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










Publication year range
1.
Mol Imaging Biol ; 19(5): 694-702, 2017 10.
Article in English | MEDLINE | ID: mdl-28233218

ABSTRACT

PURPOSE: Tumor-associated macrophages (TAMs) are often associated with a poor prognosis in cancer. To gain a better understanding of cellular recruitment and dynamics of TAM biology during cancer progression, we established a novel transgenic mouse model for in vivo imaging of luciferase-expressing macrophages. PROCEDURES: B6.129P2-Lyz2tm1(cre)Ifo/J mice, which express Cre recombinase under the control of the lysozyme M promoter (LysM) were crossed to Cre-lox Luc reporter mice (RLG), to produce LysM-LG mice whose macrophages express luciferase. Cell-type-specific luciferase expression in these mice was verified by flow cytometry, and via in vivo bioluminescence imaging under conditions where macrophages were either stimulated with lipopolysaccharide or depleted with clodronate liposomes. The distribution of activated macrophages was longitudinally imaged in two immunocompetent LysM-LG mouse models with either B16 melanoma or ID8 ovarian cancer cells. RESULTS: In vivo imaging of LysM-LG mice showed luciferase activity was generated by macrophages. Clodronate liposome-mediated depletion of macrophages lowered overall bioluminescence while lipopolysaccharide injection increased macrophage bioluminescence in both the B16 and ID8 models. Tracking macrophages weekly in tumor-bearing animals after intraperitoneal (i.p.) or intraovarian (i.o.) injection resulted in distinct, dynamic patterns of macrophage activity. Animals with metastatic ovarian cancer after i.p. injection exhibited significantly higher peritoneal macrophage activity compared to animals after i.o. injection. CONCLUSION: The LysM-LG model allows tracking of macrophage recruitment and activation during disease initiation and progression in a noninvasive manner. This model provides a tool to visualize and monitor the benefit of pharmacological interventions targeting macrophages in preclinical models.


Subject(s)
Macrophages/pathology , Molecular Imaging/methods , Ovarian Neoplasms/pathology , Animals , Disease Models, Animal , Female , