Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Med Sci Monit ; 26: e925482, 2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32876075

ABSTRACT

BACKGROUND Proliferation and migration play crucial roles in various physiological processes, especially in injured endothelial repair. Endothelial progenitor cells (EPCs), as the precursors of endothelial cell, are involved in the regeneration of the endothelial lining of blood vessels. Furthermore, EPCs were found to be a potential choice for venous thrombosis (VT) treatment. MATERIAL AND METHODS EPCs were isolated from human peripheral blood of healthy adults and VT patients. Differently expressed micro(mi)RNAs were examined by quantitative real-time polymerase chain reaction, after which proliferative capacity and migration effect were tested by Cell-Counting Kit 8, scratch wound assay, and transwell assays. Bioinformatic analysis was applied to investigate the potential target messenger ribonucleic acid and a dual-luciferase reporting system was utilized to confirm the binding of miR-22-3p to its target gene. Western blot was carried out to detect candidate protein expression level. Finally, miR-22-3p expression was monitored in VT patients during follow-up to assess its correlation with prognosis of VT. RESULTS Our data revealed that miR-22-3p was upregulated in EPCs derived from deep VT (DVT) individuals and suppression of miR-22-3p contributed to proliferation and migration of EPCs. In addition, miR-22-3p/onecut 1 (OC1)/vascular endothelial growth factor A (VEGFA) signaling pathway was involved in regulating EPC migration and proliferation. In addition, lower expression of miR-22-3p in DVT patients indicated decreased risk of VT recurrence. CONCLUSIONS Our results suggest that miR-22-3p regulates OC1/VEGFA signaling and is involved in regulating EPC proliferation and migration. The expression level of miR-22-3p could be monitored to predict DVT patients' prognosis.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Endothelial Progenitor Cells/cytology , MicroRNAs/physiology , Onecut Transcription Factors/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Venous Thrombosis/metabolism , Adult , Case-Control Studies , Humans , Prognosis , Venous Thrombosis/pathology
2.
Sci Rep ; 10(1): 996, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31969659

ABSTRACT

In the developing spinal cord, Onecut transcription factors control the diversification of motor neurons into distinct neuronal subsets by ensuring the maintenance of Isl1 expression during differentiation. However, other genes downstream of the Onecut proteins and involved in motor neuron diversification have remained unidentified. In the present study, we generated conditional mutant embryos carrying specific inactivation of Onecut genes in the developing motor neurons, performed RNA-sequencing to identify factors downstream of Onecut proteins in this neuron population, and employed additional transgenic mouse models to assess the role of one specific Onecut-downstream target, the transcription factor Nkx6.2. Nkx6.2 expression was up-regulated in Onecut-deficient motor neurons, but strongly downregulated in Onecut-deficient V2a interneurons, indicating an opposite regulation of Nkx6.2 by Onecut factors in distinct spinal neuron populations. Nkx6.2-null embryos, neonates and adult mice exhibited alterations of locomotor pattern and spinal locomotor network activity, likely resulting from defective survival of a subset of limb-innervating motor neurons and abnormal migration of V2a interneurons. Taken together, our results indicate that Nkx6.2 regulates the development of spinal neuronal populations and the formation of the spinal locomotor circuits downstream of the Onecut transcription factors.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Motor Neurons/metabolism , Onecut Transcription Factors/metabolism , Spinal Cord/metabolism , Transcription Factors/metabolism , Animals , Gene Expression , Homeodomain Proteins/genetics , Locomotion/physiology , Mice , Mice, Transgenic , Onecut Transcription Factors/genetics , Transcription Factors/genetics
3.
Mol Cell Neurosci ; 101: 103411, 2019 12.
Article in English | MEDLINE | ID: mdl-31648029

ABSTRACT

Onecut transcription factors are required to maintain Islet1 (Isl1) expression in developing spinal motor neurons (MNs), and this process is critical for proper MN differentiation. However, the mechanisms whereby OC stimulate Isl1 expression remain unknown. CREB-binding protein (CBP) and p300 paralogs are transcriptional coactivators that interact with OC proteins in hepatic cells. In the embryonic spinal cord, CBP and p300 play key roles in neurogenesis and MN differentiation. Here, using chromatin immunoprecipitation and in ovo electroporation in chicken spinal cord, we provide evidence that CBP and p300 contribute to the regulation of Isl1 expression by the OC factors in embryonic spinal MNs. CBP and p300 are detected on the CREST2 enhancer of Isl1 where OC factors are also bound. Inhibition of CBP and p300 activity inhibits activation of the CREST2 enhancer and prevents the stimulation of Isl1 expression by the OC factors. These observations suggest that CBP and p300 coactivators cooperate with OC factors to maintain Isl1 expression in postmitotic MNs.


Subject(s)
CREB-Binding Protein/metabolism , Enhancer Elements, Genetic , LIM-Homeodomain Proteins/genetics , Motor Neurons/metabolism , Onecut Transcription Factors/metabolism , Spinal Cord/metabolism , Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism , Animals , Chick Embryo , LIM-Homeodomain Proteins/metabolism , Spinal Cord/cytology , Transcription Factors/metabolism
4.
Nucleic Acids Res ; 47(11): 5587-5602, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31049588

ABSTRACT

Remodeling of chromatin accessibility is necessary for successful reprogramming of fibroblasts to neurons. However, it is still not fully known which transcription factors can induce a neuronal chromatin accessibility profile when overexpressed in fibroblasts. To identify such transcription factors, we used ATAC-sequencing to generate differential chromatin accessibility profiles between human fibroblasts and iNeurons, an in vitro neuronal model system obtained by overexpression of Neurog2 in induced pluripotent stem cells (iPSCs). We found that the ONECUT transcription factor sequence motif was strongly associated with differential chromatin accessibility between iNeurons and fibroblasts. All three ONECUT transcription factors associated with this motif (ONECUT1, ONECUT2 and ONECUT3) induced a neuron-like morphology and expression of neuronal genes within two days of overexpression in fibroblasts. We observed widespread remodeling of chromatin accessibility; in particular, we found that chromatin regions that contain the ONECUT motif were in- or lowly accessible in fibroblasts and became accessible after the overexpression of ONECUT1, ONECUT2 or ONECUT3. There was substantial overlap with iNeurons, still, many regions that gained accessibility following ONECUT overexpression were not accessible in iNeurons. Our study highlights both the potential and challenges of ONECUT-based direct neuronal reprogramming.


Subject(s)
Cellular Reprogramming , Chromatin/genetics , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Onecut Transcription Factors/genetics , Cell Differentiation , Cell Line , Chromatin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Ontology , Hepatocyte Nuclear Factor 6/genetics , Hepatocyte Nuclear Factor 6/metabolism , Homeodomain Proteins , Humans , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Onecut Transcription Factors/metabolism , Transcription Factors
5.
Int J Dev Biol ; 61(8-9): 537-543, 2017.
Article in English | MEDLINE | ID: mdl-29139539

ABSTRACT

Studying the formation of endoskeleton in many species is complex and difficult. The sea urchin embryo offers an unparalleled platform for understanding this process because of the ease with which its skeletogenic mesenchyme cells can be manipulated. In this study, preliminary evidence from biochemical studies towards understanding the role of the Onecut transcription factor during sea urchin skeletogenic mesenchyme cell specification is presented. Based on the evidence, an empirical model is proposed showing how Onecut, together with associated co-factors, may be using the C-element of the SM50 gene regulatory region in advance of the sea urchin Strongylocentrotus purpuratus spicule development. In the model, Onecut recognizes and binds the DNA sequence CATCGATCTC in the C-element without temporal restriction. Onecut then utilizes different sets of co-factors to switch from its unknown function early in development (four cell stage to the mesenchyme blastula stage), to its known role in the oral-aboral boundary thereafter. At the writing of this report, definitive evidence as to whether the "early" factors are expressed in all cells except the micromere lineages, or whether the "late" factors are expressed in micromere descendants or ectodermal precursors only are lacking. The former would suggest a possible Onecut repression function for the early co-factors outside the micromere lineages; the latter scenario would suggest a Onecut activation function for the late co-factors in the presumptive ciliary band.


Subject(s)
Embryo, Nonmammalian/cytology , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Onecut Transcription Factors/metabolism , Regulatory Sequences, Nucleic Acid , Sea Urchins/genetics , Animals , Cell Differentiation , Embryo, Nonmammalian/metabolism , Extracellular Matrix Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Onecut Transcription Factors/genetics , Sea Urchins/growth & development
6.
Dev Biol ; 409(1): 310-318, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26522848

ABSTRACT

Specification of the ciliated band (CB) of echinoid embryos executes three spatial functions essential for postgastrular organization. These are establishment of a band about 5 cells wide which delimits and bounds other embryonic territories; definition of a neurogenic domain within this band; and generation within it of arrays of ciliary cells that bear the special long cilia from which the structure derives its name. In Strongylocentrotus purpuratus the spatial coordinates of the future ciliated band are initially and exactly determined by the disposition of a ring of cells that transcriptionally activate the onecut homeodomain regulatory gene, beginning in blastula stage, long before the appearance of the CB per se. Thus the cis-regulatory apparatus that governs onecut expression in the blastula directly reveals the genomic sequence code by which these aspects of the spatial organization of the embryo are initially determined. We screened the entire onecut locus and its flanking region for transcriptionally active cis-regulatory elements, and by means of BAC recombineered deletions identified three separated and required cis-regulatory modules that execute different functions. The operating logic of the crucial spatial control module accounting for the spectacularly precise and beautiful early onecut expression domain depends on spatial repression. Previously predicted oral ectoderm and aboral ectoderm repressors were identified by cis-regulatory mutation as the products of goosecoid and irxa genes respectively, while the pan-ectodermal activator SoxB1 supplies a transcriptional driver function.


Subject(s)
Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Onecut Transcription Factors/genetics , Sea Urchins/embryology , Sea Urchins/genetics , Animals , Chromosomes, Artificial, Bacterial/genetics , Gastrula/metabolism , Genes, Reporter , Introns/genetics , Models, Genetic , Onecut Transcription Factors/metabolism , Regulatory Elements, Transcriptional , Repressor Proteins/metabolism
7.
Dev Biol ; 390(2): 273-87, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24680893

ABSTRACT

The expression pattern of Onecut genes in the central and peripheral nervous systems is highly conserved in invertebrates and vertebrates but the regulatory networks in which they are involved are still largely unknown. The presence of three gene copies in vertebrates has revealed the functional roles of the Onecut genes in liver, pancreas and some populations of motor neurons. Urochordates have only one Onecut gene and are the closest living relatives of vertebrates and thus represent a good model system to understand its regulatory network and involvement in nervous system formation. In order to define the Onecut genetic cascade, we extensively characterized the Onecut upstream cis-regulatory DNA in the ascidian Ciona intestinalis. Electroporation experiments using a 2.5kb genomic fragment and of a series of deletion constructs identified a small region of 262bp able to reproduce most of the Onecut expression profile during embryonic development. Further analyses, both bioinformatic and in vivo using transient transgenes, permitted the identification of transcription factors responsible for Onecut endogenous expression. We provide evidence that Neurogenin is a direct activator of Onecut and that an autoregulatory loop is responsible for the maintenance of its expression. Furthermore, for the first time we propose the existence of a direct connection among Neurogenin, Onecut and Rx transcription factors in photoreceptor cell formation.


Subject(s)
Gene Expression Regulation/genetics , Nerve Tissue Proteins/metabolism , Nervous System/metabolism , Onecut Transcription Factors/metabolism , Photoreceptor Cells/physiology , Regulatory Elements, Transcriptional/genetics , Urochordata/genetics , Animals , Electroporation , Histocytochemistry , In Situ Hybridization , Italy , Mediterranean Sea , Nerve Tissue Proteins/genetics , Nervous System/embryology , Onecut Transcription Factors/genetics , Photoreceptor Cells/metabolism , Transcriptome
8.
Dev Biol ; 386(2): 358-70, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24374159

ABSTRACT

Motor neurons in the vertebrate spinal cord are stereotypically organized along the rostro-caudal axis in discrete columns that specifically innervate peripheral muscle domains. Originating from the same progenitor domain, the generation of spinal motor neurons is orchestrated by a spatially and temporally tightly regulated set of secreted molecules and transcription factors such as retinoic acid and the Lim homeodomain transcription factors Isl1 and Lhx1. However, the molecular interactions between these factors remained unclear. In this study we examined the role of the microRNA 9 (miR-9) in the specification of spinal motor neurons and identified Onecut1 (OC1) as one of its targets. miR-9 and OC1 are expressed in mutually exclusive patterns in the developing chick spinal cord, with high OC1 levels in early-born motor neurons and high miR-9 levels in late-born motor neurons. miR-9 efficiently represses OC1 expression in vitro and in vivo. Overexpression of miR-9 leads to an increase in late-born neurons, while miR-9 loss-of-function induces additional OC1(+) motor neurons that display a transcriptional profile typical of early-born neurons. These results demonstrate that regulation of OC1 by miR-9 is a crucial step in the specification of spinal motor neurons and support a model in which miR-9 expression in late-born LMCl neurons downregulates Isl1 expression through inhibition of OC1. In conclusion, our study contributes essential factors to the molecular network specifying spinal motor neurons and emphasizes the importance of microRNAs as key players in the generation of neuronal diversity.


Subject(s)
Gene Expression Regulation, Developmental/physiology , MicroRNAs/metabolism , Motor Neurons/physiology , Onecut Transcription Factors/metabolism , Spinal Cord/embryology , Analysis of Variance , Animals , Base Sequence , Chick Embryo , Electroporation , Fluorescence , Gene Expression Regulation, Developmental/genetics , Immunohistochemistry , In Situ Hybridization , Luciferases , MicroRNAs/genetics , Molecular Sequence Data , Motor Neurons/metabolism , Onecut Transcription Factors/genetics
9.
Development ; 139(17): 3109-19, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22833130

ABSTRACT

During development, spinal motoneurons (MNs) diversify into a variety of subtypes that are specifically dedicated to the motor control of particular sets of skeletal muscles or visceral organs. MN diversification depends on the coordinated action of several transcriptional regulators including the LIM-HD factor Isl1, which is crucial for MN survival and fate determination. However, how these regulators cooperate to establish each MN subtype remains poorly understood. Here, using phenotypic analyses of single or compound mutant mouse embryos combined with gain-of-function experiments in chick embryonic spinal cord, we demonstrate that the transcriptional activators of the Onecut family critically regulate MN subtype diversification during spinal cord development. We provide evidence that Onecut factors directly stimulate Isl1 expression in specific MN subtypes and are therefore required to maintain Isl1 production at the time of MN diversification. In the absence of Onecut factors, we observed major alterations in MN fate decision characterized by the conversion of somatic to visceral MNs at the thoracic levels of the spinal cord and of medial to lateral MNs in the motor columns that innervate the limbs. Furthermore, we identify Sip1 (Zeb2) as a novel developmental regulator of visceral MN differentiation. Taken together, these data elucidate a comprehensive model wherein Onecut factors control multiple aspects of MN subtype diversification. They also shed light on the late roles of Isl1 in MN fate decision.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation, Developmental/genetics , LIM-Homeodomain Proteins/metabolism , Motor Neurons/physiology , Onecut Transcription Factors/metabolism , Spinal Cord/cytology , Transcription Factors/metabolism , Animals , Chick Embryo , Chromatin Immunoprecipitation , DNA Primers/genetics , Electroporation , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental/physiology , In Situ Hybridization , Mice
10.
J Comp Neurol ; 520(7): 1424-41, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22102297

