Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
PLoS Genet ; 19(8): e1010874, 2023 08.
Article in English | MEDLINE | ID: mdl-37594984

ABSTRACT

In the mammalian cerebral cortex, the hippocampal primordium (Hcp) occupies a discrete position in the dorsal telencephalic neuroepithelium adjacent to the neocortical primordium (Ncp). We examined transcriptomic and chromatin-level features that distinguish the Hcp from the Ncp in the mouse during the early neurogenic period, embryonic day (E)12.5. ATAC-seq revealed that the Hcp was more accessible than the Ncp at this stage. Motif analysis of the differentially accessible loci in these tissues revealed LHX2 as a candidate transcription factor for modulating gene regulatory networks (GRNs). We analyzed LHX2 occupancy profiles and compared these with transcriptomic data from control and Lhx2 mutant Hcp and Ncp at E12.5. Our results revealed that LHX2 directly regulates distinct genes in the Hcp and Ncp within a set of common pathways that control fundamental aspects of development namely pluripotency, axon pathfinding, Wnt, and Hippo signaling. Loss of Lhx2 caused a decrease in accessibility, specifically in hippocampal chromatin, suggesting that this factor may play a unique role in hippocampal development. We identified 14 genes that were preferentially enriched in the Hcp, for which LHX2 regulates both chromatin accessibility and mRNA expression, which have not thus far been examined in hippocampal development. Together, these results provide mechanistic insight into how LHX2 function in the Hcp may contribute to the process by which the hippocampus acquires features distinct from the neocortex.


Subject(s)
Chromatin , Neocortex , Animals , Mice , Hippocampus , LIM-Homeodomain Proteins , Mammals , Transcription Factors , Transcriptome
2.
J Mol Evol ; 91(5): 616-627, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37341745

ABSTRACT

Hox genes encode Homeodomain-containing transcription factors, which specify segmental identities along the anterior-posterior axis. Functional changes in Hox genes have been directly implicated in the evolution of body plans across the metazoan lineage. The Hox protein Ultrabithorax (Ubx) is expressed and required in developing third thoracic (T3) segments in holometabolous insects studied so far, particularly, of the order Coleoptera, Lepidoptera and Diptera. Ubx function is key to specify differential development of the second (T2) and T3 thoracic segments in these insects. While Ubx is expressed in the third thoracic segment in developing larvae of Hymenopteran Apis mellifera, the morphological differences between T2 and T3 are subtle. To identify evolutionary changes that are behind the differential function of Ubx in Drosophila and Apis, which are diverged for more than 350 million years, we performed comparative analyses of genome wide Ubx-binding sites between these two insects. Our studies reveal that a motif with a TAAAT core is a preferred binding site for Ubx in Drosophila, but not in Apis. Biochemical and transgenic assays suggest that in Drosophila, the TAAAT core sequence in the Ubx binding sites is required for Ubx-mediated regulation of two of its target genes studied here; CG13222, a gene that is normally upregulated by Ubx and vestigial (vg), whose expression is repressed by Ubx in T3. Interestingly, changing the TAAT site to a TAAAT site was sufficient to bring an otherwise unresponsive enhancer of the vg gene from Apis under the control of Ubx in a Drosophila transgenic assay. Taken together, our results suggest an evolutionary mechanism by which critical wing patterning genes might have come under the regulation of Ubx in the Dipteran lineage.

3.
Immunology ; 156(4): 384-401, 2019 04.
Article in English | MEDLINE | ID: mdl-30556901

ABSTRACT

We have previously demonstrated co-receptor level-associated functional heterogeneity in apparently homogeneous naive peripheral CD4 T cells, dependent on MHC-mediated tonic signals. Maturation pathways can differ between naive CD4 and naive CD8 cells, so we tested whether the latter showed similar co-receptor level-associated functional heterogeneity. We report that, when either polyclonal and T-cell receptor (TCR)-transgenic monoclonal peripheral naive CD8 T cells from young mice were separated into CD8hi and CD8lo subsets, CD8lo cells responded poorly, but CD8hi and CD8lo subsets of CD8 single-positive (SP) thymocytes responded similarly. CD8lo naive CD8 T cells were smaller and showed lower levels of some cell-surface molecules, but higher levels of the negative regulator CD5. In addition to the expected peripheral decline in CD8 levels on transferred naive CD8 T cells in wild-type (WT) but not in MHC class I-deficient recipient mice, short-duration naive T-cell-dendritic cell (DC) co-cultures in vitro also caused co-receptor down-modulation in CD8 T cells but not in CD4 T cells. Constitutive pZAP70/pSyk and pERK levels ex vivo were lower in CD8lo naive CD8 T cells and dual-specific phosphatase inhibition partially rescued their hypo-responsiveness. Bulk mRNA sequencing showed major differences in the transcriptional landscapes of CD8hi and CD8lo naive CD8 T cells. CD8hi naive CD8 T cells showed enrichment of genes involved in positive regulation of cell cycle and survival. Our data show that naive CD8 T cells show major differences in their signaling, transcriptional and functional landscapes associated with subtly altered CD8 levels, consistent with the possibility of peripheral cellular aging.


Subject(s)
CD8 Antigens/immunology , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Transcriptome , Adult , Animals , Cellular Senescence/immunology , Female , Healthy Volunteers , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Young Adult
4.
J Neurosci ; 37(46): 11245-11254, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29025924

ABSTRACT

Regulation of the neuron-glia cell-fate switch is a critical step in the development of the CNS. Previously, we demonstrated that Lhx2 is a necessary and sufficient regulator of this process in the mouse hippocampal primordium, such that Lhx2 overexpression promotes neurogenesis and suppresses gliogenesis, whereas loss of Lhx2 has the opposite effect. We tested a series of transcription factors for their ability to mimic Lhx2 overexpression and suppress baseline gliogenesis, and also to compensate for loss of Lhx2 and suppress the resulting enhanced level of gliogenesis in the hippocampus. Here, we demonstrate a novel function of Dmrt5/Dmrta2 as a neurogenic factor in the developing hippocampus. We show that Dmrt5, as well as known neurogenic factors Neurog2 and Pax6, can each not only mimic Lhx2 overexpression, but also can compensate for loss of Lhx2 to different extents. We further uncover a reciprocal regulatory relationship between Dmrt5 and Lhx2, such that each can compensate for loss of the other. Dmrt5 and Lhx2 also have opposing regulatory control on Pax6 and Neurog2, indicating a complex bidirectionally regulated network that controls the neuron-glia cell-fate switch.SIGNIFICANCE STATEMENT We identify Dmrt5 as a novel regulator of the neuron-glia cell-fate switch in the developing hippocampus. We demonstrate Dmrt5 to be neurogenic, and reciprocally regulated by Lhx2: loss of either factor promotes gliogenesis; overexpression of either factor suppresses gliogenesis and promotes neurogenesis; each can substitute for loss of the other. Furthermore, each factor has opposing effects on established neurogenic genes Neurog2 and Pax6 Dmrt5 is known to suppress their expression, and we show that Lhx2 is required to maintain it. Our study reveals a complex regulatory network with bidirectional control of a fundamental feature of CNS development, the control of the production of neurons versus astroglia in the developing hippocampus.Finally, we confirm that Lhx2 binds a highly conserved putative enhancer of Dmrt5, suggesting an evolutionarily conserved regulatory relationship between these factors. Our findings uncover a complex network that involves Lhx2, Dmrt5, Neurog2, and Pax6, and that ensures the appropriate amount and timing of neurogenesis and gliogenesis in the developing hippocampus.


Subject(s)
Hippocampus/physiology , LIM-Homeodomain Proteins/physiology , Neurogenesis/physiology , Neuroglia/physiology , Neurons/physiology , Transcription Factors/physiology , Animals , Base Sequence , Cell Differentiation/physiology , Cells, Cultured , Female , Hippocampus/cytology , Hippocampus/embryology , Male , Mice , Mice, Transgenic , Pregnancy
5.
J Neurosci ; 37(1): 194-203, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28053041

ABSTRACT

In the developing cerebral cortex, sequential transcriptional programs take neuroepithelial cells from proliferating progenitors to differentiated neurons with unique molecular identities. The regulatory changes that occur in the chromatin of the progenitors are not well understood. During deep layer neurogenesis, we show that transcription factor LHX2 binds to distal regulatory elements of Fezf2 and Sox11, critical determinants of neuron subtype identity in the mouse neocortex. We demonstrate that LHX2 binds to the nucleosome remodeling and histone deacetylase histone remodeling complex subunits LSD1, HDAC2, and RBBP4, which are proximal regulators of the epigenetic state of chromatin. When LHX2 is absent, active histone marks at the Fezf2 and Sox11 loci are increased. Loss of LHX2 produces an increase, and overexpression of LHX2 causes a decrease, in layer 5 Fezf2 and CTIP2-expressing neurons. Our results provide mechanistic insight into how LHX2 acts as a necessary and sufficient regulator of genes that control cortical neuronal subtype identity. SIGNIFICANCE STATEMENT: The functional complexity of the cerebral cortex arises from an array of distinct neuronal subtypes with unique connectivity patterns that are produced from common progenitors. This study reveals that transcription factor LHX2 regulates the numbers of specific cortical output neuron subtypes by controlling the genes that are required to produce them. Loss or increase in LHX2 during neurogenesis is sufficient to increase or decrease, respectively, a particular subcerebrally projecting population. Mechanistically, LHX2 interacts with chromatin modifying protein complexes to edit the chromatin landscape of its targets Fezf2 and Sox11, which regulates their expression and consequently the identities of the neurons produced. Thus, LHX2 is a key component of the control network for producing neurons that will participate in cortical circuitry.


Subject(s)
Cerebral Cortex/cytology , DNA-Binding Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , SOXC Transcription Factors/metabolism , Transcription Factors/metabolism , Animals , Cerebral Cortex/diagnostic imaging , Chromatin/genetics , Epigenesis, Genetic , Female , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Male , Mice , Nucleosomes/metabolism , Pregnancy
6.
Oxf Open Neurosci ; 3: kvae001, 2024.
Article in English | MEDLINE | ID: mdl-38595939

ABSTRACT

PRDM16 is a dynamic transcriptional regulator of various stem cell niches, including adipocytic, hematopoietic, cardiac progenitors, and neural stem cells. PRDM16 has been suggested to contribute to 1p36 deletion syndrome, one of the most prevalent subtelomeric microdeletion syndromes. We report a patient with a de novo nonsense mutation in the PRDM16 coding sequence, accompanied by lissencephaly and microcephaly features. Human stem cells were genetically modified to mimic this mutation, generating cortical organoids that exhibited altered cell cycle dynamics. RNA sequencing of cortical organoids at day 32 unveiled changes in cell adhesion and WNT-signaling pathways. ChIP-seq of PRDM16 identified binding sites in postmortem human fetal cortex, indicating the conservation of PRDM16 binding to developmental genes in mice and humans, potentially at enhancer sites. A shared motif between PRDM16 and LHX2 was identified and further examined through comparison with LHX2 ChIP-seq data from mice. These results suggested a collaborative partnership between PRDM16 and LHX2 in regulating a common set of genes and pathways in cortical radial glia cells, possibly via their synergistic involvement in cortical development.

7.
bioRxiv ; 2023 Aug 12.
Article in English | MEDLINE | ID: mdl-37609127

ABSTRACT

PRDM16 is a dynamic transcriptional regulator of various stem cell niches, including adipocytic, hematopoietic, cardiac progenitors, and neural stem cells. PRDM16 has been suggested to contribute to 1p36 deletion syndrome, one of the most prevalent subtelomeric microdeletion syndromes. We report a patient with a de novo nonsense mutation in the PRDM16 coding sequence, accompanied by lissencephaly and microcephaly features. Human stem cells were genetically modified to mimic this mutation, generating cortical organoids that exhibited altered cell cycle dynamics. RNA sequencing of cortical organoids at day 32 unveiled changes in cell adhesion and WNT-signaling pathways. ChIP-seq of PRDM16 identified binding sites in postmortem human fetal cortex, indicating the conservation of PRDM16 binding to developmental genes in mice and humans, potentially at enhancer sites. A shared motif between PRDM16 and LHX2 was identified and further examined through comparison with LHX2 ChIP-seq data from mice. These results suggested a collaborative partnership between PRDM16 and LHX2 in regulating a common set of genes and pathways in cortical radial glia cells, possibly via their synergistic involvement in cortical development.

8.
Cell Stem Cell ; 29(7): 1102-1118.e8, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35803228

ABSTRACT

The embryo instructs the allocation of cell states to spatially regulate functions. In the blastocyst, patterning of trophoblast (TR) cells ensures successful implantation and placental development. Here, we defined an optimal set of molecules secreted by the epiblast (inducers) that captures in vitro stable, highly self-renewing mouse trophectoderm stem cells (TESCs) resembling the blastocyst stage. When exposed to suboptimal inducers, these stem cells fluctuate to form interconvertible subpopulations with reduced self-renewal and facilitated differentiation, resembling peri-implantation cells, known as TR stem cells (TSCs). TESCs have enhanced capacity to form blastoids that implant more efficiently in utero due to inducers maintaining not only local TR proliferation and self-renewal, but also WNT6/7B secretion that stimulates uterine decidualization. Overall, the epiblast maintains sustained growth and decidualization potential of abutting TR cells, while, as known, distancing imposed by the blastocyst cavity differentiates TR cells for uterus adhesion, thus patterning the essential functions of implantation.


Subject(s)
Embryo Implantation , Placenta , Animals , Blastocyst , Female , Germ Layers , Mice , Pregnancy , Stem Cells , Trophoblasts/metabolism
9.
Nat Commun ; 12(1): 6094, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667153

ABSTRACT

Zygotic genome activation (ZGA) initiates regionalized transcription underlying distinct cellular identities. ZGA is dependent upon dynamic chromatin architecture sculpted by conserved DNA-binding proteins. However, the direct mechanistic link between the onset of ZGA and the tissue-specific transcription remains unclear. Here, we have addressed the involvement of chromatin organizer Satb2 in orchestrating both processes during zebrafish embryogenesis. Integrative analysis of transcriptome, genome-wide occupancy and chromatin accessibility reveals contrasting molecular activities of maternally deposited and zygotically synthesized Satb2. Maternal Satb2 prevents premature transcription of zygotic genes by influencing the interplay between the pluripotency factors. By contrast, zygotic Satb2 activates transcription of the same group of genes during neural crest development and organogenesis. Thus, our comparative analysis of maternal versus zygotic function of Satb2 underscores how these antithetical activities are temporally coordinated and functionally implemented highlighting the evolutionary implications of the biphasic and bimodal regulation of landmark developmental transitions by a single determinant.


Subject(s)
Matrix Attachment Region Binding Proteins/metabolism , Transcription Factors/metabolism , Vertebrates/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , Embryonic Development , Female , Gene Expression Regulation, Developmental , Male , Matrix Attachment Region Binding Proteins/genetics , Transcription Factors/genetics , Transcriptome , Vertebrates/genetics , Vertebrates/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zygote/metabolism
10.
J Biosci ; 452020.
Article in English | MEDLINE | ID: mdl-31965986

ABSTRACT

Multicellular organisms have evolved sophisticated mechanisms for responding to various developmental, environmental and physical stimuli by regulating transcription. The correlation of distribution of RNA Polymerase II (RNA Pol II) with transcription is well established in higher metazoans, however genome-wide information about its distribution in early metazoans, such as Hydra, is virtually absent. To gain insights into RNA Pol II-mediated transcription and chromatin organization in Hydra, we performed chromatin immunoprecipitation (ChIP)-coupled high-throughput sequencing (ChIP-seq) for RNA Pol II and Histone H3. Strikingly, we found that Hydra RNA Pol II is uniformly distributed across the entire gene body, as opposed to its counterparts in bilaterians such as human and mouse. Furthermore, correlation with transcriptome data revealed that the levels of RNA Pol II correlate with the magnitude of gene expression. Strikingly, the characteristic peak of RNA Pol II pause typically observed in bilaterians at the transcription start sites (TSSs) was not observed in Hydra. The RNA Pol II traversing ratio in Hydra was found to be intermediate to yeast and bilaterians. The search for factors involved in RNA Pol II pause revealed that RNA Pol II pausing machinery was most likely acquired first in Cnidaria. However, only a small subset of genes exhibited the promoter proximal RNP Pol II pause. Interestingly, the nucleosome occupancy is highest over the subset of paused genes as compared to total Hydra genes, which is another indication of paused RNA Pol II at these genes. Thus, this study provides evidence for the molecular basis of RNA Pol II pause early during the evolution of multicellular organisms.


Subject(s)
Chromatin/genetics , Evolution, Molecular , Hydra/genetics , RNA Polymerase II/genetics , Animals , Chromatin/ultrastructure , Gene Expression Regulation/genetics , High-Throughput Nucleotide Sequencing , Histones/genetics , Humans , Mice , Promoter Regions, Genetic , Transcriptome/genetics
11.
Commun Biol ; 2: 434, 2019.
Article in English | MEDLINE | ID: mdl-31799436

ABSTRACT

Wnt/ß-catenin signalling has been shown to play a critical role during head organizer formation in Hydra. Here, we characterized the Wnt signalling regulatory network involved in formation of the head organizer. We found that Wnt signalling regulates genes that are important in tissue morphogenesis. We identified that majority of transcription factors (TFs) regulated by Wnt/ß-catenin signalling belong to the homeodomain and forkhead families. Silencing of Margin, one of the Wnt regulated homeodomain TFs, results in loss of the ectopic tentacle phenotype typically seen upon activation of Wnt signalling. Furthermore, we show that the Margin promoter is directly bound and regulated by ß-catenin. Ectopic expression of Margin in zebrafish embryos results in body axis abnormalities suggesting that Margin plays a role in axis patterning. Our findings suggest that homeobox TFs came under the regulatory umbrella of Wnt/ß-catenin signalling presumably resulting in the evolution of primary body axis in animal phyla.


Subject(s)
Body Patterning/genetics , Gene Expression Regulation, Developmental , Hydra/embryology , Hydra/physiology , Wnt Signaling Pathway , Animals , Computational Biology/methods , Data Curation , Gene Expression Profiling , Transcription Factors/metabolism , Transcriptome , Wnt Proteins/metabolism
12.
Mol Cell Biol ; 38(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30181396

ABSTRACT

The ubiquitous transcription factor specificity protein 1 (SP1) is heavily modified posttranslationally. These modifications are critical for switching its functions and modulation of its transcriptional activity and DNA binding and stability. However, the mechanism governing the stability of SP1 by cellular signaling pathways is not well understood. Here, we provide biochemical and functional evidence that SP1 is an integral part of the Wnt signaling pathway. We identified a phosphodegron motif in SP1 that is specific to mammals. In the absence of Wnt signaling, glycogen synthase kinase 3ß (GSK3ß)-mediated phosphorylation and ß-TrCP E3 ubiquitin ligase-mediated ubiquitination are required to induce SP1 degradation. When Wnt signaling is on, SP1 is stabilized in a ß-catenin-dependent manner. SP1 directly interacts with ß-catenin, and Wnt signaling induces the stabilization of SP1 by impeding its interaction with ß-TrCP and axin1, components of the destruction complex. Wnt signaling suppresses ubiquitination and subsequent proteosomal degradation of SP1. Furthermore, SP1 regulates Wnt-dependent stability of ß-catenin and their mutual stabilization is critical for target gene expression, suggesting a feedback mechanism. Upon stabilization, SP1 and ß-catenin cooccupy the promoters of TCFL2/ß-catenin target genes. Collectively, this study uncovers a direct link between SP1 and ß-catenin in the Wnt signaling pathway.


Subject(s)
Axin Signaling Complex/genetics , Sp1 Transcription Factor/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Amino Acid Sequence , Animals , Cell Line , Cell Line, Tumor , Glycogen Synthase Kinase 3 beta/genetics , HCT116 Cells , HEK293 Cells , Humans , Phosphorylation/genetics , Sequence Alignment , Transcription, Genetic/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , beta-Transducin Repeat-Containing Proteins/genetics
13.
Dev Cell ; 43(2): 198-211.e12, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29033362

ABSTRACT

Cell-cell contact formation constitutes an essential step in evolution, leading to the differentiation of specialized cell types. However, remarkably little is known about whether and how the interplay between contact formation and fate specification affects development. Here, we identify a positive feedback loop between cell-cell contact duration, morphogen signaling, and mesendoderm cell-fate specification during zebrafish gastrulation. We show that long-lasting cell-cell contacts enhance the competence of prechordal plate (ppl) progenitor cells to respond to Nodal signaling, required for ppl cell-fate specification. We further show that Nodal signaling promotes ppl cell-cell contact duration, generating a positive feedback loop between ppl cell-cell contact duration and cell-fate specification. Finally, by combining mathematical modeling and experimentation, we show that this feedback determines whether anterior axial mesendoderm cells become ppl or, instead, turn into endoderm. Thus, the interdependent activities of cell-cell signaling and contact formation control fate diversification within the developing embryo.


Subject(s)
Cell Communication , Cell Lineage , Feedback, Physiological , Gastrula/metabolism , Morphogenesis/physiology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Body Patterning , Cell Differentiation , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryonic Development , Gastrula/growth & development , Gastrulation/physiology , Gene Expression Regulation, Developmental , Models, Theoretical , Nodal Protein/genetics , Nodal Protein/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics
14.
Cell Rep ; 16(3): 866-77, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27396324

ABSTRACT

During metazoan development, the temporal pattern of morphogen signaling is critical for organizing cell fates in space and time. Yet, tools for temporally controlling morphogen signaling within the embryo are still scarce. Here, we developed a photoactivatable Nodal receptor to determine how the temporal pattern of Nodal signaling affects cell fate specification during zebrafish gastrulation. By using this receptor to manipulate the duration of Nodal signaling in vivo by light, we show that extended Nodal signaling within the organizer promotes prechordal plate specification and suppresses endoderm differentiation. Endoderm differentiation is suppressed by extended Nodal signaling inducing expression of the transcriptional repressor goosecoid (gsc) in prechordal plate progenitors, which in turn restrains Nodal signaling from upregulating the endoderm differentiation gene sox17 within these cells. Thus, optogenetic manipulation of Nodal signaling identifies a critical role of Nodal signaling duration for organizer cell fate specification during gastrulation.


Subject(s)
Body Patterning/physiology , Gastrulation/physiology , Nodal Protein/metabolism , SOXF Transcription Factors/metabolism , Signal Transduction/physiology , Zebrafish Proteins/metabolism , Animals , Base Sequence , Body Patterning/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Embryo, Nonmammalian/physiology , Endoderm/metabolism , Endoderm/physiology , Gastrulation/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Morphogenesis/genetics , Morphogenesis/physiology , Optogenetics/methods , Signal Transduction/genetics , Transcription, Genetic/genetics , Up-Regulation/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/physiology , Zebrafish Proteins/genetics
15.
J Biosci ; 40(5): 891-907, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26648035

ABSTRACT

The Asian elephant Elephas maximus and the African elephant Loxodonta africana that diverged 5-7 million years ago exhibit differences in their physiology, behaviour and morphology. A comparative genomics approach would be useful and necessary for evolutionary and functional genetic studies of elephants. We performed sequencing of E. maximus and map to L. africana at ~15X coverage. Through comparative sequence analyses, we have identified Asian elephant specific homozygous, non-synonymous single nucleotide variants (SNVs) that map to 1514 protein coding genes, many of which are involved in olfaction. We also present the first report of a high-coverage transcriptome sequence in E. maximus from peripheral blood lymphocytes. We have identified 103 novel protein coding transcripts and 66-long non-coding (lnc)RNAs. We also report the presence of 181 protein domains unique to elephants when compared to other Afrotheria species. Each of these findings can be further investigated to gain a better understanding of functional differences unique to elephant species, as well as those unique to elephantids in comparison with other mammals. This work therefore provides a valuable resource to explore the immense research potential of comparative analyses of transcriptome and genome sequences in the Asian elephant.


Subject(s)
Elephants/genetics , Genetic Variation , Transcriptome , Animals , Gene Expression , Gene Expression Profiling , Genome , Homozygote , Lymphocytes/physiology , Molecular Sequence Annotation , RNA, Long Noncoding , Sequence Analysis
16.
Article in English | MEDLINE | ID: mdl-26388940

ABSTRACT

BACKGROUND: Role of epigenetic mechanisms towards regulation of the complex life cycle/pathogenesis of Plasmodium falciparum, the causative agent of malaria, has been poorly understood. To elucidate stage-specific epigenetic regulation, we performed genome-wide mapping of multiple histone modifications of P. falciparum. Further to understand the differences in transcription regulation in P. falciparum and its host, human, we compared their histone modification profiles. RESULTS: Our comprehensive comparative analysis suggests distinct mode of transcriptional regulation in malaria parasite by virtue of poised genes and differential histone modifications. Furthermore, analysis of histone modification profiles predicted 562 genes producing anti-sense RNAs and 335 genes having bidirectional promoter activity, which raises the intriguing possibility of RNA-mediated regulation of transcription in P. falciparum. Interestingly, we found that H3K36me2 acts as a global repressive mark and gene regulation is fine tuned by the ratio of activation marks to H3K36me2 in P. falciparum. This novel mechanism of gene regulation is supported by the fact that knockout of SET genes (responsible for H3K36 methylation) leads to up-regulation of genes with highest occupancy of H3K36me2 in wild-type P. falciparum. Moreover, virulence (var) genes are mostly poised and marked by a unique set of activation (H4ac) and repression (H3K9me3) marks, which are mutually exclusive to other Plasmodium housekeeping genes. CONCLUSIONS: Our study reveals unique plasticity in the epigenetic regulation in P. falciparum which can influence parasite virulence and pathogenicity. The observed differences in the histone code and transcriptional regulation in P. falciparum and its host will open new avenues for epigenetic drug development against malaria parasite.

17.
Sci Rep ; 4: 6076, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25123547

ABSTRACT

In this study, we have investigated genome-wide occurrence of Histone Acetyltransferases (HATs) in genomes of Mus musculus and Danio rerio on the basis of presence of HAT domain. Our study identified a group of proteins that lacks characteristic features of known HAT families, relatively smaller in size and has no other associated domains. Most of the proteins in this unclassified group are Camello proteins, which are not yet known and classified as functional HATs. Our in vitro and in vivo analysis revealed that Camello family proteins are active HATs and exhibit specificity towards histone H4. Interestingly, Camello proteins are among the first identified HATs showing perinuclear localization. Moreover, Camello proteins are evolutionarily conserved in all chordates and are observed for the first time in cnidarians in phylogeny. Furthermore, knockdown of Camello protein (CMLO3) in zebrafish embryos exhibited defects in axis elongation and head formation. Thus, our study identified a novel family of active HATs that is specific for histone H4 acetylation, exhibits perinuclear localization and is essential for zebrafish development.


