involvement in tumour aetiology, although several in-vitro studies link Fgf4 overexpression to malignant transformation and metastatization of culture cells. We generated a transgenic mouse model in which the whey acidic protein (WAP) promoter directs expression of human Fgf4 to mammary tissues during late pregnancy and throughout lactation, with the purpose of studying the involvement of this growth factor in mammary tumorigenesis. Expression of the transgene was specifically detected in lobular-alveolar cells of lactating mammary glands that, by histological analysis, displayed hyperplastic areas and a disorganized structure. This was accompanied by an increased number of red blood cells and expression, in alveolar epithelial cells, of the vascular endothelial growth factor, which is absent in wild type controls. The most striking effect caused by FGF4 overexpression was on the remodelling of mammary tissue at the end of lactation. Indeed, transgenic animals showed a delayed involution of the gland due to a dramatic reduction in the overall number of apoptotic cells, which are normally present in the organ after weaning. Nevertheless, none of the animals examined developed neoplastic lesions of the mammary gland even after several pregnancies and at old age. Our work represents the first in-vivo demonstration of the anti-apoptotic and angiogenic properties of FGF4.
Collapse
MESH Headings
- Aging/physiology
- Animals
- Apoptosis
- Blotting, Western
- Cell Transformation, Neoplastic
- Endothelial Growth Factors/genetics
- Endothelial Growth Factors/metabolism
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Fibroblast Growth Factor 4
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/physiology
- Gene Expression Regulation
- Humans
- Hyperplasia/blood
- Hyperplasia/genetics
- Hyperplasia/metabolism
- Hyperplasia/pathology
- Immunohistochemistry
- Lactation
- Lymphokines/genetics
- Lymphokines/metabolism
- Mammary Glands, Animal/abnormalities
- Mammary Glands, Animal/blood supply
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Animal/blood supply
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Transgenic
- Milk Proteins/analysis
- Milk Proteins/biosynthesis
- Milk Proteins/genetics
- Neovascularization, Pathologic
- Phenotype
- Pregnancy
- Promoter Regions, Genetic/genetics
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/physiology
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Transgenes/genetics
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- M Morini
- Unità Transgenici, Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
She P, Shiota M, Shelton KD, Chalkley R, Postic C, Magnuson MA. Phosphoenolpyruvate carboxykinase is necessary for the integration of hepatic energy metabolism. Mol Cell Biol 2000; 20:6508-17. [PMID: 10938127 PMCID: PMC86125 DOI: 10.1128/mcb.20.17.6508-6517.2000] [Citation(s) in RCA: 189] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We used an allelogenic Cre/loxP gene targeting strategy in mice to determine the role of cytosolic phosphoenolpyruvate carboxykinase (PEPCK) in hepatic energy metabolism. Mice that lack this enzyme die within 3 days of birth, while mice with at least a 90% global reduction of PEPCK, or a liver-specific knockout of PEPCK, are viable. Surprisingly, in both cases these animals remain euglycemic after a 24-h fast. However, mice without hepatic PEPCK develop hepatic steatosis after fasting despite up-regulation of a variety of genes encoding free fatty acid-oxidizing enzymes. Also, marked alterations in the expression of hepatic genes involved in energy metabolism occur in the absence of any changes in plasma hormone concentrations. Given that a ninefold elevation of the hepatic malate concentration occurs in the liver-specific PEPCK knockout mice, we suggest that one or more intermediary metabolites may directly regulate expression of the affected genes. Thus, hepatic PEPCK may function more as an integrator of hepatic energy metabolism than as a determinant of gluconeogenesis.
Collapse
Affiliation(s)
- P She
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
44
|
Pérez-Sánchez C, Arias-de-la-Fuente C, Gómez-Ferrería MA, Granadino B, Rey-Campos J. FHX.L and FHX.S, two isoforms of the human fork-head factor FHX (FOXJ2) with differential activity. J Mol Biol 2000; 301:795-806. [PMID: 10966786 DOI: 10.1006/jmbi.2000.3999] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Many biological phenomena are dependent on mechanisms that fine-tune the expression levels of particular genes. This can be achieved by altering the relative activity of a single transcription factor, by post-translational modifications or by interaction with regulatory molecules. An alternative mechanism is based on competition between two or more differently active isoforms of the same transcription factor. We found that FHX, a recently characterized human fork-head transcriptional activator, may show such a mechanism for balancing its activity by expressing two differently sized isoforms, FHX.S and FHX.L, encoded by a single gene located on human chromosome 12. FHX. L and FHX.S showed different transcriptional capacities, the larger form, FHX.L, behaving as the more potent transactivator. A transactivation domain of the acidic type present only in FHX.L would account for this functional difference. The relative concentrations of these two FHX isoforms appear to vary in a number of cell types, a circumstance that may regulate the final activity of this transcription factor.
Collapse
Affiliation(s)
- C Pérez-Sánchez
- Centro de Investigaciones Biológicas, CSIC, Velázquez 144, Madrid, 28006, Spain
| | | | | | | | | |
Collapse
|
45
|
Kume T, Deng K, Hogan BL. Murine forkhead/winged helix genes Foxc1 (Mf1) and Foxc2 (Mfh1) are required for the early organogenesis of the kidney and urinary tract. Development 2000; 127:1387-95. [PMID: 10704385 DOI: 10.1242/dev.127.7.1387] [Citation(s) in RCA: 225] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The murine genes, Foxc1 and Foxc2 (previously, Mf1 and Mfh1), encode forkhead/winged helix transcription factors with virtually identical DNA-binding domains and overlapping expression patterns in various embryonic tissues. Foxc1/Mf1 is disrupted in the mutant, congenital hydrocephalus (Foxc1/Mf1(ch)), which has multiple developmental defects. We show here that, depending on the genetic background, most Foxc1 homozygous mutants are born with abnormalities of the metanephric kidney, including duplex kidneys and double ureters, one of which is a hydroureter. Analysis of embryos reveals that Foxc1 homozygotes have ectopic mesonephric tubules and ectopic anterior ureteric buds. Moreover, expression in the intermediate mesoderm of Glial cell-derived neurotrophic factor (Gdnf), a primary inducer of the ureteric bud, is expanded more anteriorly in Foxc1 homozygous mutants compared with wild type. These findings support the hypothesis of Mackie and Stephens concerning the etiology of duplex kidney and hydroureter in human infants with congenital kidney abnormalities (Mackie, G. G. and Stephens, F. G. (1975) J. Urol. 114, 274–280). Previous studies established that most Foxc1(lacZ)Foxc2(tm1) compound heterozygotes have the same spectrum of cardiovascular defects as single homozygous null mutants, demonstrating interaction between the two genes in the cardiovascular system. Here, we show that most compound heterozygotes have hypoplastic kidneys and a single hydroureter, while all heterozygotes are normal. This provides evidence that the two genes interact in kidney as well as heart development.
Collapse
Affiliation(s)
- T Kume
- Howard Hughes Medical Institute and Department of Cell Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2175, USA
| | | | | |
Collapse
|
46
|
Alvarez-Bolado G, Zhou X, Voss AK, Thomas T, Gruss P. Winged helix transcription factor Foxb1 is essential for access of mammillothalamic axons to the thalamus. Development 2000; 127:1029-38. [PMID: 10662642 DOI: 10.1242/dev.127.5.1029] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Our aim was to study the mechanisms of brain histogenesis. As a model, we have used the role of winged helix transcription factor gene Foxb1 in the emergence of a very specific morphological trait of the diencephalon, the mammillary axonal complex. Foxb1 is expressed in a large hypothalamic neuronal group (the mammillary body), which gives origin to a major axonal bundle with branches to thalamus, tectum and tegmentum. We have generated mice carrying a targeted mutation of Foxb1 plus the tau-lacZ reporter. In these mutants, a subpopulation of dorsal thalamic ventricular cells “thalamic palisade” show abnormal persistence of Foxb1 transcriptional activity; the thalamic branch of the mammillary axonal complex is not able to grow past these cells and enter the thalamus. The other two branches of the mammillary axonal complex (to tectum and tegmentum) are unaffected by the mutation. Most of the neurons that originate the mammillothalamic axons suffer apoptosis after navigational failure. Analysis of chimeric brains with wild-type and Foxb1 mutant cells suggests that correct expression of Foxb1 in the thalamic palisade is sufficient to rescue the normal phenotype. Our results indicate that Foxb1 is essential for diencephalic histogenesis and that it exerts its effects by controlling access to the target by one particular axonal branch.
Collapse
Affiliation(s)
- G Alvarez-Bolado
- Max-Planck Institute of Biophysical Chemistry, Department of Molecular Cell Biology, D-37077 Goettingen, Germany
| | | | | | | | | |
Collapse
|
47
|
Kume T, Deng K, Hogan BL. Minimal phenotype of mice homozygous for a null mutation in the forkhead/winged helix gene, Mf2. Mol Cell Biol 2000; 20:1419-25. [PMID: 10648626 PMCID: PMC85298 DOI: 10.1128/mcb.20.4.1419-1425.2000] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mf2 (mesoderm/mesenchyme forkhead 2) encodes a forkhead/winged helix transcription factor expressed in numerous tissues of the mouse embryo, including paraxial mesoderm, somites, branchial arches, vibrissae, developing central nervous system, and developing kidney. We have generated mice homozygous for a null mutation in the Mf2 gene (Mf2(lacZ)) to examine its role during embryonic development. The lacZ allele also allows monitoring of Mf2 gene expression. Homozygous null mutants are viable and fertile and have no major developmental defects. Some mutants show renal abnormalities, including kidney hypoplasia and hydroureter, but the penetrance of this phenotype is only 40% or lower, depending on the genetic background. These data suggest that Mf2 can play a unique role in kidney development, but there is functional redundancy in this organ and other tissues with other forkhead/winged helix genes.
Collapse
Affiliation(s)
- T Kume
- Howard Hughes Medical Institute, Nashville, Tennessee 37232-2175, USA
| | | | | |
Collapse
|
48
|
Daubas P, Tajbakhsh S, Hadchouel J, Primig M, Buckingham M. Myf5 is a novel early axonal marker in the mouse brain and is subjected to post-transcriptional regulation in neurons. Development 2000; 127:319-31. [PMID: 10603349 DOI: 10.1242/dev.127.2.319] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Myf5 is a key basic Helix-Loop-Helix transcription factor capable of converting many non-muscle cells into muscle. Together with MyoD it is essential for initiating the skeletal muscle programme in the embryo. We previously identified unexpected restricted domains of Myf5 transcription in the embryonic mouse brain, first revealed by Myf5-nlacZ(+/)(−) embryos (Tajbakhsh, S. and Buckingham, M. (1995) Development 121, 4077–4083). We have now further characterized these Myf5 expressing neurons. Retrograde labeling with diI, and the use of a transgenic mouse line expressing lacZ under the control of Myf5 regulatory sequences, show that Myf5 transcription provides a novel axonal marker of the medial longitudinal fasciculus (mlf) and the mammillotegmental tract (mtt), the earliest longitudinal tracts to be established in the embryonic mouse brain. Tracts projecting caudally from the developing olfactory system are also labelled. nlacZ and lacZ expression persist in the adult brain, in a few ventral domains such as the mammillary bodies of the hypothalamus and the interpeduncular nucleus, potentially derived from the embryonic structures where the Myf5 gene is transcribed. To investigate the role of Myf5 in the brain, we monitored Myf5 protein accumulation by immunofluorescence and immunoblotting in neurons transcribing the gene. Although Myf5 was detected in muscle myotomal cells, it was absent in neurons. This would account for the lack of myogenic conversion in brain structures and the absence of a neural phenotype in homozygous null mutants. RT-PCR experiments show that the splicing of Myf5 primary transcripts occurs correctly in neurons, suggesting that the lack of Myf5 protein accumulation is due to regulation at the level of mRNA translation or protein stability. In the embryonic neuroepithelium, Myf5 is transcribed in differentiated neurons after the expression of neural basic Helix-Loop-Helix transcription factors. The signalling molecules Wnt1 and Sonic hedgehog, implicated in the activation of Myf5 in myogenic progenitor cells in the somite, are also produced in the viscinity of the Myf5 expression domain in the mesencephalon. We show that cells expressing Wnt1 can activate neuronal Myf5-nlacZ gene expression in dissected head explants isolated from E9.5 embryos. Furthermore, the gene encoding the basic Helix-Loop-Helix transcription factor mSim1 is expressed in adjacent cells in both the somite and the brain, suggesting that signalling molecules necessary for the activation of mSim1 as well as Myf5 are present at these different sites in the embryo. This phenomenon may be widespread and it remains to be seen how many other potentially potent regulatory genes, in addition to Myf5, when activated do not accumulate protein at inappropriate sites in the embryo.
Collapse
Affiliation(s)
- P Daubas
- CNRS URA 1947, Départment de Biologie Moléculaire, Institut Pasteur, 75724 Paris Cedex 15, France.
| | | | | | | | | |
Collapse
|
49
|
Lewis MT. Homeobox genes in mammary gland development and neoplasia. Breast Cancer Res 2000; 2:158-69. [PMID: 11250705 PMCID: PMC138770 DOI: 10.1186/bcr49] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/1999] [Revised: 01/25/2000] [Accepted: 02/04/2000] [Indexed: 02/03/2023] Open
Abstract
Both normal development and neoplastic progression involve cellular transitions from one physiological state to another. Whereas much is being discovered about signal transduction networks involved in regulating these transitions, little progress has been made in identifying the higher order genetic determinants that establish and maintain mammary cell identity and dictate cell type-specific responses to mammotropic signals. Homeobox genes are a large superfamily of genes whose members function in establishing and maintaining cell fate and cell identity throughout embryonic development. Recent genetic and expression analyses strongly suggest that homeobox genes may perform similar functions at specific developmental transition points in the mammary gland. These analyses also suggest that homeobox genes may play a contributory or causal role in breast cancer.
Collapse
Affiliation(s)
- M T Lewis
- Department of Physiology and Biophysics, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| |
Collapse
|
50
|
Abstract
The spectrum of neuronal migration disorders (NMD) in humans encompasses developmental brain defects with a range of clinical and pathological features. A simple classification distinguishes agyria/pachygyria, heterotopia, polymicrogyria and cortical dysplasia as distinct clinico-pathological entities. Many of these conditions are associated with intractable epilepsy. When considering the pathogenesis of NMD, a critical developmental process is the migration of neuroblasts along the processes of radial glia during the formation of the layered structure of the cerebral cortex. In addition, faulty cytodifferentiation and programmed cell death play important roles in the generation of dysplasias and heterotopias respectively. A number of genes have been identified that participate in the regulation of neuronal migration. Mouse models, in which these genes are mutated, provide insight into the developmental pathways that underlie normal and abnormal neuronal migration.
Collapse
Affiliation(s)
- A J Copp
- Neural Development Unit, Institute of Child Health, University College London, UK.
| | | |
Collapse
|