ABSTRACT

The A13 dopaminergic nucleus belongs to the incerto-hypothalamic area. It is thought to exert autonomous roles by integrating sensory input to autonomic, neuroendocrine, and motor output. Although its early development has been well characterized, the factors that contribute to later steps of its formation remain unknown. Transcription factors of the Onecut family have been detected in the A13 nucleus, raising the question of possible roles of these factors during A13 development. Using a combination of immunofluorescence analyses on sections and after whole-mount labeling followed by 3D reconstructions, we further characterized the second phase of development of the A13 nucleus in the mouse, described the distribution of the Onecut proteins throughout A13 development, and analyzed the phenotype of this nucleus in single or compound mutant embryos for the Onecut factors. Here we show that A13 development can be divided into two successive phases. First, during radial migration toward the pial surface the A13 cells differentiate into dopaminergic neurons. Second, these cells gather in the vicinity of the third ventricle. Onecut factors are dynamically and differentially expressed in the A13 nucleus during these two phases of development. In Onecut mutant embryos, the A13 neurons differentiate normally but scatter in the diencephalon and fail to properly gather close to the third ventricle. Hence, Onecut factors are markers of the A13 nucleus throughout embryonic development. They are dispensable for the first phase of A13 development but are required for the second phase of development and for maintenance of this nucleus.


Subject(s)
Diencephalon/embryology , Diencephalon/metabolism , Neurogenesis/physiology , Onecut Transcription Factors/metabolism , Animals , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Fluorescent Antibody Technique , In Situ Hybridization , Mice , Mice, Mutant Strains
11.
Dev Biol ; 355(2): 358-71, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21600895

ABSTRACT

Retinal homeobox (Rx) genes play a crucial and conserved role in the development of the anterior neural plate of metazoans. During chordate evolution, they have also acquired a novel function in the control of eye formation and neurogenesis. To characterize the Rx genetic cascade and shed light on the mechanisms that led to the acquisition of this new role in eye development, we studied Rx transcriptional regulation using the ascidian, Ciona intestinalis. Through deletion analysis of the Ci-Rx promoter, we have identified two distinct enhancer elements able to induce Ci-Rx specific expression in the anterior part of the CNS and in the photosensory organ at tailbud and larva stages. Bioinformatic analysis highlighted the presence of two Onecut binding sites contained in these enhancers, so we explored the role of this transcription factor in the regulation of Ci-Rx. By in situ hybridization, we first confirmed that these genes are co-expressed in the same cells. Through a series of in vivo and in vitro experiments, we then demonstrated that the two Onecut sites are responsible for enhancer activation in Ci-Rx endogenous territories. We also demonstrated in vivo that Onecut misexpression is able to induce ectopic activation of the Rx promoter. Finally, we demonstrated that Ci-Onecut is able to promote Ci-Rx expression in the sensory vesicle. Together, these results support the conclusion that in Ciona embryogenesis, Ci-Rx expression is under the control of the Onecut transcription factor and that this factor is necessary and sufficient to specifically activate Ci-Rx through two enhancer elements.


