51
|
Wolf L, Harrison W, Huang J, Xie Q, Xiao N, Sun J, Kong L, Lachke SA, Kuracha MR, Govindarajan V, Brindle PK, Ashery-Padan R, Beebe DC, Overbeek PA, Cvekl A. Histone posttranslational modifications and cell fate determination: lens induction requires the lysine acetyltransferases CBP and p300. Nucleic Acids Res 2013; 41:10199-214. [PMID: 24038357 PMCID: PMC3905850 DOI: 10.1093/nar/gkt824] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/15/2013] [Accepted: 08/20/2013] [Indexed: 12/15/2022] Open
Abstract
Lens induction is a classical embryologic model to study cell fate determination. It has been proposed earlier that specific changes in core histone modifications accompany the process of cell fate specification and determination. The lysine acetyltransferases CBP and p300 function as principal enzymes that modify core histones to facilitate specific gene expression. Herein, we performed conditional inactivation of both CBP and p300 in the ectodermal cells that give rise to the lens placode. Inactivation of both CBP and p300 resulted in the dramatic discontinuation of all aspects of lens specification and organogenesis, resulting in aphakia. The CBP/p300(-/-) ectodermal cells are viable and not prone to apoptosis. These cells showed reduced expression of Six3 and Sox2, while expression of Pax6 was not upregulated, indicating discontinuation of lens induction. Consequently, expression of αB- and αA-crystallins was not initiated. Mutant ectoderm exhibited markedly reduced levels of histone H3 K18 and K27 acetylation, subtly increased H3 K27me3 and unaltered overall levels of H3 K9ac and H3 K4me3. Our data demonstrate that CBP and p300 are required to establish lens cell-type identity during lens induction, and suggest that posttranslational histone modifications are integral to normal cell fate determination in the mammalian lens.
Collapse
Affiliation(s)
- Louise Wolf
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Wilbur Harrison
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Jie Huang
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Qing Xie
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Ningna Xiao
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Jian Sun
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Lingkun Kong
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Salil A. Lachke
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Murali R. Kuracha
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Venkatesh Govindarajan
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Paul K. Brindle
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Ruth Ashery-Padan
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - David C. Beebe
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Paul A. Overbeek
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| | - Ales Cvekl
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY10461, USA, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA, Departments of Ophthalmology and Visual Sciences, Washington University Saint Louis, Saint Louis, MO 63110, USA, Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA, Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA, Department of Surgery, Creighton University, Omaha, NE 68178, USA, Department of Biochemistry, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA and Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Israel 69978
| |
Collapse
|
52
|
Antosova B, Smolikova J, Borkovcova R, Strnad H, Lachova J, Machon O, Kozmik Z. Ectopic activation of Wnt/β-catenin signaling in lens fiber cells results in cataract formation and aberrant fiber cell differentiation. PLoS One 2013; 8:e78279. [PMID: 24205179 PMCID: PMC3813504 DOI: 10.1371/journal.pone.0078279] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 09/09/2013] [Indexed: 12/04/2022] Open
Abstract
The Wnt/β-catenin signaling pathway controls many processes during development, including cell proliferation, cell differentiation and tissue homeostasis, and its aberrant regulation has been linked to various pathologies. In this study we investigated the effect of ectopic activation of Wnt/β-catenin signaling during lens fiber cell differentiation. To activate Wnt/β-catenin signaling in lens fiber cells, the transgenic mouse referred to as αA-CLEF was generated, in which the transactivation domain of β-catenin was fused to the DNA-binding protein LEF1, and expression of the transgene was controlled by αA-crystallin promoter. Constitutive activation of Wnt/β-catenin signaling in lens fiber cells of αA-CLEF mice resulted in abnormal and delayed fiber cell differentiation. Moreover, adult αA-CLEF mice developed cataract, microphthalmia and manifested downregulated levels of γ-crystallins in lenses. We provide evidence of aberrant expression of cell cycle regulators in embryonic lenses of αA-CLEF transgenic mice resulting in the delay in cell cycle exit and in the shift of fiber cell differentiation to the central fiber cell compartment. Our results indicate that precise regulation of the Wnt/β-catenin signaling activity during later stages of lens development is essential for proper lens fiber cell differentiation and lens transparency.
Collapse
Affiliation(s)
- Barbora Antosova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- Department of Genetics and Microbiology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Jana Smolikova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Romana Borkovcova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Hynek Strnad
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jitka Lachova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Ondrej Machon
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Zbynek Kozmik
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
53
|
Sugiyama Y, Shelley EJ, Wen L, Stump RJW, Shimono A, Lovicu FJ, McAvoy JW. Sfrp1 and Sfrp2 are not involved in Wnt/β-catenin signal silencing during lens induction but are required for maintenance of Wnt/β-catenin signaling in lens epithelial cells. Dev Biol 2013; 384:181-93. [PMID: 24140542 DOI: 10.1016/j.ydbio.2013.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 10/03/2013] [Accepted: 10/07/2013] [Indexed: 02/07/2023]
Abstract
During eye lens development, regulation of Wnt/β-catenin signaling is critical for two major processes: initially it must be silent in the lens placode for lens development to proceed, but subsequently it is required for maintenance of the lens epithelium. It is not known how these different phases of Wnt/β-catenin activity/inactivity are regulated. Secreted frizzled related protein-2 (Sfrp2), a putative Wnt-Fz antagonist, is expressed in lens placode and in lens epithelial cells and has been put forward as a candidate for regional Wnt/β-catenin pathway regulation. Here we show its closely-related isoform, Sfrp1, has a complimentary pattern of expression in the lens, being absent from the placode and epithelium but expressed in the fibers. As mice with single knockouts of Sfrp1 or Sfrp2 had no defects in lens formation, we examined lenses of Sfrp1 and Sfrp2 double knockout (DKO) mice and showed that they formed lens placode and subsequent lens structures. Consistent with this we did not observe ectopic TCF/Lef activity in lens placode of DKOs. This indicates that Sfrp1 and Sfrp2 individually, or together, do not constitute the putative negative regulator that blocks Wnt/β-catenin signaling during lens induction. In contrast, Sfrp1 and Sfrp2 appear to have a positive regulatory function because Wnt/β-catenin signaling in lens epithelial cells was reduced in Sfrp1 and Sfrp2 DKO mice. Lenses that formed in DKO mice were smaller than controls and exhibited a deficient epithelium. Thus Sfrps play a role in lens development, at least in part, by regulating aspects of Wnt/β-catenin signaling in lens epithelial cells.
Collapse
Affiliation(s)
- Yuki Sugiyama
- Save Sight Institute, The University of Sydney, Australia
| | | | | | | | | | | | | |
Collapse
|
54
|
Cancian L, Hansen A, Boshoff C. Cellular origin of Kaposi's sarcoma and Kaposi's sarcoma-associated herpesvirus-induced cell reprogramming. Trends Cell Biol 2013; 23:421-32. [PMID: 23685018 DOI: 10.1016/j.tcb.2013.04.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 04/02/2013] [Accepted: 04/03/2013] [Indexed: 01/05/2023]
Abstract
Kaposi's sarcoma (KS) is the most common malignancy in untreated HIV patients. KS is characterised by abnormal neoangiogenesis, inflammation, and proliferation of tumour cells [KS spindle cells (SCs)]. Kaposi's sarcoma-associated herpesvirus (KSHV) is the aetiological agent of KS. KS SCs are the predominant KSHV-infected cells in KS lesions. In this review, we report advances in understanding of the cellular origin of the KS SC, a contentious topic in KSHV research. KS SCs are now known to be of endothelial cell (EC) origin, phenotypically most similar to lymphatic ECs (LECs), but poorly differentiated. We focus on recent insights into KSHV's ability to exploit the normal differentiation pathway and intrinsic plasticity of ECs, through manipulation of EC-specific transcriptional regulators [i.e., prospero homeobox 1 (PROX1) and MAF] and discuss how this may contribute to viral persistence and KS sarcomagenesis.
Collapse
Affiliation(s)
- Laila Cancian
- UCL Cancer Institute, 72 Huntley Street, University College London, London WC1E 6BT, UK
| | | | | |
Collapse
|
55
|
Sousounis K, Tsonis PA. Patterns of gene expression in microarrays and expressed sequence tags from normal and cataractous lenses. Hum Genomics 2012; 6:14. [PMID: 23244575 PMCID: PMC3563465 DOI: 10.1186/1479-7364-6-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 05/14/2012] [Indexed: 11/30/2022] Open
Abstract
In this contribution, we have examined the patterns of gene expression in normal and cataractous lenses as presented in five different papers using microarrays and expressed sequence tags. The purpose was to evaluate unique and common patterns of gene expression during development, aging and cataracts.
Collapse
Affiliation(s)
- Konstantinos Sousounis
- Department of Biology and Center for Tissue Regeneration and Engineering, University of Dayton, Dayton, OH 45469-2320, USA
| | | |
Collapse
|
56
|
de Thonel A, Le Mouël A, Mezger V. Transcriptional regulation of small HSP-HSF1 and beyond. Int J Biochem Cell Biol 2012; 44:1593-612. [PMID: 22750029 DOI: 10.1016/j.biocel.2012.06.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 06/07/2012] [Accepted: 06/08/2012] [Indexed: 12/16/2022]
Abstract
The members of the small heat shock protein (sHSP) family are molecular chaperones that play major roles in development, stress responses, and diseases, and have been envisioned as targets for therapy, particularly in cancer. The molecular mechanisms that regulate their transcription, in normal, stress, or pathological conditions, are characterized by extreme complexity and subtlety. Although historically linked to the heat shock transcription factors (HSFs), the stress-induced or developmental expression of the diverse members, including HSPB1/Hsp27/Hsp25, αA-crystallin/HSPB4, and αB-crystallin/HSPB5, relies on the combinatory effects of many transcription factors. Coupled with remarkably different cis-element architectures in the sHsp regulatory regions, they confer to each member its developmental expression or stress-inducibility. For example, multiple regulatory pathways coordinate the spatio-temporal expression of mouse αA-, αB-crystallin, and Hsp25 genes during lens development, through the action of master genes, like the large Maf family proteins and Pax6, but also HSF4. The inducibility of Hsp27 and αB-crystallin transcription by various stresses is exerted by HSF-dependent mechanisms, by which concomitant induction of Hsp27 and αB-crystallin expression is observed. In contrast, HSF-independent pathways can lead to αB-crystallin expression, but not to Hsp27 induction. Not surprisingly, deregulation of the expression of sHSP is associated with various pathologies, including cancer, neurodegenerative, or cardiac diseases. However, many questions remain to be addressed, and further elucidation of the developmental mechanisms of sHsp gene transcription might help to unravel the tissue- and stage-specific functions of this fascinating class of proteins, which might prove to be crucial for future therapeutic strategies. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.
Collapse
|
57
|
Ho DWH, Yap MKH, Ng PW, Fung WY, Yip SP. Association of high myopia with crystallin beta A4 (CRYBA4) gene polymorphisms in the linkage-identified MYP6 locus. PLoS One 2012; 7:e40238. [PMID: 22792142 PMCID: PMC3389832 DOI: 10.1371/journal.pone.0040238] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 06/03/2012] [Indexed: 12/03/2022] Open
Abstract
Background Myopia is the most common ocular disorder worldwide and imposes tremendous burden on the society. It is a complex disease. The MYP6 locus at 22 q12 is of particular interest because many studies have detected linkage signals at this interval. The MYP6 locus is likely to contain susceptibility gene(s) for myopia, but none has yet been identified. Methodology/Principal Findings Two independent subject groups of southern Chinese in Hong Kong participated in the study an initial study using a discovery sample set of 342 cases and 342 controls, and a follow-up study using a replication sample set of 316 cases and 313 controls. Cases with high myopia were defined by spherical equivalent ≤ -8 dioptres and emmetropic controls by spherical equivalent within ±1.00 dioptre for both eyes. Manual candidate gene selection from the MYP6 locus was supported by objective in silico prioritization. DNA samples of discovery sample set were genotyped for 178 tagging single nucleotide polymorphisms (SNPs) from 26 genes. For replication, 25 SNPs (tagging or located at predicted transcription factor or microRNA binding sites) from 4 genes were subsequently examined using the replication sample set. Fisher P value was calculated for all SNPs and overall association results were summarized by meta-analysis. Based on initial and replication studies, rs2009066 located in the crystallin beta A4 (CRYBA4) gene was identified to be the most significantly associated with high myopia (initial study: P = 0.02; replication study: P = 1.88e-4; meta-analysis: P = 1.54e-5) among all the SNPs tested. The association result survived correction for multiple comparisons. Under the allelic genetic model for the combined sample set, the odds ratio of the minor allele G was 1.41 (95% confidence intervals, 1.21-1.64). Conclusions/Significance A novel susceptibility gene (CRYBA4) was discovered for high myopia. Our study also signified the potential importance of appropriate gene prioritization in candidate selection.
Collapse
Affiliation(s)
- Daniel W. H. Ho
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Maurice K. H. Yap
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Po Wah Ng
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Wai Yan Fung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
- * E-mail:
| |
Collapse
|
58
|
Wende H, Lechner SG, Cheret C, Bourane S, Kolanczyk ME, Pattyn A, Reuter K, Munier FL, Carroll P, Lewin GR, Birchmeier C. The transcription factor c-Maf controls touch receptor development and function. Science 2012; 335:1373-6. [PMID: 22345400 DOI: 10.1126/science.1214314] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The sense of touch relies on detection of mechanical stimuli by specialized mechanosensory neurons. The scarcity of molecular data has made it difficult to analyze development of mechanoreceptors and to define the basis of their diversity and function. We show that the transcription factor c-Maf/c-MAF is crucial for mechanosensory function in mice and humans. The development and function of several rapidly adapting mechanoreceptor types are disrupted in c-Maf mutant mice. In particular, Pacinian corpuscles, a type of mechanoreceptor specialized to detect high-frequency vibrations, are severely atrophied. In line with this, sensitivity to high-frequency vibration is reduced in humans carrying a dominant mutation in the c-MAF gene. Thus, our work identifies a key transcription factor specifying development and function of mechanoreceptors and their end organs.
Collapse
Affiliation(s)
- Hagen Wende
- Developmental Biology, Max Delbrück Center (MDC) for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Ogino H, Ochi H, Reza HM, Yasuda K. Transcription factors involved in lens development from the preplacodal ectoderm. Dev Biol 2012; 363:333-47. [PMID: 22269169 DOI: 10.1016/j.ydbio.2012.01.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 12/14/2011] [Accepted: 01/09/2012] [Indexed: 12/14/2022]
Abstract
Lens development is a stepwise process accompanied by the sequential activation of transcription factors. Transcription factor genes can be classified into three groups according to their functions: the first group comprises preplacodal genes, which are implicated in the formation of the preplacodal ectoderm that serves as a common primordium for cranial sensory tissues, including the lens. The second group comprises lens-specification genes, which establish the lens-field within the preplacodal ectoderm. The third group comprises lens-differentiation genes, which promote lens morphogenesis after the optic vesicle makes contact with the presumptive lens ectoderm. Analyses of the regulatory interactions between these genes have provided an overview of lens development, highlighting crucial roles for positive cross-regulation in fate specification and for feed-forward regulation in the execution of terminal differentiation. This overview also sheds light upon the mechanisms of how preplacodal gene activities lead to the activation of genes involved in lens-specification.
Collapse
Affiliation(s)
- Hajime Ogino
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma, Nara, 630-0192, Japan.
| | | | | | | |
Collapse
|
60
|
Xie Q, Cvekl A. The orchestration of mammalian tissue morphogenesis through a series of coherent feed-forward loops. J Biol Chem 2011; 286:43259-71. [PMID: 21998302 DOI: 10.1074/jbc.m111.264580] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Tissue morphogenesis requires intricate temporal and spatial control of gene expression that is executed through specific gene regulatory networks (GRNs). GRNs are comprised from individual subcircuits of different levels of complexity. An important question is to elucidate the mutual relationship between those genes encoding DNA-binding factors that trigger the subcircuit with those that play major "later" roles during terminal differentiation via expression of specific genes that constitute the phenotype of individual tissues. The ocular lens is a classical model system to study tissue morphogenesis. Pax6 is essential for both lens placode formation and subsequent stages of lens morphogenesis, whereas c-Maf controls terminal differentiation of lens fibers, including regulation of crystallins, key lens structural proteins required for its transparency and refraction. Here, we show that Pax6 directly regulates c-Maf expression during lens development. A 1.3-kb c-Maf promoter with a 1.6-kb upstream enhancer (CR1) recapitulated the endogenous c-Maf expression pattern in lens and retinal pigmented epithelium. ChIP assays revealed binding of Pax6 and c-Maf to multiple regions of the c-Maf locus in lens chromatin. To predict functional Pax6-binding sites, nine novel variants of Pax6 DNA-binding motifs were identified and characterized. Two of these motifs predicted a pair of Pax6-binding sites in the CR1. Mutagenesis of these Pax6-binding sites inactivated transgenic expression in the lens but not in retinal pigmented epithelium. These data establish a novel regulatory role for Pax6 during lens development, link together the Pax6/c-Maf/crystallin regulatory network, and suggest a novel type of GRN subcircuit that controls a major part of embryonic lens development.
Collapse
Affiliation(s)
- Qing Xie
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
61
|
The lens in focus: a comparison of lens development in Drosophila and vertebrates. Mol Genet Genomics 2011; 286:189-213. [PMID: 21877135 DOI: 10.1007/s00438-011-0643-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/04/2011] [Indexed: 12/24/2022]
Abstract
The evolution of the eye has been a major subject of study dating back centuries. The advent of molecular genetics offered the surprising finding that morphologically distinct eyes rely on conserved regulatory gene networks for their formation. While many of these advances often stemmed from studies of the compound eye of the fruit fly, Drosophila melanogaster, and later translated to discoveries in vertebrate systems, studies on vertebrate lens development far outnumber those in Drosophila. This may be largely historical, since Spemann and Mangold's paradigm of tissue induction was discovered in the amphibian lens. Recent studies on lens development in Drosophila have begun to define molecular commonalities with the vertebrate lens. Here, we provide an overview of Drosophila lens development, discussing intrinsic and extrinsic factors controlling lens cell specification and differentiation. We then summarize key morphological and molecular events in vertebrate lens development, emphasizing regulatory factors and networks strongly associated with both systems. Finally, we provide a comparative analysis that highlights areas of research that would help further clarify the degree of conservation between the formation of dioptric systems in invertebrates and vertebrates.
Collapse
|
62
|
c-Maf plays a crucial role for the definitive erythropoiesis that accompanies erythroblastic island formation in the fetal liver. Blood 2011; 118:1374-85. [PMID: 21628412 DOI: 10.1182/blood-2010-08-300400] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
c-Maf is one of the large Maf (musculoaponeurotic fibrosarcoma) transcription factors that belong to the activated protein-1 super family of basic leucine zipper proteins. Despite its overexpression in hematologic malignancies, the physiologic roles c-Maf plays in normal hematopoiesis have been largely unexplored. On a C57BL/6J background, c-Maf(-/-) embryos succumbed from severe erythropenia between embryonic day (E) 15 and E18. Flow cytometric analysis of fetal liver cells showed that the mature erythroid compartments were significantly reduced in c-Maf(-/-) embryos compared with c-Maf(+/+) littermates. Interestingly, the CFU assay indicated there was no significant difference between c-Maf(+/+) and c-Maf(-/-) fetal liver cells in erythroid colony counts. This result indicated that impaired definitive erythropoiesis in c-Maf(-/-) embryos is because of a non-cell-autonomous effect, suggesting a defective erythropoietic microenvironment in the fetal liver. As expected, the number of erythroblasts surrounding the macrophages in erythroblastic islands was significantly reduced in c-Maf(-/-) embryos. Moreover, decreased expression of VCAM-1 was observed in c-Maf(-/-) fetal liver macrophages. In conclusion, these results strongly suggest that c-Maf is crucial for definitive erythropoiesis in fetal liver, playing an important role in macrophages that constitute erythroblastic islands.
Collapse
|
63
|
Wiley LA, Rajagopal R, Dattilo LK, Beebe DC. The tumor suppressor gene Trp53 protects the mouse lens against posterior subcapsular cataracts and the BMP receptor Acvr1 acts as a tumor suppressor in the lens. Dis Model Mech 2011; 4:484-95. [PMID: 21504908 PMCID: PMC3124053 DOI: 10.1242/dmm.006593] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We previously found that lenses lacking the Acvr1 gene, which encodes a bone morphogenetic protein (BMP) receptor, had abnormal proliferation and cell death in epithelial and cortical fiber cells. We tested whether the tumor suppressor protein p53 (encoded by Trp53) affected this phenotype. Acvr1 conditional knockout (Acvr1CKO) mouse fiber cells had increased numbers of nuclei that stained for p53 phosphorylated on serine 15, an indicator of p53 stabilization and activation. Deletion of Trp53 rescued the Acvr1CKO cell death phenotype in embryos and reduced Acvr1-dependent apoptosis in postnatal lenses. However, deletion of Trp53 alone increased the number of fiber cells that failed to withdraw from the cell cycle. Trp53CKO and Acvr1;Trp53DCKO (double conditional knockout), but not Acvr1CKO, lenses developed abnormal collections of cells at the posterior of the lens that resembled posterior subcapsular cataracts. Cells from human posterior subcapsular cataracts had morphological and molecular characteristics similar to the cells at the posterior of mouse lenses lacking Trp53. In Trp53CKO lenses, cells in the posterior plaques did not proliferate but, in Acvr1;Trp53DCKO lenses, many cells in the posterior plaques continued to proliferate, eventually forming vascularized tumor-like masses at the posterior of the lens. We conclude that p53 protects the lens against posterior subcapsular cataract formation by suppressing the proliferation of fiber cells and promoting the death of any fiber cells that enter the cell cycle. Acvr1 acts as a tumor suppressor in the lens. Enhancing p53 function in the lens could contribute to the prevention of steroid- and radiation-induced posterior subcapsular cataracts.
Collapse
Affiliation(s)
- Luke A Wiley
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO 63110, USA
| | | | | | | |
Collapse
|
64
|
Lachke SA, Maas RL. Building the developmental oculome: systems biology in vertebrate eye development and disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2011; 2:305-323. [PMID: 20836031 DOI: 10.1002/wsbm.59] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The vertebrate eye is a sophisticated multicomponent organ that has been actively studied for over a century, resulting in the identification of the major embryonic and molecular events involved in its complex developmental program. Data gathered so far provides sufficient information to construct a rudimentary network of the various signaling molecules, transcription factors, and their targets for several key stages of this process. With the advent of genomic technologies, there has been a rapid expansion in our ability to collect and process biological information, and the use of systems-level approaches to study specific aspects of vertebrate eye development has already commenced. This is beginning to result in the definition of the dynamic developmental networks that operate in ocular tissues, and the interactions of such networks between coordinately developing ocular tissues. Such an integrative understanding of the eye by a comprehensive systems-level analysis can be termed the 'oculome', and that of serial developmental stages of the eye as it transits from its initiation to a fully formed functional organ represents the 'developmental oculome'. Construction of the developmental oculome will allow novel mechanistic insights that are essential for organ regeneration-based therapeutic applications, and the generation of computational models for eye disease states to predict the effects of drugs. This review discusses our present understanding of two of the individual components of the developing vertebrate eye--the lens and retina--at both the molecular and systems levels, and outlines the directions and tools required for construction of the developmental oculome.
Collapse
Affiliation(s)
- Salil A Lachke
- Division of Genetics, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Richard L Maas
- Division of Genetics, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
65
|
Morito N, Yoh K, Maeda A, Nakano T, Fujita A, Kusakabe M, Hamada M, Kudo T, Yamagata K, Takahashi S. A Novel Transgenic Mouse Model of the Human Multiple Myeloma Chromosomal Translocation t(14;16)(q32;q23). Cancer Res 2011; 71:339-48. [DOI: 10.1158/0008-5472.can-10-1057] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Multiple myeloma (MM) is a currently incurable neoplasm of terminally differentiated B cells. The translocation and/or overexpression of c-MAF have been observed in human MM. Although c-MAF might function as an oncogene in human MM, there has been no report thus far describing the direct induction of MM by c-MAF overexpression in vivo. In this study, we have generated transgenic (TG) mice that express c-Maf specifically in the B-cell compartment. Aged c-Maf TG mice developed B-cell lymphomas with some clinical features that resembled those of MM, namely, plasma cell expansion and hyperglobulinemia. Quantitative RT-PCR analysis demonstrated that Ccnd2 and Itgb7, which are known target genes of c-Maf, were highly expressed in the lymphoma cells. This novel TG mouse model of the human MM t(14;16)(q32;q23) chromosomal translocation should serve to provide new insight into the role of c-MAF in tumorigenesis. Cancer Res; 71(2); 339–48. ©2011 AACR.
Collapse
Affiliation(s)
- Naoki Morito
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Keigyou Yoh
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Atsuko Maeda
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takako Nakano
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akiko Fujita
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Manabu Kusakabe
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Michito Hamada
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takashi Kudo
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kunihiro Yamagata
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Authors' Affiliations: Departments of 1Nephrology and 2Anatomy and Embryology, Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
66
|
Abstract
Chondrocyte differentiation in the growth plate is an important process for the longitudinal growth of endochondral bones. Sox9 and Runx2 are the most often-studied transcriptional regulators of the chondrocyte differentiation process, but the importance of additional factors is also becoming apparent. Mafs are a subfamily of the basic ZIP (bZIP) transcription factor superfamily, which act as key regulators of tissue-specific gene expression and terminal differentiation in many tissues. There is increasing evidence that c-Maf and its splicing variant Lc-Maf play a role in chondrocyte differentiation in a temporal-spatial manner. This review summarizes the functions of c-Maf in chondrocyte differentiation and discusses the possible role of c-Maf in osteoarthritis progression.
Collapse
Affiliation(s)
| | | | - Dominik R. Haudenschild
- Dominik R. Haudenschild, Department of Orthopaedic Surgery, Division of Orthopaedic Research, University of California Davis Medical Center, 4635 Second Street, Sacramento, CA 95817, USA
| |
Collapse
|
67
|
Abstract
Lens regeneration among vertebrates is basically restricted to some amphibians. The most notable cases are the ones that occur in premetamorphic frogs and in adult newts. Frogs and newts regenerate their lens in very different ways. In frogs the lens is regenerated by transdifferentiation of the cornea and is limited only to a time before metamorphosis. On the other hand, regeneration in newts is mediated by transdifferentiation of the pigment epithelial cells of the dorsal iris and is possible in adult animals as well. Thus, the study of both systems could provide important information about the process. Molecular tools have been developed in frogs and recently also in newts. Thus, the process has been studied at the molecular and cellular levels. A synthesis describing both systems was long due. In this review we describe the process in both Xenopus and the newt. The known molecular mechanisms are described and compared.
Collapse
Affiliation(s)
- Jonathan J Henry
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61801, USA.
| | | |
Collapse
|
68
|
Kanai K, Reza HM, Kamitani A, Hamazaki Y, Han SI, Yasuda K, Kataoka K. SUMOylation negatively regulates transcriptional and oncogenic activities of MafA. Genes Cells 2010; 15:971-82. [PMID: 20718938 DOI: 10.1111/j.1365-2443.2010.01431.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dysregulated expression of Maf proteins (namely c-Maf, MafA and MafB) leads to multiple myeloma in humans and oncogenic transformation of chicken embryonic fibroblasts. Maf proteins are transcriptional activators of tissue-specific gene expression and regulators of cell differentiation. For example, MafA is a critical regulator of crystallin genes and the lens differentiation program in chickens. In mammals, MafA is essential for the development of mature insulin-producing beta-cells of pancreas. It has been shown that MafA protein stability is regulated by phosphorylations at multiple serine and threonine residues. Here, we report that Maf proteins are also post-translationally modified by small ubiquitin-like modifier (SUMO) proteins at a conserved lysine residue in the amino-terminal transactivator domain. A SUMOylation-deficient mutant of MafA (K32R) was more potent than wild-type MafA in transactivating luciferase reporter construct driven by alphaA-crystallin or insulin gene promoter. In ovo electroporation into developing chicken embryo showed that the K32R mutant induced ectopic delta-crystallin gene expression more efficiently than the wild-type MafA. We also demonstrated that the K32R mutant had enhanced ability to induce colony formation of a chicken fibroblast cell line DF-1. Therefore, SUMOylation is a functional post-translational modification of MafA that negatively regulates its transcriptional and transforming activities.
Collapse
Affiliation(s)
- Kenichi Kanai
- Nara Institute of Science and Technology, Takayama-cho, Ikoma, Japan
| | | | | | | | | | | | | |
Collapse
|
69
|
Wang WL, Li Q, Xu J, Cvekl A. Lens fiber cell differentiation and denucleation are disrupted through expression of the N-terminal nuclear receptor box of NCOA6 and result in p53-dependent and p53-independent apoptosis. Mol Biol Cell 2010; 21:2453-68. [PMID: 20484573 PMCID: PMC2903674 DOI: 10.1091/mbc.e09-12-1031] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nuclear receptor coactivator 6 (NCOA6) is a multifunctional protein implicated in embryonic development, cell survival, and homeostasis. An 81-amino acid fragment, dnNCOA6, containing the N-terminal nuclear receptor box (LXXLL motif) of NCOA6, acts as a dominant-negative (dn) inhibitor of NCOA6. Here, we expressed dnNCOA6 in postmitotic transgenic mouse lens fiber cells. The transgenic lenses showed reduced growth; a wide spectrum of lens fiber cell differentiation defects, including reduced expression of gamma-crystallins; and cataract formation. Those lens fiber cells entered an alternate proapoptotic pathway, and the denucleation (karyolysis) process was stalled. Activation of caspase-3 at embryonic day (E)13.5 was followed by double-strand breaks (DSBs) formation monitored via a biomarker, gamma-H2AX. Intense terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) signals were found at E16.5. Thus, a window of approximately 72 h between these events suggested prolonged though incomplete apoptosis in the lens fiber cell compartment that preserved nuclei in its cells. Genetic experiments showed that the apoptotic-like processes in the transgenic lens were both p53-dependent and p53-independent. Lens-specific deletion of Ncoa6 also resulted in disrupted lens fiber cell differentiation. Our data demonstrate a cell-autonomous role of Ncoa6 in lens fiber cell differentiation and suggest novel insights into the process of lens fiber cell denucleation and apoptosis.
Collapse
Affiliation(s)
- Wei-Lin Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
70
|
Hansen A, Henderson S, Lagos D, Nikitenko L, Coulter E, Roberts S, Gratrix F, Plaisance K, Renne R, Bower M, Kellam P, Boshoff C. KSHV-encoded miRNAs target MAF to induce endothelial cell reprogramming. Genes Dev 2010; 24:195-205. [PMID: 20080955 DOI: 10.1101/gad.553410] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Kaposi sarcoma herpesvirus (KSHV) induces transcriptional reprogramming of endothelial cells. In particular, KSHV-infected lymphatic endothelial cells (LECs) show an up-regulation of genes associated with blood vessel endothelial cells (BECs). Consequently, KSHV-infected tumor cells in Kaposi sarcoma are poorly differentiated endothelial cells, expressing markers of both LECs and BECs. MicroRNAs (miRNAs) are short noncoding RNA molecules that act post-transcriptionally to negatively regulate gene expression. Here we validate expression of the KSHV-encoded miRNAs in Kaposi sarcoma lesions and demonstrate that these miRNAs contribute to viral-induced reprogramming by silencing the cellular transcription factor MAF (musculoaponeurotic fibrosarcoma oncogene homolog). MAF is expressed in LECs but not in BECs. We identify a novel role for MAF as a transcriptional repressor, preventing expression of BEC-specific genes, thereby maintaining the differentiation status of LECs. These findings demonstrate that viral miRNAs could influence the differentiation status of infected cells, and thereby contribute to KSHV-induced oncogenesis.
Collapse
Affiliation(s)
- Amy Hansen
- Cancer Research UK Viral Oncology Group, University College London Cancer Institute, University College London, London WC1E 6BT, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Miyai M, Tanaka YG, Kamitani A, Hamada M, Takahashi S, Kataoka K. c-Maf and MafB transcription factors are differentially expressed in Huxley's and Henle's layers of the inner root sheath of the hair follicle and regulate cuticle formation. J Dermatol Sci 2010; 57:178-82. [PMID: 20060689 DOI: 10.1016/j.jdermsci.2009.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Revised: 11/13/2009] [Accepted: 12/10/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND The hair follicle of mammalian skin consists of a group of concentric epithelial cell layers. The inner root sheath (IRS), which surrounds the hardening hair shaft beneath the skin surface, is subdivided into three layers, termed the cuticle of the IRS, Huxley's layer, and Henle's layer. The IRS forms a follicular wall in the hair canal and helps guide the developing hair shaft. c-Maf and MafB, members of the Maf family of transcription factors, play important roles in the developmental processes of various tissues and in cell type-specific gene expression. OBJECTIVE The aim of this study is to reveal the pattern of expression and functional roles of c-Maf and MafB in the hair follicle. METHODS We determined the precise location of c-Maf and MafB expression using immunofluorescent staining of mouse skin sections with layer-specific markers. We also analyzed whiskers of c-maf- and mafB-null mice (c-maf(-/-) and mafB(-/-), respectively) using scanning electron microscopy. RESULTS c-Maf and MafB were differentially expressed in the Huxley's and Henle's layers of the IRS. Scanning electron microscopic analysis showed irregular cuticle patterning of whiskers of c-maf(-/-) and mafB(-/-) mice. The cuticles of mafB(-/-) mice were also thinner than those of wild-type mice. CONCLUSION c-Maf and MafB are expressed in the IRS layers in a lineage-restricted manner and are involved in hair morphogenesis.
Collapse
Affiliation(s)
- Masashi Miyai
- Laboratory of Molecular and Developmental Biology, Graduate School of Biological Science, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | | | | | | | | | | |
Collapse
|
72
|
|
73
|
Maeda A, Moriguchi T, Hamada M, Kusakabe M, Fujioka Y, Nakano T, Yoh K, Lim KC, Engel JD, Takahashi S. Transcription factor GATA-3 is essential for lens development. Dev Dyn 2009; 238:2280-91. [PMID: 19623612 DOI: 10.1002/dvdy.22035] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
During vertebrate lens development, the anterior, ectoderm-derived lens vesicle cells differentiate into a monolayer of epithelial cells that retain proliferative potential. Subsequently, they exit the cell cycle and give rise to posterior lens fiber cells that form the lens body. In the present study, we demonstrate that the transcription factor GATA-3 is expressed in the posterior lens fiber cells during embryogenesis, and that GATA-3 deficiency impairs lens development. Interestingly, expression of E-cadherin, a premature lens vesicle marker, is abnormally prolonged in the posterior region of Gata3 homozygous mutant lenses. Furthermore, expression of gamma-crystallin, a differentiation marker for fiber cells, is reduced. This suppressed differentiation is accompanied by an abnormal cellular proliferation, as well as with diminished levels of the cell-cycle inhibitors Cdkn1b/p27 and Cdkn1c/p57 and increased Ccnd2/cyclin D2 abundance. Thus, these observations suggest that GATA-3 is essential for lens cells differentiation and proper cell cycle control.
Collapse
Affiliation(s)
- Atsuko Maeda
- Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Gage PJ, Zacharias AL. Signaling "cross-talk" is integrated by transcription factors in the development of the anterior segment in the eye. Dev Dyn 2009; 238:2149-62. [PMID: 19623614 DOI: 10.1002/dvdy.22033] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Extracellular signaling "cross-talk" between tissues is an important requirement for development of many organs yet the underlying mechanisms generally remain poorly understood. The anterior segment of the eye, which is constructed from four embryonic lineages, provides a unique opportunity to genetically dissect developmental processes such as signaling "cross-talk" without fear of inducing lethality. In the current review, we summarize recent data showing that PITX2, a homeodomain transcription factor, integrates retinoic acid and canonical Wnt/beta-catenin signaling during anterior segment development. Because the requirements for retinoic acid signaling, canonical Wnt/beta-catenin signaling, and PITX2 are not unique to the eye, this newly identified pathway may have relevance elsewhere during development and in tissue homeostasis.
Collapse
Affiliation(s)
- Philip J Gage
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan 48105, USA.
| | | |
Collapse
|
75
|
Malloch EL, Perry KJ, Fukui L, Johnson VR, Wever J, Beck CW, King MW, Henry JJ. Gene expression profiles of lens regeneration and development in Xenopus laevis. Dev Dyn 2009; 238:2340-56. [PMID: 19681139 PMCID: PMC2773617 DOI: 10.1002/dvdy.21998] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Seven hundred and thirty-four unique genes were recovered from a cDNA library enriched for genes up-regulated during the process of lens regeneration in the frog Xenopus laevis. The sequences represent transcription factors, proteins involved in RNA synthesis/processing, components of prominent cell signaling pathways, genes involved in protein processing, transport, and degradation (e.g., the ubiquitin/proteasome pathway), matrix metalloproteases (MMPs), as well as many other proteins. The findings implicate specific signal transduction pathways in the process of lens regeneration, including the FGF, TGF-beta, MAPK, Retinoic acid, Wnt, and hedgehog signaling pathways, which are known to play important roles in eye/lens development and regeneration in various systems. In situ hybridization revealed that the majority of genes recovered are expressed during embryogenesis, including in eye tissues. Several novel genes specifically expressed in lenses were identified. The suite of genes was compared to those up-regulated in other regenerating tissues/organisms, and a small degree of overlap was detected.
Collapse
Affiliation(s)
- Erica L. Malloch
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Kimberly J. Perry
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Lisa Fukui
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Verity R. Johnson
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Jason Wever
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| | - Caroline W. Beck
- University of Otago, Department of Zoology, 340 Great King Street, Dunedin, New Zealand
| | - Michael W. King
- Indiana University School of Medicine and Center for Regenerative Biology and Medicine, Terre Haute, IN 47809
| | - Jonathan J. Henry
- University of Illinois, Department of Cell & Developmental Biology, 601 S. Goodwin Ave. Urbana, IL 61801
| |
Collapse
|
76
|
Takeuchi T, Kudo T, Ogata K, Hamada M, Nakamura M, Kito K, Abe Y, Ueda N, Yamamoto M, Engel JD, Takahashi S. Neither MafA/L-Maf nor MafB is essential for lens development in mice. Genes Cells 2009; 14:941-7. [PMID: 19624757 DOI: 10.1111/j.1365-2443.2009.01321.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The importance of the large Maf transcription factor family has been investigated in lens development in the chick, Xenopus and mammals. Previously we reported that c-maf-deficient mice exhibit severe defects in lens fibre cells. Here, we report the roles of other large Mafs, MafA/L-Maf and MafB, during mouse lens development. MafA/L-Maf and MafB were expressed in lens epithelial cells and fibre cells at E12.5 but had largely disappeared from the lens at E18.5. The lens of mafA-, mafB-deficient and mafA::mafB double-deficient mice developed normally. In c-maf-deficient mice, the pattern of expression of MafA and MafB differed from their expression in wild-type mice. Moreover, the expression of crystallin genes was unchanged in mafA-, mafB- and mafA::mafB double-deficient lens. These results indicate that c-Maf alone is essential for lens development, and that MafA/L-Maf and MafB are dispensable in mice.
Collapse
Affiliation(s)
- Takashi Takeuchi
- Department of Anatomy and Embryology, Doctoral Program in Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Nwosu BU, Raygada M, Tsilou ET, Rennert OM, Stratakis CA. Rieger's Anomaly and Other Ocular Abnormalities in Association with Osteogenesis Imperfecta and aCOL1A1Mutation. Ophthalmic Genet 2009; 26:135-8. [PMID: 16272059 DOI: 10.1080/13816810500228993] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A patient with osteogenesis imperfecta (OI) and some features of Ehlers-Danlos syndrome had Rieger's anomaly and other associated ocular abnormalities. He carried a COL1A1 mutation (c.3313delA) that has only rarely been seen in OI. The association of ocular anterior chamber abnormalities with OI has not been reported previously, while OI with Ehlers-Danlos syndrome features has only been described in some kindreds. The patient had serious complications as a result of his ocular anomalies. We speculate that the course of his disease and, perhaps, its co-existence with OI could be exacerbated by his collagen type-I defect, although no causality can be established by this report of a single case.
Collapse
Affiliation(s)
- Benjamin U Nwosu
- Pediatric Endocrinology Inter-Institute Training Program, Developmental Endocrinology Branch, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
78
|
Goto K, Yasuda M, Sugawara A, Kuramochi T, Itoh T, Azuma N, Ito M. Small Eye Phenotypes Observed in a HumantauGene Transgenic Rat. Curr Eye Res 2009; 31:107-10. [PMID: 16421024 DOI: 10.1080/02713680500478675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
We developed a rat line showing small eye from transgenic rats that were obtained by microinjection of a DNA segment containing the human (h)tau cDNA (GenBank: BC000558: 31-677,774-1180) expressed under control of CAG promoter, which is related to Alzheimer disease, into the pronuclei rat embryos. The rat line was established by selective brother-sister mating of rats showing small eyes. Of 11 offspring in the 11th generation, there were eight animals with microphthalmia and the transgene. The remaining three rats without transgene did not show the small eyes phenotype. The globes of affected rats were 1.2 mm in length compared with normal globes (3.5 mm), and all other ocular structures were normal. The expression of hTau protein was evident immunohistochemically in the ciliary body, extraocular muscle, lens epithelium, and pigment epithelium. Cytogenetic analysis suggested that the chromosome location of the transgene was chromosome 1 (1p12). This region may include genes related to lens development, such as Cat5.
Collapse
Affiliation(s)
- Kazuo Goto
- Central Institute for Experimental Animals, Kawasaki, Japan.
| | | | | | | | | | | | | |
Collapse
|
79
|
Nishimura W, Bonner-Weir S, Sharma A. Expression of MafA in pancreatic progenitors is detrimental for pancreatic development. Dev Biol 2009; 333:108-20. [PMID: 19576197 DOI: 10.1016/j.ydbio.2009.06.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 06/11/2009] [Accepted: 06/22/2009] [Indexed: 12/27/2022]
Abstract
The transcription factor MafA regulates glucose-responsive expression of insulin. MafA-deficient mice have a normal proportion of insulin+ cells at birth but develop diabetes gradually with age, suggesting that MafA is required for maturation and not specification of pancreatic beta-cells. However, several studies show that ectopic expression of MafA may have a role in specification as it induces insulin+ cells in chicken gut epithelium, reprograms adult murine acinar cells into insulin+ cells in combination with Ngn3 and Pdx1, and triggers the lens differentiation. Hence, we examined whether MafA can induce specification of beta-cells during pancreatic development. When the MafA transgene is expressed in Pdx1+ pancreatic progenitors, both pancreatic mass and proliferation of progenitors are reduced, at least partially due to induction of cyclin kinase inhibitors p27 and p57. Expression of MafA in Pdx1+ cells until E12.5 was sufficient to cause these effects and to disproportionately inhibit the formation of endocrine cells in the remnant pancreas. Thus, in mice, MafA expression in Pdx1+ pancreatic progenitors is not sufficient to specify insulin+ cells but in fact deters pancreatic development and the differentiation of endocrine cells. These findings imply that MafA should be used to enhance maturation, rather than specification, of beta-cells from stem/progenitor cells.
Collapse
Affiliation(s)
- Wataru Nishimura
- Section of Islet Transplantation & Cell Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
80
|
Xu J, Yang Y, Qiu G, Lal G, Wu Z, Levy DE, Ochando JC, Bromberg JS, Ding Y. c-Maf regulates IL-10 expression during Th17 polarization. THE JOURNAL OF IMMUNOLOGY 2009; 182:6226-36. [PMID: 19414776 DOI: 10.4049/jimmunol.0900123] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
IL-10 production by Th17 cells is critical for limiting autoimmunity and inflammatory responses. Gene array analysis on Stat6 and T-bet double-deficient Th17 cells identified the Th2 transcription factor c-Maf to be synergistically up-regulated by IL-6 plus TGFbeta and associated with Th17 IL-10 production. Both c-Maf and IL-10 induction during Th17 polarization depended on Stat3, but not Stat6 or Stat1, and mechanistically differed from IL-10 regulation by Th2 or IL-27 signals. TGFbeta was also synergistic with IL-27 to induce c-Maf, and it induced Stat1-independent IL-10 expression in contrast to IL-27 alone. Retroviral transduction of c-Maf was able to induce IL-10 expression in Stat6-deficient CD4 and CD8 T cells, and c-Maf directly transactivated IL-10 gene expression through binding to a MARE (Maf recognition element) motif in the IL-10 promoter. Taken together, these data reveal a novel role for c-Maf in regulating T effector development, and they suggest that TGFbeta may antagonize Th17 immunity by IL-10 production through c-Maf induction.
Collapse
Affiliation(s)
- Jiangnan Xu
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Indoleamine 2,3-dioxygenase overexpression causes kynurenine-modification of proteins, fiber cell apoptosis and cataract formation in the mouse lens. J Transl Med 2009; 89:498-512. [PMID: 19308046 PMCID: PMC2722445 DOI: 10.1038/labinvest.2009.22] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) is the first enzyme in the kynurenine pathway. The kynurenines formed in this pathway chemically modify proteins and cause apoptosis in cells. Evidence suggests that kynurenines and their protein modifications are involved in cataract formation, but this has yet to be directly demonstrated. We generated transgenic (Tg) mouse lines that overexpress human IDO in the lens. Homozygous Tg (homTg) lenses had higher IDO immunoreactivity, approximately 4.5 times greater IDO mRNA, and approximately 8 times higher IDO activity compared to lenses from hemizygous Tg (hemTg) animals. The kynurenine content was threefold higher in homTg than in hemTg but was not detected in wild-type (Wt) lenses. Kynurenine modifications were approximately 2.6 times greater in homTg than in hemTg or Wt. HomTg lenses had vacuoles in the epithelium and cortical fiber cells. Kynurenine modifications coincided with apoptosis in the secondary fiber cells of homTg lenses. Caspase-3 and caspase-9 activities were markedly higher in homTg than in hemTg and Wt. The glutathione content was approximately 36% lower in homTg compared to hemTg and Wt lenses. HomTg animals also developed bilateral cataracts within 3 months of birth. Together these data demonstrate that IDO-mediated production of kynurenines results in defects in fiber cell differentiation and their apoptosis and suggest that IDO activity is kept low in the lens to prevent deleterious effects by kynurenines.
Collapse
|
82
|
Kondo T, El Khattabi I, Nishimura W, Laybutt DR, Geraldes P, Shah S, King G, Bonner-Weir S, Weir G, Sharma A. p38 MAPK is a major regulator of MafA protein stability under oxidative stress. Mol Endocrinol 2009; 23:1281-90. [PMID: 19407223 DOI: 10.1210/me.2008-0482] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mammalian MafA/RIPE3b1 is an important glucose-responsive transcription factor that regulates function, maturation, and survival of beta-cells. Increased expression of MafA results in improved glucose-stimulated insulin secretion and beta-cell function. Because MafA is a highly phosphorylated protein, we examined whether regulating activity of protein kinases can increase MafA expression by enhancing its stability. We demonstrate that MafA protein stability in MIN6 cells and isolated mouse islets is regulated by both p38 MAPK and glycogen synthase kinase 3. Inhibiting p38 MAPK enhanced MafA stability in cells grown under both low and high concentrations of glucose. We also show that the N-terminal domain of MafA plays a major role in p38 MAPK-mediated degradation; simultaneous mutation of both threonines 57 and 134 into alanines in MafA was sufficient to prevent this degradation. Under oxidative stress, a condition detrimental to beta-cell function, a decrease in MafA stability was associated with a concomitant increase in active p38 MAPK. Interestingly, inhibiting p38 MAPK but not glycogen synthase kinase 3 prevented oxidative stress-dependent degradation of MafA. These results suggest that the p38 MAPK pathway may represent a common mechanism for regulating MafA levels under oxidative stress and basal and stimulatory glucose concentrations. Therefore, preventing p38 MAPK-mediated degradation of MafA represents a novel approach to improve beta-cell function.
Collapse
Affiliation(s)
- Takuma Kondo
- Section of Islet Transplantation and Cell Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Shi X, Cui B, Wang Z, Weng L, Xu Z, Ma J, Xu G, Kong X, Hu L. Removal of Hsf4 leads to cataract development in mice through down-regulation of gamma S-crystallin and Bfsp expression. BMC Mol Biol 2009; 10:10. [PMID: 19224648 PMCID: PMC2653017 DOI: 10.1186/1471-2199-10-10] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2008] [Accepted: 02/19/2009] [Indexed: 11/10/2022] Open
Abstract
Background Heat-shock transcription factor 4 (HSF4) mutations are associated with autosomal dominant lamellar cataract and Marner cataract. Disruptions of the Hsf4 gene cause lens defects in mice, indicating a requirement for HSF4 in fiber cell differentiation during lens development. However, neither the relationship between HSF4 and crystallins nor the detailed mechanism of maintenance of lens transparency by HSF4 is fully understood. Results In an attempt to determine how the underlying biomedical and physiological mechanisms resulting from loss of HSF4 contribute to cataract formation, we generated an Hsf4 knockout mouse model. We showed that the Hsf4 knockout mouse (Hsf4-/-) partially mimics the human cataract caused by HSF4 mutations. Q-PCR analysis revealed down-regulation of several cataract-relevant genes, including γS-crystallin (Crygs) and lens-specific beaded filament proteins 1 and 2 (Bfsp1 and Bfsp2), in the lens of the Hsf4-/- mouse. Transcription activity analysis using the dual-luciferase system suggested that these cataract-relevant genes are the direct downstream targets of HSF4. The effect of HSF4 on γS-crystallin is exemplified by the cataractogenesis seen in the Hsf4-/-,rncat intercross. The 2D electrophoretic analysis of whole-lens lysates revealed a different expression pattern in 8-week-old Hsf4-/- mice compared with their wild-type counterparts, including the loss of some αA-crystallin modifications and reduced expression of γ-crystallin proteins. Conclusion Our results indicate that HSF4 is sufficiently important to lens development and disruption of the Hsf4 gene leads to cataracts via at least three pathways: 1) down-regulation of γ-crystallin, particularly γS-crystallin; 2) decreased lens beaded filament expression; and 3) loss of post-translational modification of αA-crystallin.
Collapse
Affiliation(s)
- Xiaohe Shi
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Fuchshofer R, Stephan DA, Russell P, Tamm ER. Gene expression profiling of TGFbeta2- and/or BMP7-treated trabecular meshwork cells: Identification of Smad7 as a critical inhibitor of TGF-beta2 signaling. Exp Eye Res 2009; 88:1020-32. [PMID: 19450457 DOI: 10.1016/j.exer.2009.01.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 11/20/2008] [Accepted: 01/06/2009] [Indexed: 01/11/2023]
Abstract
A distinct structural change in the trabecular meshwork (TM) of patients with primary open-angle glaucoma (POAG) is the increase in fibrillar extracellular matrix (ECM) in the juxtacanalicular region of the TM. Transforming growth factor (TGF)-beta2 signaling may be involved, as TGF-beta2 is significantly increased in the aqueous humor of patients with POAG. In cultured human TM cells, TGF-beta2 causes an increase in ECM deposition, an effect that is blunted or prevented, if BMP7 is added in combination with TGF-beta2. In order to know more about the signaling network that is induced in HTM cells treated with BMP7, TGF-beta2 or the combination of both factors, we identified differentially regulated genes by microarray analysis, and confirmed selected genes by quantitative RT-PCR, Western blotting, or immunohistochemistry. We observed multiple effects of both TGF-beta2 and BMP7 on the expression of a considerable number of genes involved in growth factor signaling, ECM structure and turnover, and modification of the cytoskeleton. Among the genes that were found to be regulated were CAPZA1, CDC42BPB, EFEMP1, FGF5, FSTL3, HBEGF, LTBP1, LTBP2, MATN2, NRP1, SERPINE1, SH3MD1, SMTN, SMAD7, TFPI2, TNFAIP6, and VEGF. Since SMAD7 encodes for Smad7, an inhibitory Smad that acts in a negative-feedback loop to inhibit TGF-beta activity, we silenced Smad7 mRNA in cultured human TM cells by a specific small interfering RNA. Silencing of its mRNA caused a substantial knock down of Smad7 in TM cells. Following combined BMP7/TGF-beta2 treatment, the antagonizing effect of BMP7 on TGF-beta2-induced CTGF expression was abolished. We conclude that Smad7 is the key molecular switch that inhibits TGF-beta2 signaling, and mediates the blunting effects of BMP7 on TGF-beta2 in TM cells. A therapeutic modulation of Smad7 might be a promising approach to influence ECM turnover in the TM and to treat POAG.
Collapse
Affiliation(s)
- Rudolf Fuchshofer
- Institute of Human Anatomy and Embryology, University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany
| | | | | | | |
Collapse
|
85
|
Ho HY, Chang KH, Nichols J, Li M. Homeodomain protein Pitx3 maintains the mitotic activity of lens epithelial cells. Mech Dev 2008; 126:18-29. [PMID: 19007884 DOI: 10.1016/j.mod.2008.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Revised: 09/15/2008] [Accepted: 10/22/2008] [Indexed: 11/18/2022]
Abstract
Pitx3 is a bicoid like homeobox transcription factor of which deficiency in mice is linked with the aphakia phenotype. Mutation in human PITX3 gene is associated with autosomal dominant cataract with variable anterior segment mesenchymal dysgenesis. However, the molecular events causing the morphological changes in aphakia remains unknown. In this study we investigated the behaviour of GFP tagged Pitx3 null embryonic stem cells in chimeric lens, as well as the molecular features of the Pitx3-deficient lens of homozygous Pitx3 knockout mice. We show that the lack of colonisation of Pitx3-deficient ES cell derivatives in Pitx3 wild-type<-->Pitx3 null chimeric lens was due to the depletion of the epithelial cells in lens epithelium manifested by aberrant cell cycle exit and precocious onset of fibre cell differentiation of the Pitx3 null cells at the lens vesicle stage. This was demonstrated by the early activation of the cell cycle inhibitors p27Kip1 and p57Kip2, and the expression of beta-and gamma-crystallins. These defects are at least partially attributed to the loss of FoxE3 and misexpression of Prox1 in the lens vesicle epithelial cells. Thus, Pitx3 is essential to maintain lens epithelial phenotype and prevent inappropriate fibre cell differentiation during lens development.
Collapse
Affiliation(s)
- Hsin-Yi Ho
- Institute for Stem Cell Research, University of Edinburgh, Edinburgh, UK
| | | | | | | |
Collapse
|
86
|
Carmona FD, Jiménez R, Collinson JM. The molecular basis of defective lens development in the Iberian mole. BMC Biol 2008; 6:44. [PMID: 18939978 PMCID: PMC2587461 DOI: 10.1186/1741-7007-6-44] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Accepted: 10/21/2008] [Indexed: 11/24/2022] Open
Abstract
Background Fossorial mammals face natural selection pressures that differ from those acting on surface dwelling animals, and these may lead to reduced visual system development. We have studied eye development in a species of true mole, the Iberian mole Talpa occidentalis, and present the molecular basis of abnormal lens development. This is the first embryological developmental study of the eyes of any fossorial mammal at the molecular level. Results Lens fibre differentiation is not completed in the Iberian mole. Although eye development starts normally (similar to other model species), defects are seen after closure of the lens vesicle. PAX6 is not down-regulated in developing lens fibre nuclei, as it is in other species, and there is ectopic expression of FOXE3, a putative downstream effector of PAX6, in some, but not all lens fibres. FOXE3-positive lens fibres continue to proliferate within the posterior compartment of the embryonic lens, but unlike in the mouse, no proliferation was detected anywhere in the postnatal mole lens. The undifferentiated status of the anterior epithelial cells was compromised, and most of them undergo apoptosis. Furthermore, β-crystallin and PROX1 expression patterns are abnormal and our data suggest that genes encoding β-crystallins are not directly regulated by PAX6, c-MAF and PROX1 in the Iberian mole, as they are in other model vertebrates. Conclusion In other model vertebrates, genetic pathways controlling lens development robustly compartmentalise the lens into a simple, undifferentiated, proliferative anterior epithelium, and quiescent, anuclear, terminally differentiated posterior lens fibres. These pathways are not as robust in the mole, and lead to loss of the anterior epithelial phenotype and only partial differentiation of the lens fibres, which continue to express 'epithelial' genes. Paradigms of genetic regulatory networks developed in other vertebrates appear not to hold true for the Iberian mole.
Collapse
Affiliation(s)
- F David Carmona
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | | | | |
Collapse
|
87
|
Abstract
Like JUN and FOS, the Maf transcription factors belong to the AP1 family. Besides their established role in human cancer--overexpression of the large Maf genes promotes the development of multiple myeloma--they can display tumour suppressor-like activity in specific cellular contexts, which is compatible with their physiological role in terminal differentiation. However, their oncogenic activity relies mostly on the acquisition of new biological functions relevant to cell transformation, the most striking characteristic of Maf oncoproteins being their ability to enhance pathological interactions between tumour cells and the stroma.
Collapse
Affiliation(s)
- Alain Eychène
- Institut Curie, Centre de Recherche, Orsay F-91405, France
| | | | | |
Collapse
|
88
|
Cain S, Martinez G, Kokkinos MI, Turner K, Richardson RJ, Abud HE, Huelsken J, Robinson ML, de Iongh RU. Differential requirement for beta-catenin in epithelial and fiber cells during lens development. Dev Biol 2008; 321:420-33. [PMID: 18652817 DOI: 10.1016/j.ydbio.2008.07.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Revised: 06/27/2008] [Accepted: 07/01/2008] [Indexed: 11/18/2022]
Abstract
Recent studies implicate Wnt/beta-catenin signaling in lens differentiation (Stump, R. J., et al., 2003. A role for Wnt/beta-catenin signaling in lens epithelial differentiation. Dev Biol;259:48-61). Beta-catenin is a component of adherens junctions and functions as a transcriptional activator in canonical Wnt signaling. We investigated the effects of Cre/LoxP-mediated deletion of beta-catenin during lens development using two Cre lines that specifically deleted beta-catenin in whole lens or only in differentiated fibers, from E13.5. We found that beta-catenin was required in lens epithelium and during early fiber differentiation but appeared to be redundant in differentiated fiber cells. Complete loss of beta-catenin resulted in an abnormal and deficient epithelial layer with loss of E-cadherin and Pax6 expression as well as abnormal expression of c-Maf and p57(kip2) but not Prox1. There was also disrupted fiber cell differentiation, characterized by poor cell elongation, decreased beta-crystallin expression, epithelial cell cycle arrest at G(1)-S transition and premature cell cycle exit. Despite cell cycle arrest there was no induction of apoptosis. Mutant fiber cells displayed altered apical-basal polarity as evidenced by altered distribution of the tight junction protein, ZO1, disruption of apical actin filaments and abnormal deposition of extracellular matrix, resulting in a deficient lens capsule. Loss of beta-catenin also affected the formation of adhesion junctions as evidenced by dissociation of N-cadherin and F-actin localization in differentiating fiber cells. However, loss of beta-catenin from terminally differentiating fibers had no apparent effects on adhesion junctions between adjacent embryonic fibers. These data indicate that beta-catenin plays distinct functions during lens fiber differentiation and is involved in both Wnt signaling and adhesion-related mechanisms that regulate lens epithelium and early fiber differentiation.
Collapse
Affiliation(s)
- Sarah Cain
- Ocular Development Laboratory, Anatomy and Cell Biology Department, University of Melbourne, Victoria 3010, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Notch signaling regulates growth and differentiation in the mammalian lens. Dev Biol 2008; 321:111-22. [PMID: 18588871 DOI: 10.1016/j.ydbio.2008.06.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 06/02/2008] [Accepted: 06/03/2008] [Indexed: 11/23/2022]
Abstract
The Notch signal transduction pathway regulates the decision to proliferate versus differentiate. Although there are a myriad of mouse models for the Notch pathway, surprisingly little is known about how these genes regulate early eye development, particularly in the anterior lens. We employed both gain-of-function and loss-of-function approaches to determine the role of Notch signaling in lens development. Here we analyzed mice containing conditional deletion of the Notch effector Rbpj or overexpression of the activated Notch1 intracellular domain during lens formation. We demonstrate distinct functions for Notch signaling in progenitor cell growth, fiber cell differentiation and maintenance of the transition zone. In particular, Notch signaling controls the timing of primary fiber cell differentiation and is essential for secondary fiber cell differentiation. Either gain or loss of Notch signaling leads to formation of a dysgenic lens, which in loss-of-function mice undergoes a profound postnatal degeneration. Our data suggest both Cyclin D1 and Cyclin D2, and the p27(Kip1) cyclin-dependent kinase inhibitor act downstream of Notch signaling, and define multiple critical functions for this pathway during lens development.
Collapse
|
90
|
Vanhoose AM, Samaras S, Artner I, Henderson E, Hang Y, Stein R. MafA and MafB regulate Pdx1 transcription through the Area II control region in pancreatic beta cells. J Biol Chem 2008; 283:22612-9. [PMID: 18522939 PMCID: PMC2504898 DOI: 10.1074/jbc.m802902200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pancreatic-duodenal homeobox factor-1 (Pdx1) is highly enriched in islet β cells and integral to proper cell development and adult function. Of the four conserved 5′-flanking sequence blocks that contribute to transcription in vivo, Area II (mouse base pairs -2153/-1923) represents the only mammalian specific control domain. Here we demonstrate that regulation of β-cell-enriched Pdx1 expression by the MafA and MafB transcription factors is exclusively through Area II. Thus, these factors were found to specifically activate through Area II in cell line transfection-based assays, and MafA, which is uniquely expressed in adult islet β cells was only bound to this region in quantitative chromatin immunoprecipitation studies. MafA and MafB are produced in β cells during development and were both bound to Area II at embryonic day 18.5. Expression of a transgene driven by Pdx1 Areas I and II was also severely compromised during insulin+ cell formation in MafB-/- mice, consistent with the importance of this large Maf in β-cell production and Pdx1 expression. These findings illustrate the significance of large Maf proteins to Pdx1 expression in β cells, and in particular MafB during pancreatic development.
Collapse
Affiliation(s)
- Amanda M Vanhoose
- Department of Molecular Physiology and Biophysics, Vanderbilt Medical School, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
91
|
Zhao H, Yang T, Madakashira BP, Thiels CA, Bechtle CA, Garcia CM, Zhang H, Yu K, Ornitz DM, Beebe DC, Robinson ML. Fibroblast growth factor receptor signaling is essential for lens fiber cell differentiation. Dev Biol 2008; 318:276-88. [PMID: 18455718 DOI: 10.1016/j.ydbio.2008.03.028] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 03/14/2008] [Accepted: 03/17/2008] [Indexed: 11/19/2022]
Abstract
The vertebrate lens provides an excellent model to study the mechanisms that regulate terminal differentiation. Although fibroblast growth factors (FGFs) are thought to be important for lens cell differentiation, it is unclear which FGF receptors mediate these processes during different stages of lens development. Deletion of three FGF receptors (Fgfr1-3) early in lens development demonstrated that expression of only a single allele of Fgfr2 or Fgfr3 was sufficient for grossly normal lens development, while mice possessing only a single Fgfr1 allele developed cataracts and microphthalmia. Profound defects were observed in lenses lacking all three Fgfrs. These included lack of fiber cell elongation, abnormal proliferation in prospective lens fiber cells, reduced expression of the cell cycle inhibitors p27(kip1) and p57(kip2), increased apoptosis and aberrant or reduced expression of Prox1, Pax6, c-Maf, E-cadherin and alpha-, beta- and gamma-crystallins. Therefore, while signaling by FGF receptors is essential for lens fiber differentiation, different FGF receptors function redundantly.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Enlargement
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Cyclin-Dependent Kinase Inhibitor p57/metabolism
- Eye Abnormalities/embryology
- Fibroblast Growth Factors/metabolism
- Gene Targeting
- Homeodomain Proteins/metabolism
- Lens, Crystalline/cytology
- Lens, Crystalline/embryology
- Mice
- Mutation
- Proto-Oncogene Proteins c-maf/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Signal Transduction
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Haotian Zhao
- Graduate Program in Molecular, Cellular and Developmental Biology, College of Biological Sciences, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Kwakowsky A, Schwirtlich M, Zhang Q, Eisenstat DD, Erdélyi F, Baranyi M, Katarova ZD, Szabó G. GAD isoforms exhibit distinct spatiotemporal expression patterns in the developing mouse lens: correlation with Dlx2 and Dlx5. Dev Dyn 2008; 236:3532-44. [PMID: 17969168 DOI: 10.1002/dvdy.21361] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gamma-aminobutyric acid (GABA), the major inhibitory neurotransmitter of the adult nervous system and its biosynthetic enzyme glutamic acid decarboxylase (GAD) are abundantly expressed in the embryonic nervous system and are involved in the modulation of cell proliferation, migration, and differentiation. Here we describe for the first time the expression of GABA and embryonic and adult GAD isoforms in the developing mouse lens. We show that the GAD isoforms are sequentially induced with specific spatiotemporal profiles: GAD65 and embryonic GAD isoforms prevail in primary fibers, while GAD67 is the predominant GAD expressed in the postnatal secondary fibers. This pattern correlates well with the expression of Dlx2 and Dlx5, known as upstream regulators of GAD. GABA and GAD are most abundant at the tips of elongating fibers and are absent from organelle-free cells, suggesting their involvement is primarily in shaping of the cytoskeleton during fiber elongation stages.
Collapse
Affiliation(s)
- Andrea Kwakowsky
- Department of Gene Technology and Developmental Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Nakayama Y, Miyake A, Nakagawa Y, Mido T, Yoshikawa M, Konishi M, Itoh N. Fgf19 is required for zebrafish lens and retina development. Dev Biol 2007; 313:752-66. [PMID: 18089288 DOI: 10.1016/j.ydbio.2007.11.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Revised: 11/06/2007] [Accepted: 11/09/2007] [Indexed: 02/01/2023]
Abstract
Fgf signaling plays crucial roles in morphogenesis. Fgf19 is required for zebrafish forebrain development. Here, we examined the roles of Fgf19 in the formation of the lens and retina in zebrafish. Knockdown of Fgf19 caused a size reduction of the lens and the retina, failure of closure of the choroids fissure, and a progressive expansion of the retinal tissue to the midline of the forebrain. Fgf19 expressed in the nasal retina and lens was involved in cell survival but not cell proliferation during embryonic lens and retina development. Fgf19 was essential for the differentiation of lens fiber cells in the lens but not for the neuronal differentiation and lamination in the retina. Loss of nasal fate in the retina caused by the knockdown of Fgf19, expansion of nasal fate in the retina caused by the overexpression of Fgf19 and eye transplantation indicated that Fgf19 in the retina was crucial for the nasal-temporal patterning of the retina that is critical for the guidance of retinal ganglion cell axons. Knockdown of Fgf19 also caused incorrect axon pathfinding. The present findings indicate that Fgf19 positively regulates the patterning and growth of the retina, and the differentiation and growth of the lens in zebrafish.
Collapse
Affiliation(s)
- Yoshiaki Nakayama
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto 606-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
94
|
Roth CL, Mastronardi C, Lomniczi A, Wright H, Cabrera R, Mungenast AE, Heger S, Jung H, Dubay C, Ojeda SR. Expression of a tumor-related gene network increases in the mammalian hypothalamus at the time of female puberty. Endocrinology 2007; 148:5147-61. [PMID: 17615149 DOI: 10.1210/en.2007-0634] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Much has been learned in recent years about the central mechanisms controlling the initiation of mammalian puberty. It is now clear that this process requires the interactive participation of several genes. Using a combination of high throughput, molecular, and bioinformatics strategies, in combination with a system biology approach, we singled out from the hypothalamus of nonhuman primates and rats a group of related genes whose expression increases at the time of female puberty. Although these genes [henceforth termed tumor-related genes (TRGs)] have diverse cellular functions, they share the common feature of having been earlier identified as involved in tumor suppression/tumor formation. A prominent member of this group is KiSS1, a gene recently shown to be essential for the occurrence of puberty. Cis-regulatory analysis revealed the presence of a hierarchically arranged gene set containing five major hubs (CDP/CUTL1, MAF, p53, YY1, and USF2) controlling the network at the transcriptional level. In turn, these hubs are heavily connected to non-TRGs involved in the transcriptional regulation of the pubertal process. TRGs may be expressed in the mammalian hypothalamus as components of a regulatory gene network that facilitates and integrates cellular and cell-cell communication programs required for the acquisition of female reproductive competence.
Collapse
Affiliation(s)
- Christian L Roth
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Reza HM, Nishi H, Kataoka K, Takahashi Y, Yasuda K. L-Maf regulates p27kip1 expression during chick lens fiber differentiation. Differentiation 2007; 75:737-44. [PMID: 17428264 DOI: 10.1111/j.1432-0436.2007.00171.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Organ formation requires spatio-temporal proliferation and differentiation of precursor cells. During lens development, placodal cells in the posterior lens vesicle exit from the cell cycle and enter into the process of differentiation. Cyclin-dependent kinase inhibitors play critical roles in cell cycle exit and promote differentiation in several tissues. We have found that p27kip1 is expressed in the posterior lens cells that undergo differentiation to form the differentiated fiber cells. The transcription factor L-Maf is expressed in these cells earlier than p27kip1. From in ovo gain- or loss-of-function experiments, we have found that L-Maf can, respectively, induce or inhibit the expression of p27kip1 in lens cells. Promoter assays using the 5' upstream sequences of the human p27kip1 gene indicate that L-Maf can activate p27kip1 transcription through the basal regulatory region. We suggest that L-Maf regulates cell cycle exit of the posterior lens cells by activating p27kip1, and thus directs fiber cell differentiation during lens formation in chick.
Collapse
Affiliation(s)
- Hasan Mahmud Reza
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma 630-0101, Japan.
| | | | | | | | | |
Collapse
|
96
|
Cvekl A, Duncan MK. Genetic and epigenetic mechanisms of gene regulation during lens development. Prog Retin Eye Res 2007; 26:555-97. [PMID: 17905638 PMCID: PMC2136409 DOI: 10.1016/j.preteyeres.2007.07.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Recent studies demonstrated a number of links between chromatin structure, gene expression, extracellular signaling and cellular differentiation during lens development. Lens progenitor cells originate from a pool of common progenitor cells, the pre-placodal region (PPR) which is formed from a combination of extracellular signaling between the neural plate, naïve ectoderm and mesendoderm. A specific commitment to the lens program over alternate choices such as the formation of olfactory epithelium or the anterior pituitary is manifested by the formation of a thickened surface ectoderm, the lens placode. Mouse lens progenitor cells are characterized by the expression of a complement of lens lineage-specific transcription factors including Pax6, Six3 and Sox2, controlled by FGF and BMP signaling, followed later by c-Maf, Mab21like1, Prox1 and FoxE3. Proliferation of lens progenitors together with their morphogenetic movements results in the formation of the lens vesicle. This transient structure, comprised of lens precursor cells, is polarized with its anterior cells retaining their epithelial morphology and proliferative capacity, whereas the posterior lens precursor cells initiate terminal differentiation forming the primary lens fibers. Lens differentiation is marked by expression and accumulation of crystallins and other structural proteins. The transcriptional control of crystallin genes is characterized by the reiterative use of transcription factors required for the establishment of lens precursors in combination with more ubiquitously expressed factors (e.g. AP-1, AP-2alpha, CREB and USF) and recruitment of histone acetyltransferases (HATs) CBP and p300, and chromatin remodeling complexes SWI/SNF and ISWI. These studies have poised the study of lens development at the forefront of efforts to understand the connections between development, cell signaling, gene transcription and chromatin remodeling.
Collapse
Affiliation(s)
- Ales Cvekl
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | |
Collapse
|
97
|
Yang Y, Wolf LV, Cvekl A. Distinct embryonic expression and localization of CBP and p300 histone acetyltransferases at the mouse alphaA-crystallin locus in lens. J Mol Biol 2007; 369:917-26. [PMID: 17467007 PMCID: PMC2063435 DOI: 10.1016/j.jmb.2007.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 03/26/2007] [Accepted: 04/02/2007] [Indexed: 10/23/2022]
Abstract
Mouse alphaA-crystallin gene encodes the most abundant protein of the mammalian lens. Expression of alphaA-crystallin is regulated temporally and spatially during lens development with initial expression in the lens vesicle followed by strong upregulation in the differentiating primary lens fibers. Lens-specific expression of alphaA-crystallin is mediated by DNA-binding transcription factors Pax6, c-Maf and CREB bound to its promoter region. Its 5'-distal enhancer, DCR1, mediates regulation of alphaA-crystallin via FGF signaling, while its 3'-distal enhancer, DCR3, functions only in elongated primary lens fibers via other lens differentiation pathways. DCR1 and DCR3 establish outside borders of a lens-specific chromatin region marked by histone H3 K9 acetylation. Here, we identified CREB-binding protein (CBP) and p300 as major histone acetyltransferases (HATs) associated in vivo with the mouse alphaA-crystallin locus. Both HATs are expressed in embryonic lens. Expression of CBP in primary lens fiber cells coincides with alphaA-crystallin. In the chromatin of lens epithelial cells, chromatin immunoprecipitations (ChIPs) show that the alphaA-crystallin promoter is notably devoid of any significant presence of CBP and p300, though DCR1 and a few other regions show the presence of these HATs. In the chromatin obtained from newborn lens, CBP was localized specifically at the promoter region with about ten times higher abundance compared to the entire alphaA-crystallin locus. In contrast, p300 is distributed more evenly across the entire locus. Analysis of total histone H3 and H3 K9 acetylation revealed potential lower density of nucleosomes 2 kb upstream from the promoter region. Collectively, our data suggest that moderate level of alphaA-crystallin gene expression in lens epithelial cells does not require the presence of CBP and p300 in the promoter. However, the lens-specific chromatin domain contains both promoter localized CBP on the "background" of locus-spread presence of CBP and p300.
Collapse
Affiliation(s)
- Ying Yang
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY10461, USA
| | | | | |
Collapse
|
98
|
Perveen R, Favor J, Jamieson RV, Ray DW, Black GCM. A heterozygous c-Maf transactivation domain mutation causes congenital cataract and enhances target gene activation. Hum Mol Genet 2007; 16:1030-8. [PMID: 17374726 DOI: 10.1093/hmg/ddm048] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
MAF, one of a family of large Maf bZIP transcription factors, is mutated in human developmental ocular disorders that include congenital cataract, microcornea, coloboma and anterior segment dysgenesis. Expressed early in the developing lens vesicle, it is central to regulation of lens crystallin gene expression. We report a semi-dominant mouse c-Maf mutation recovered after ENU mutatgenesis which results in the substitution, D90V, at a highly conserved residue within the N-terminal 35 amino-acid minimal transactivation domain (MTD). Unlike null and loss-of-function c-Maf mutations, which cause severe runting and renal abnormalities, the phenotype caused by the D90V mutation is isolated cataract. In reporter assays, D90V results in increased promoter activation, a situation similar to MTD mutations of NRL that also cause human disease. In contrast to wild-type protein, the c-Maf D90V mutant protein is not inhibited by protein kinase A-dependent pathways. The MTD of large Maf proteins has been shown to interact with the transcriptional co-activator p300 and we demonstrate that c-Maf D90V enhances p300 recruitment in a cell-type dependent manner. We observed the same for the pathogenic human NRL MTD mutation S50T, which suggests a common mechanism of action.
Collapse
Affiliation(s)
- R Perveen
- Academic Unit of Medical Genetics and Regional Genetics Service Department of Clinical Genetics, Central Manchester and Manchester Children's University Hospitals NHS Trust, St Mary's Hospital, Hathersage Road, Manchester M13 0JH, UK
| | | | | | | | | |
Collapse
|
99
|
Donner AL, Ko F, Episkopou V, Maas RL. Pax6 is misexpressed in Sox1 null lens fiber cells. Gene Expr Patterns 2007; 7:606-13. [PMID: 17306631 PMCID: PMC2246053 DOI: 10.1016/j.modgep.2007.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Revised: 01/03/2007] [Accepted: 01/03/2007] [Indexed: 01/03/2023]
Abstract
Sox1 null lens fiber cells fail to elongate and have disrupted expression of gamma-crystallin. We have evaluated the expression of Sox1 and Pax6 proteins during critical stages of lens morphogenesis, with particular focus on fiber cell differentiation. While Pax6 and Sox1 are co-expressed during early stages of fiber cell differentiation, Sox1 up-regulation coincides temporally with the down-regulation of Pax6, and these proteins therefore display a striking inverse expression pattern in the lens fiber cell compartment. Furthermore, Pax6 is inappropriately expressed in the fiber cells of Sox1 null mice and the Pax6 target, alpha5 integrin, is simultaneously misexpressed. Finally, we demonstrate a genetic interaction between Sox1 and Pax6, as Sox1 heterozygosity partially rescues the diameter of Pax6(Sey) lenses by increasing the number of cells in the fiber cell compartment.
Collapse
Affiliation(s)
- Amy L Donner
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, NRB 458, 77 Louis Pasteur Avenue, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
100
|
Abstract
Crystallins are the predominant structural proteins in the lens that are evolutionarily related to stress proteins. They were first discovered outside the vertebrate eye lens by Bhat and colleagues in 1989 who found alphaB-crystallin expression in the retina, heart, skeletal muscles, skin, brain and other tissues. With the advent of microarray and proteome analysis, there is a clearer demonstration that crystallins are prominent proteins both in the normal retina and in retinal pathologies, emphasizing the importance of understanding crystallin functions outside of the lens. There are two main crystallin gene families: alpha-crystallins, and betagamma-crystallins. alpha-crystallins are molecular chaperones that prevent aberrant protein interactions. The chaperone properties of alpha-crystallin are thought to allow the lens to tolerate aging-induced deterioration of the lens proteins without showing signs of cataracts until older age. alpha-crystallins not only possess chaperone-like activity in vitro, but can also remodel and protect the cytoskeleton, inhibit apoptosis, and enhance the resistance of cells to stress. Recent advances in the field of structure-function relationships of alpha-crystallins have provided the first clues to their underlying roles in tissues outside the lens. Proteins of the betagamma-crystallin family have been suggested to affect lens development, and are also expressed in tissues outside the lens. The goal of this paper is to highlight recent work with lens epithelial cells from alphaA- and alphaB-crystallin knockout mice. The use of lens epithelial cells suggests that crystallins have important cellular functions in the lens epithelium and not just the lens fiber cells as previously thought. These studies may be directly relevant to understanding the general cellular functions of crystallins.
Collapse
Affiliation(s)
- Usha P Andley
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|