Subject(s)
Body Patterning/genetics , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/metabolism , Zebrafish/embryology , Acetylation , Amino Acid Sequence , Animals , Cell Line, Tumor , Gene Knockdown Techniques , HeLa Cells , Humans , Lysine/chemistry , Mice , Molecular Sequence Data , Morpholinos/genetics , Protein Processing, Post-Translational , Sequence Alignment , Zinc Fingers/genetics
18.
Curr Drug Targets ; 13(13): 1603-15, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22998183

ABSTRACT

Cancer progression and metastasis involve series of alterations in the expression of multitude of genes. The structure and organization of chromatin play an important role in spatial arrangement of genes inside the nucleus thereby allowing different machineries to activate or silence the transcription of genes governed by various epigenetic events. Epigenetic modifications and dynamic changes in chromatin organization by organizer proteins have recently been shown to play an instrumental role in regulating cancer-promoting genes. Special AT-rich binding protein (SATB1) is a unique type of global regulator that integrates higher-order chromatin organization with regulation of gene expression. Aberrant expression of SATB1 has been shown to promote breast, hepatocellular, prostate and various other cancers. In this review we highlight upon the role of SATB1in chromatin organization and as global regulator of gene expression during cancer development. The expression of SATB1 progressively increases with the progression of cancers and it dynamically reprograms the expression of genes that are involved in epithelial-mesenchymal transition. SATB1 directly regulates the expression of ERRB2, MMP2, ABL1, E-cadherin and hence acts as key regulator in cancer development. Understanding the molecular mechanisms of regulation of SATB1 expression would therefore be extremely essential towards designing strategies to control it. Recent studies have provided important insights into regulation of SATB1 by FOXP3 and microRNAs. In this review we evaluate the potential of SATB1 as molecular target for cancer therapy.


Subject(s)
Chromatin/metabolism , Gene Targeting/trends , Genetic Therapy/trends , Matrix Attachment Region Binding Proteins/metabolism , Neoplasms/therapy , Animals , Chromatin/genetics , Humans , Matrix Attachment Region Binding Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
19.
Exp Ther Med ; 1(6): 915-920, 2010 Nov.
Article in English | MEDLINE | ID: mdl-22993618

ABSTRACT

Malignant melanoma is one of the most common types of cancer in the US and worldwide. The epidemiological data suggest that dietary modification may reduce the incidence of this disease. Quercetin (3,5,7,3',4'-tetrahydroxyflavone), a flavonoid isolated from onion, exhibits anti-oxidant, anti-inflammatory and anti-cancer effects. D,L-sulforaphane [1-isothiocyanato-4-(methylsulfinyl)-butane], a cruciferous vegetable-derived isomer isolated from broccoli, is highly effective in protection against cancer. Matrix metalloproteinases (MMPs), extracellular matrix degrading enzymes, are involved in embryogenesis, inflammation, angiogenesis and cancer. MMP-9 in particular plays a crucial role in the regulation of invasion, tumor growth and metastasis. Previous studies have reported that both quercetin and sulforaphane independently reduce tumor growth and metastasis in breast, prostate, lung and other types of cancers. However, the combined effects of quercetin and sulforaphane on the regulation of tumor growth and the mechanism(s) of actions underlying this process have not yet been investigated. In the present study, we report for the first time that quercetin and sulforaphane in combination inhibit the proliferation and migration of melanoma (B16F10) cells more effectively than either compound used alone. Moreover, these compounds in combination significantly suppressed melanoma growth as compared to their individual use in a mouse model. This combined effect was predominantly due to a decrease in MMP-9 expression in the mouse tumors. Taken together, our findings revealed that the administration of quercetin and sulforaphane in combination rather than alone may be a more effective approach for the treatment of malignant melanoma.

SELECTION OF CITATIONS
SEARCH DETAIL