Subject(s)
Ciona intestinalis/embryology , Eye Proteins/metabolism , Eye/embryology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Onecut Transcription Factors/metabolism , Animals , Central Nervous System/metabolism , Ciona intestinalis/genetics , Computational Biology , Electrophoretic Mobility Shift Assay , Electroporation , Homeodomain Proteins/genetics , In Situ Hybridization , Promoter Regions, Genetic/genetics , beta-Galactosidase/metabolism
12.
Genetics ; 177(3): 1621-37, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17720939

ABSTRACT

Sex is determined in Caenorhabditis elegans through a dose-dependent signal that communicates the number of X chromosomes relative to the ploidy, the number of sets of autosomes. The sex switch gene xol-1 is the direct molecular target of this X:A signal and integrates both X and autosomal components to determine sexual fate. X chromosome number is relayed by X signal elements (XSEs) that act cumulatively to repress xol-1 in XX animals, thereby inducing hermaphrodite fate. Ploidy is relayed by autosomal signal elements (ASEs), which counteract the single dose of XSEs in XO animals to activate xol-1 and induce the male fate. Our goal was to identify and characterize new XSEs and further analyze known XSEs to understand the principles by which a small difference in the concentration of an intracellular signal is amplified to induce dramatically different developmental fates. We identified a new XSE, the ONECUT homeodomain protein CEH-39, and showed that it acts as a dose-dependent repressor of xol-1 transcript levels. Unexpectedly, most other XSEs also repress xol-1 predominantly, but not exclusively, at the transcript level. The twofold difference in X dose between XO and XX animals is translated into the male vs. hermaphrodite fate by the synergistic action of multiple, independent XSEs that render xol-1 active or inactive, primarily through transcriptional regulation.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Onecut Transcription Factors/metabolism , Sex Determination Processes , X Chromosome/genetics , Animals , Animals, Genetically Modified , Base Sequence , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , Chromosome Mapping , DNA Primers/genetics , DNA, Helminth/genetics , Dosage Compensation, Genetic , Female , Gene Dosage , Genes, Helminth , Male , Models, Biological , Onecut Transcription Factors/genetics , RNA Interference , Sex Differentiation/genetics , Signal Transduction
13.
J Biol Chem ; 281(43): 32263-71, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16950765

ABSTRACT

An intestine-specific gene regulatory region was previously identified near the second exon of the human adenosine deaminase (ADA) gene. In mammalian intestine, ADA is expressed at high levels only along the villi of the duodenal epithelium, principally if not exclusively in enterocytes. Within the ADA intestinal regulatory region, a potent duodenum-specific enhancer was identified that controls this pattern of expression. This enhancer has been shown to rely on PDX-1, GATA factors, and Cdx factors for its function. Upstream of the enhancer, a separate temporal regulatory region was identified that has no independent enhancer capability but controls the timing of enhancer activation. DNase I footprinting and electrophoretic mobility shift assays were used to begin to characterize temporal region function at the molecular level. In this manner, binding sites for the Onecut (OC) family of factors, YY1, and NFI family members were identified. Identification of the OC site was especially interesting, because almost nothing is known about the function of OC factors in intestine. In transgenic mice, mutation of the OC site to ablate binding resulted in a delay of 2-3 weeks in enhancer activation in the developing postnatal intestine, a result very similar to that observed previously when the entire temporal region was deleted. In mammals, the OC family is comprised of OC-1/HNF-6, OC-2, and OC-3. An examination of intestinal expression patterns showed that all three OC factors are expressed at detectable levels in adult mouse duodenum, with OC-2 predominant. In postnatal day 2 mice only OC-2 and OC-3 were detected in intestine, with OC-2 again predominant.


Subject(s)
Enhancer Elements, Genetic , Intestinal Mucosa/physiology , Onecut Transcription Factors/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adult , Animals , Animals, Newborn , Base Sequence , Binding, Competitive , Chromosome Mapping , DNA Footprinting , Deoxyribonuclease I/metabolism , Duodenum/enzymology , Duodenum/physiology , Gene Expression Regulation, Enzymologic , Humans , Intestinal Mucosa/enzymology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutagenesis , Onecut Transcription Factors/chemistry , Onecut Transcription Factors/genetics , Plasmids , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL