Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073089

ABSTRACT

Sox proteins are known as crucial transcription factors for many developmental processes and for a wide range of common diseases. They were believed to specifically bind and bend DNA with other transcription factors and elicit transcriptional activation or repression activities in the early stage of transcription. However, their functions are not limited to transcription initiation. It has been showed that Sox proteins are involved in the regulation of alternative splicing regulatory networks and translational control. In this review, we discuss the current knowledge on how Sox transcription factors such as Sox2, Sry, Sox6, and Sox9 allow the coordination of co-transcriptional splicing and also the mechanism of SOX4-mediated translational control in the context of RNA polymerase III.


Subject(s)
SOX Transcription Factors , Alternative Splicing , Animals , Humans , Protein Biosynthesis , RNA Polymerase III , RNA Splicing , SOX Transcription Factors/genetics , SOX Transcription Factors/physiology , Spliceosomes/metabolism , Transcriptional Activation
2.
Aging Cell ; 20(5): e13343, 2021 05.
Article in English | MEDLINE | ID: mdl-33721419

ABSTRACT

Although important factors governing the meiosis have been reported in the embryonic ovary, meiosis in postnatal testis remains poorly understood. Herein, we first report that SRY-box 30 (Sox30) is an age-related and essential regulator of meiosis in the postnatal testis. Sox30-null mice exhibited uniquely impaired testis, presenting the abnormal arrest of germ-cell differentiation and irregular Leydig cell proliferation. In aged Sox30-null mice, the observed testicular impairments were more severe. Furthermore, the germ-cell arrest occurred at the stage of meiotic zygotene spermatocytes, which is strongly associated with critical regulators of meiosis (such as Cyp26b1, Stra8 and Rec8) and sex differentiation (such as Rspo1, Foxl2, Sox9, Wnt4 and Ctnnb1). Mechanistically, Sox30 can activate Stra8 and Rec8, and inhibit Cyp26b1 and Ctnnb1 by direct binding to their promoters. A different Sox30 domain required for regulating the activity of these gene promoters, providing a "fail-safe" mechanism for Sox30 to facilitate germ-cell differentiation. Indeed, retinoic acid levels were reduced owing to increased degradation following the elevation of Cyp26b1 in Sox30-null testes. Re-expression of Sox30 in Sox30-null mice successfully restored germ-cell meiosis, differentiation and Leydig cell proliferation. Moreover, the restoration of actual fertility appeared to improve over time. Consistently, Rec8 and Stra8 were reactivated, and Cyp26b1 and Ctnnb1 were reinhibited in the restored testes. In summary, Sox30 is necessary, sufficient and age-associated for germ-cell meiosis and differentiation in testes by direct regulating critical regulators. This study advances our understanding of the regulation of germ-cell meiosis and differentiation in the postnatal testis.


Subject(s)
SOX Transcription Factors/physiology , Spermatozoa/cytology , Testis/cytology , Aging , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Gene Expression Regulation , Male , Meiosis , Meiotic Prophase I , Mice , Promoter Regions, Genetic , Protein Domains , SOX Transcription Factors/chemistry , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , Sex Differentiation , Testis/metabolism , Tretinoin/metabolism
3.
Exp Eye Res ; 161: 174-192, 2017 08.
Article in English | MEDLINE | ID: mdl-28577895

ABSTRACT

Sox2 is a well-established neuronal stem cell-associated transcription factor that regulates neural development and adult neurogenesis in vertebrates, and is one of the critical genes used to reprogram differentiated cells into induced pluripotent stem cells. We examined if Sox2 was involved in the early reprogramming-like events that Müller glia undergo as they upregulate many pluripotency- and neural stem cell-associated genes required for proliferation in light-damaged adult zebrafish retinas. In the undamaged adult zebrafish retina, Sox2 is expressed in Müller glia and a subset of amacrine cells, similar to other vertebrates. Following 31 h of light damage, Sox2 expression significantly increased in proliferating Müller glia. Morpholino-mediated knockdown of Sox2 expression resulted in decreased numbers of proliferating Müller glia, while induced overexpression of Sox2 stimulated Müller glia proliferation in the absence of retinal damage. Thus, Sox2 is necessary and sufficient for Müller glia proliferation. We investigated the role of Wnt/ß-catenin signaling, which is a known regulator of sox2 expression during vertebrate retinal development. While ß-catenin 2, but not ß-catenin 1, was necessary for Müller glia proliferation, neither ß-catenin paralog was required for sox2 expression following retinal damage. Sox2 expression was also necessary for ascl1a (neurogenic) and lin28a (reprogramming) expression, but not stat3 expression following retinal damage. Furthermore, Sox2 was required for Müller glial-derived neuronal progenitor cell amplification and expression of the pro-neural marker Tg(atoh7:EGFP). Finally, loss of Sox2 expression prevented complete regeneration of cone photoreceptors. This study is the first to identify a functional role for Sox2 during Müller glial-based regeneration of the vertebrate retina.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation/physiology , Ependymoglial Cells/metabolism , Nerve Regeneration/physiology , RNA-Binding Proteins/metabolism , Retina/physiology , SOX Transcription Factors/physiology , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Cell Differentiation , Fluorescent Antibody Technique, Indirect , Gene Knockdown Techniques , Immunoblotting , In Situ Nick-End Labeling , Light , Neural Stem Cells/metabolism , Neurogenesis/physiology , Radiation Injuries, Experimental/metabolism , Real-Time Polymerase Chain Reaction , Retina/radiation effects , Transcription Factors , Zebrafish
4.
Sci Rep ; 6: 32874, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27611740

ABSTRACT

The sex-determining gene SRY induces SOX9 expression in the testes of eutherian mammals via two pathways. SRY binds to testis-specific enhancer of Sox9 (TESCO) with SF1 to activate SOX9 transcription. SRY also up-regulates ER71 expression, and ER71 activates Sox9 transcription. After the initiation of testis differentiation, SOX9 enhances Amh expression by binding to its promoter with SF1. SOX8, SOX9 and SOX10, members of the SOXE gene family, also enhance the activities of the Amh promoter and TESCO. In this study, we investigated the regulation of these sexual differentiation genes in Tokudaia osimensis, which lacks a Y chromosome and the SRY gene. The activity of the AMH promoter was stimulated by SOXE genes and SF1. Mutant AMH promoters, with mutations in its SOX and SF1 binding sites, did not show significant activity by SOX9 and SF1. These results indicate that AMH expression was regulated by the binding of SOX9 and SF1. By contrast, SOXE genes could not enhance TESCO activity. These results indicate that TESCO enhancer activity was lost in this species. Furthermore, the activity of the SOX9 promoter was enhanced by ER71, indicating that ER71 may play an important role in the testis-specific expression of SOX9.


Subject(s)
Murinae/genetics , SOX Transcription Factors/physiology , Sex Determination Processes , Testis/physiology , Animals , Gene Expression Regulation , Male , Murinae/metabolism , Murinae/physiology , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , Testis/metabolism , Y Chromosome
5.
Eur J Cell Biol ; 94(12): 547-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26340821

ABSTRACT

In metazoa, SOX family transcription factors play many diverse roles. In vertebrate, they are well-known regulators of numerous developmental processes. Wide-ranging studies have demonstrated the co-expression of SOX proteins in various developing tissues and that they occur in an overlapping manner and show functional redundancy. In particular, studies focusing on the HMG box of SOX proteins have revealed that the HMG box regulates DNA-binding properties, and mediates both the nucleocytoplasmic shuttling of SOX proteins and their physical interactions with partner proteins. Posttranslational modifications are further implicated in the regulation of the transcriptional activities of SOX proteins. In this review, we discuss the underlying molecular mechanisms involved in the SOX-partner factor interactions and the functional modes of SOX-partner complexes during development. We particularly emphasize the representative roles of the SOX group proteins in major tissues during developmental and physiological processes.


Subject(s)
SOX Transcription Factors/physiology , Active Transport, Cell Nucleus , Animals , Cardiovascular System/embryology , Cardiovascular System/growth & development , Cell Differentiation , Gene Expression Regulation, Developmental , Humans , Nervous System/embryology , Nervous System/growth & development , Protein Binding , Protein Processing, Post-Translational
6.
Mol Reprod Dev ; 81(12): 1103-14, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25394196

ABSTRACT

Early chick development is a systematic process governed by the concerted action of multiple mechanisms that regulate transcription and post-transcriptional processes. Post-transcriptional microRNA-mediated regulation, with regard to lineage specification and differentiation in early chick development, requires further investigation. Here, we characterize the transcriptional and post-transcriptional regulation mechanisms in undifferentiated chick blastodermal cells. Expression of the miR-302 cluster, POUV, SOX2, and STAT5B decreased in a time-dependent manner in early chick development. We found that POUV, SOX2, and STAT5B regulate the transcription of the miR-302 cluster, as its 5'-flanking region contains binding elements for each transcription factor. Additionally, POUV, SOX2, and STAT5B maintain pluripotency by regulating genes containing the miR-302 cluster target sequence. For example, microRNAs from the miR-302 cluster can bind to PBX3 and E2F7 transcripts, thus acting as a post-transcriptional regulator that maintains the undifferentiated state of blastodermal cells by balancing the expression of genes related to pluripotency and differentiation. Based on these results, we suggest that both transcriptional and post-transcriptional regulation of the miR302 cluster is critical for intrinsically controlling the undifferentiated state of chick embryonic blastodermal cells. These findings may help our understanding of the cellular and molecular mechanisms that underlie developmental decisions during early chick development.


Subject(s)
Chick Embryo/embryology , Gene Expression Regulation, Developmental/physiology , MicroRNAs/physiology , Models, Biological , Transcription Factors/physiology , Animals , Chick Embryo/metabolism , DNA Primers/genetics , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Luciferases , MicroRNAs/metabolism , RNA Interference/physiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , SOX Transcription Factors/physiology , STAT5 Transcription Factor/metabolism , Transcription Factors/metabolism
7.
Arterioscler Thromb Vasc Biol ; 34(11): 2372-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25169934

ABSTRACT

The establishment of arterial and venous identity of endothelial cells is critical for the proper anatomic configuration and function of the vascular tree. Arterial and venous specification of endothelial cells is determined by genetic factors, although surrounding cells and hemodynamic forces may also contribute to vascular remodeling. This review provides an overview of the signaling pathways and related transcription factors implicated in differentiation of endothelial cells. We will discuss, in particular, the role of upstream and downstream effectors of Wnt, Sox, and Notch pathways. The understanding of the molecular mechanisms that orchestrate endothelial differentiation may have therapeutic relevance for diseases such as atherosclerosis, arteriovenous malformations, aneurysms, and others.


Subject(s)
Arteries/physiology , Endothelium, Vascular/physiology , Signal Transduction/physiology , Veins/physiology , Animals , Hemodynamics/physiology , Humans , Receptors, Notch/physiology , SOX Transcription Factors/physiology , Vascular Remodeling/physiology , Wnt Proteins/physiology
8.
Curr Opin Hematol ; 21(3): 229-34, 2014 May.
Article in English | MEDLINE | ID: mdl-24626043

ABSTRACT

PURPOSE OF REVIEW: The development of a functionally and anatomically correct vascular network is a complex phenomenon that requires the combined activity of different signaling pathways and transcription factors. Notch signaling activation, for instance, is crucial for arterial specification. Here, we discuss the current knowledge on how other signaling pathways cooperate with Notch to orchestrate arterial differentiation of embryonic and postnatal vasculature. RECENT FINDINGS: The role of Notch in vascular development and arterial differentiation is well known. However, it was found that canonical Wnt signaling may act upstream of Notch, upregulating Dll4 and inducing endothelial cells to acquire arterial characteristics. Furthermore, the transcription factor Sox17 may act as a link between Wnt and Notch in the induction of a correct arterio/venous differentiation. SUMMARY: In the past years, the research on vascular development was mostly focused on the mechanisms that regulate vessel growth. We now understand that in order to interfere with several vascular diseases (e.g. aneurysm, cerebral ischemia and stroke) or tumor vascularization, we need to understand the signals that direct arterio/venous specification. Here, we discuss the interplay between Notch, Wnt and Sox that exert a combined positive action on arterial differentiation.


Subject(s)
Arteries/physiology , Gene Expression Regulation, Developmental/physiology , Receptors, Notch/physiology , SOX Transcription Factors/physiology , Signal Transduction/physiology , Wnt Signaling Pathway/physiology , Cell Differentiation/physiology , Endothelial Cells/physiology , Humans
9.
Development ; 140(5): 1090-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23364327

ABSTRACT

Endoderm and mesoderm are both formed upon activation of Nodal signaling but how endoderm differentiates from mesoderm is still poorly explored. The sox-related gene casanova (sox32) acts downstream of the Nodal signal, is essential for endoderm development and requires the co-factor Pou2 (Pou5f1, Oct3, Oct4) in this process. Conversely, BMP signals have been shown to inhibit endoderm development by an as yet unexplained mechanism. In a search for Casanova regulators in zebrafish, we identified two of its binding partners as the transcription factors Pou2 and Vox, a member of the Vent group of proteins also involved in the patterning of the gastrula. In overexpression studies we show that vox and/or Vent group genes inhibit the capacity of Casanova to induce endoderm, even in the presence of its co-factor Pou2, and that Vox acts as a repressor in this process. We further show that vox, but not other members of the Vent group, is essential for defining the proper endodermal domain size at gastrulation. In this process, vox acts downstream of BMPs. Cell fate analysis further shows that Vox plays a key role downstream of BMP signals in regulating the capacity of Nodal to induce endoderm versus mesoderm by modulating the activity of the Casanova/Pou2 regulatory system.


Subject(s)
Endoderm/embryology , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Octamer Transcription Factor-3/metabolism , Repressor Proteins/metabolism , Repressor Proteins/physiology , SOX Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Down-Regulation/genetics , Embryo, Nonmammalian , Endoderm/growth & development , Endoderm/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Nodal Signaling Ligands/genetics , Nodal Signaling Ligands/metabolism , Nodal Signaling Ligands/physiology , Octamer Transcription Factor-3/physiology , Protein Binding/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Repressor Proteins/chemistry , Repressor Proteins/genetics , SOX Transcription Factors/physiology , Sequence Deletion , Tissue Distribution , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
10.
BMC Dev Biol ; 13: 1, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23289785

ABSTRACT

BACKGROUND: Group B Sox domain transcription factors play important roles in metazoan central nervous system development. They are, however, difficult to study as mutations often have pleiotropic effects and other Sox family members can mask phenotypes due to functional compensation. In Drosophila melanogaster, the Sox gene Dichaete is dynamically expressed in the embryonic CNS, where it is known to have functional roles in neuroblasts and the ventral midline. In this study, we use inducible dominant negative proteins in combination with ChIP, immunohistochemistry and genome-wide expression profiling to further dissect the role of Dichaete in these two tissues. RESULTS: We generated two dominant negative Dichaete constructs, one lacking a DNA binding domain and the other fused to the Engrailed transcriptional repressor domain. We expressed these tissue-specifically in the midline and in neuroblasts using the UAS/GAL4 system, validating their use at the phenotypic level and with known target genes. Using ChIP and immunohistochemistry, we identified two new likely direct Dichaete target genes, commisureless in the midline and asense in the neuroectoderm. We performed genome-wide expression profiling in stage 8-9 embryos, identifying almost a thousand potential tissue-specific Dichaete targets, with half of these genes showing evidence of Dichaete binding in vivo. These include a number of genes with known roles in CNS development, including several components of the Notch, Wnt and EGFR signalling pathways. CONCLUSIONS: As well as identifying commisureless as a target, our data indicate that Dichaete helps establish its expression during early midline development but has less effect on its established later expression, highlighting Dichaete action on tissue specific enhancers. An analysis of the broader range of candidate Dichaete targets indicates that Dichaete plays diverse roles in CNS development, with the 500 or so Dichaete-bound putative targets including a number of transcription factors, signalling pathway components and terminal differentiation genes. In the early neurectoderm we implicate Dichaete in the lateral inhibition pathway and show that Dichaete acts to repress the proneural gene asense. Our analysis also reveals that dominant negatives cause off-target effects, highlighting the need to use other experimental data for validating findings from dominant negative studies.


Subject(s)
Central Nervous System/metabolism , Drosophila Proteins/metabolism , SOX Transcription Factors/metabolism , Animals , Binding Sites , Body Patterning , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster , Gene Expression Regulation, Developmental , Mutation , SOX Transcription Factors/genetics , SOX Transcription Factors/physiology
11.
Mol Psychiatry ; 17(3): 267-79, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21709683

ABSTRACT

The basic helix-loop-helix PAS (Per, Arnt, Sim) domain transcription factor gene NPAS3 is a replicated genetic risk factor for psychiatric disorders. A knockout (KO) mouse model exhibits behavioral and adult neurogenesis deficits consistent with human illness. To define the location and mechanism of NPAS3 etiopathology, we combined immunofluorescent, transcriptomic and metabonomic approaches. Intense Npas3 immunoreactivity was observed in the hippocampal subgranular zone-the site of adult neurogenesis--but was restricted to maturing, rather than proliferating, neuronal precursor cells. Microarray analysis of a HEK293 cell line over-expressing NPAS3 showed that transcriptional targets varied according to circadian rhythm context and C-terminal deletion. The most highly up-regulated NPAS3 target gene, VGF, encodes secretory peptides with established roles in neurogenesis, depression and schizophrenia. VGF was just one of many NPAS3 target genes also regulated by the SOX family of transcription factors, suggesting an overlap in neurodevelopmental function. The parallel repression of multiple glycolysis genes by NPAS3 reveals a second role in the regulation of glucose metabolism. Comparison of wild-type and Npas3 KO metabolite composition using high-resolution mass spectrometry confirmed these transcriptional findings. KO brain tissue contained significantly altered levels of NAD(+), glycolysis metabolites (such as dihydroxyacetone phosphate and fructose-1,6-bisphosphate), pentose phosphate pathway components and Kreb's cycle intermediates (succinate and α-ketoglutarate). The dual neurodevelopmental and metabolic aspects of NPAS3 activity described here increase our understanding of mental illness etiology, and may provide a mechanism for innate and medication-induced susceptibility to diabetes commonly reported in psychiatric patients.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Neurogenesis/genetics , Transcription Factors/physiology , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Chemistry , Circadian Rhythm , Dentate Gyrus/metabolism , Energy Metabolism/genetics , Glycolysis/genetics , HEK293 Cells/metabolism , Humans , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Recombinant Fusion Proteins/physiology , SOX Transcription Factors/physiology , Transcription Factors/genetics , Transcriptome
12.
Acta Biochim Biophys Sin (Shanghai) ; 43(5): 387-99, 2011 May.
Article in English | MEDLINE | ID: mdl-21467072

ABSTRACT

Sox superfamily proteins are DNA-binding transcriptional factors that contain highly conserved high-mobility group (HMG) box and take part in various development process. Sox31 is a maternal factor supplied in the oocyte and starts its zygotic expression during mid-blastula transition (MBT). From gastrulation stage, it mainly resides in neural tissue. Ectopically expression of Sox31 mRNA leads to cyclopia, fusion eyes, or totally loss of anterior head structure, in accompany with severe notochord defects. Molecular markers indicate that forebrain tissue reduces sharply while the posterior neural tissue expands anteriorly. In addition, organizer specification is also suppressed. Oppositely, an antisense morpholino designed functionally knockdown Sox31 causes typically dorsalized phenotype and reversed central nervous system (CNS) anteroposterior (AP) patterning. Gain of function with chimeric construct, where Sox31 HMG DNA binding domain is fused to a transcription activation domain (VP16) or transcription suppression domain (EnR), suggests that Sox31 acts as a transcriptional suppressor in vivo. The expression of Bozozok (Dharma), a direct target gene of pre-MBT Wnt/ß-catenin signal, is suppressed by Sox31. Thus, to unveil the relationship between Sox31 and ß-catenin-related transcriptional activity, we designed Top/Fop luciferase assay in HEK293T cells, and found that Sox31 could indeed suppress Tcf/Lef-dependent transcriptional activity without influencing the stability of ß-catenin. Moreover, post-MBT Wnt signal was reduced in Sox31 morphants corresponding to the suppressed hindbrain structure, while phenotypic defects caused by excessive Sox31 could be rescued by Wnt antagonist dkk1. Taken together, Sox31 functions as an essential CNS AP patterning determinant and coordinates the CNS AP patterning process with organizer specification.


Subject(s)
Body Patterning , Central Nervous System/embryology , SOX Transcription Factors/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Base Sequence , DNA Primers , Gene Knockdown Techniques , RNA, Messenger/genetics , SOX Transcription Factors/genetics , Zebrafish Proteins/genetics
13.
Am J Physiol Gastrointest Liver Physiol ; 300(4): G503-15, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21292996

ABSTRACT

The genetic mechanisms underlying tissue maintenance of the gastrointestinal tract are critical for the proper function of the digestive system under normal physiological stress. The identification of transcription factors and related signal transduction pathways that regulate stem cell maintenance and lineage allocation is attractive from a clinical standpoint in that it may provide targets for novel cell- or drug-based therapies. Sox [sex-determining region Y (Sry) box-containing] factors are a family of transcription factors that are emerging as potent regulators of stem cell maintenance and cell fate decisions in multiple organ systems and might provide valuable insight toward the understanding of these processes in endodermally derived tissues of the gastrointestinal tract. In this review, we focus on the known genetic functions of Sox factors and their roles in epithelial tissues of the esophagus, stomach, intestine, colon, pancreas, and liver. Additionally, we discuss pathological conditions in the gastrointestinal tract that are associated with a dysregulation of Sox factors. Further study of Sox factors and their role in gastrointestinal physiology and pathophysiology may lead to advances that facilitate control of tissue maintenance and development of advanced clinical therapies.


Subject(s)
Gastrointestinal Diseases/metabolism , Gastrointestinal Tract/physiology , SOX Transcription Factors/physiology , Animals , Cell Proliferation , Gastrointestinal Diseases/physiopathology , Humans
14.
Neuroscience ; 172: 329-41, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20951776

ABSTRACT

Adult zebrafish have the ability to recover from spinal cord injury and exhibit re-growth of descending axons from the brainstem to the spinal cord. We performed gene expression analysis using microarray to find damage-induced genes after spinal cord injury, and found that Sox11b mRNA is up-regulated at 11 days after injury. However, the functional relevance of Sox11b for regeneration is not known. Here, we report that the up-regulation of Sox11b mRNA after spinal cord injury is mainly localized in ependymal cells lining the central canal and in newly differentiating neuronal precursors or immature neurons. Using an in vivo morpholino-based gene knockout approach, we demonstrate that Sox11b is essential for locomotor recovery after spinal cord injury. In the injured spinal cord, expression of the neural stem cell associated gene Nestin, and the proneural gene Ascl1a (Mash1a), which are involved in the self-renewal and cell fate specification of endogenous neural stem cells, respectively, is regulated by Sox11b. Our data indicate that Sox11b promotes neuronal determination of endogenous stem cells and regenerative neurogenesis following spinal cord injury in the adult zebrafish. Enhancing Sox11b expression to promote proliferation and neurogenic determination of endogenous neural stem cells after injury may be a promising strategy in restorative therapy after spinal cord injury in mammals.


Subject(s)
Neurogenesis/genetics , Recovery of Function/genetics , Regeneration/genetics , SOX Transcription Factors/physiology , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Zebrafish Proteins/physiology , Animals , Disease Models, Animal , SOX Transcription Factors/genetics , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Zebrafish , Zebrafish Proteins/genetics
15.
Leuk Lymphoma ; 51(11): 1962-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20919851

ABSTRACT

SOX11 is a transcription factor involved in embryonic neurogenesis and tissue remodeling. Its role in lymphopoiesis is still unknown. Recent studies have shown the specific overexpression of SOX11 mRNA and nuclear protein in both cyclin D1-positive and -negative mantle cell lymphoma (MCL). In addition to MCL, SOX11 is strongly expressed in hairy cell leukemia, Burkitt lymphoma, and immature lymphocytic neoplasms. Expression of SOX11 in MCL is not only a new diagnostic marker, but may also carry information related to the clinical and biological behavior. Further study of the biology may reveal common pathways to neoplasia related to SOX11 expression.


Subject(s)
Lymphoma, Mantle-Cell/genetics , SOXC Transcription Factors/genetics , Animals , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, Mantle-Cell/mortality , SOX Transcription Factors/genetics , SOX Transcription Factors/physiology
16.
Med Sci (Paris) ; 26(4): 411-6, 2010 Apr.
Article in French | MEDLINE | ID: mdl-20412747

ABSTRACT

OCT4 encoded by pou5f1 is one of the most ancient and early transcription factors identified in the embryo. It has been longwise recognized as a gatekeeper for pluripotency of embryonic stem (ES) cell. Uncovered twenty years ago, its fame was built up from its key role in maintaining embryonic stem cell pluripotency in 1998. Since, OCT4 was reported to also instruct stem cell fate through a gene dosage effect. It reached recently a novel glorious hit with its master role in reprogramming somatic cells.


Subject(s)
Octamer Transcription Factor-3/physiology , Animals , Cell Differentiation , Embryonic Development/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/physiology , Fetal Heart/cytology , Fetal Heart/metabolism , Gene Dosage , Gene Expression Regulation, Developmental , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , Models, Biological , Myocardium/metabolism , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Paracrine Communication , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic/genetics , SOX Transcription Factors/genetics , SOX Transcription Factors/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/physiology
17.
BMC Biol ; 8: 5, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20089153

ABSTRACT

BACKGROUND: Dedifferentiation occurs naturally in mature cell types during epimorphic regeneration in fish and some amphibians. Dedifferentiation also occurs in the induction of pluripotent stem cells when a set of transcription factors (Oct4, Sox2, Klf4 and c-Myc) is over expressed in mature cell types. RESULTS: We hypothesised that there are parallels between dedifferentiation or reprogramming of somatic cells to induced pluripotent stem cells and the natural process of dedifferentiation during epimorphic regeneration. We analysed expression levels of the most commonly used pluripotency associated factors in regenerating and non-regenerating tissue and compared them with levels in a pluripotent reference cell. We found that some of the pluripotency associated factors (oct4/pou5f1, sox2, c-myc, klf4, tert, sall4, zic3, dppa2/4 and fut1, a homologue of ssea1) were expressed before and during regeneration and that at least two of these factors (oct4, sox2) were also required for normal fin regeneration in the zebrafish. However these factors were not upregulated during regeneration as would be expected if blastema cells acquired pluripotency. CONCLUSIONS: By comparing cells from the regeneration blastema with embryonic pluripotent reference cells we found that induced pluripotent stem and blastema cells do not share pluripotency. However, during blastema formation some of the key reprogramming factors are both expressed and are also required for regeneration to take place. We therefore propose a link between partially reprogrammed induced pluripotent stem cells and the half way state of blastema cells and suggest that a common mechanism might be regulating these two processes.


Subject(s)
Cell Dedifferentiation/physiology , Cellular Reprogramming/physiology , Regeneration/physiology , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Dedifferentiation/genetics , Cellular Reprogramming/genetics , Electroporation , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Flow Cytometry , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , In Situ Hybridization , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/physiology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Regeneration/genetics , SOX Transcription Factors/genetics , SOX Transcription Factors/physiology , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
18.
Dev Biol ; 338(2): 262-9, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20025865

ABSTRACT

Sox2 has been variously implicated in maintenance of pluripotent stem cells or, alternatively, early stages of cell differentiation, depending on context. In the developing inner ear, Sox2 initially marks all cells in the nascent sensory epithelium and, in mouse, is required for sensory epithelium formation. Sox2 is eventually downregulated in hair cells but is maintained in support cells, the functional significance of which is unknown. Here we describe regulation and function of sox2 in the zebrafish inner ear. Expression of sox2 begins after the onset of sensory epithelium development and is regulated by Atoh1a/b, Fgf and Notch. Knockdown of sox2 does not prevent hair cell production, but the rate of accumulation is reduced due to sporadic death of differentiated hair cells. We next tested the capacity for hair cell regeneration following laser ablation of mature brn3c:gfp-labeled hair cells. In control embryos, regeneration of lost hair cells begins by 12 h post-ablation and involves transdifferentiation of support cells rather than asymmetric cell division. In contrast, regeneration does not occur in sox2-depleted embryos. These data show that zebrafish sox2 is required for hair cell survival, as well as for transdifferentiation of support cells into hair cells during regeneration.


Subject(s)
Ear, Inner/cytology , Hair Cells, Auditory/cytology , Regeneration , SOX Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Cell Differentiation , Cell Survival , Gene Expression Regulation , SOX Transcription Factors/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/physiology , Zebrafish , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...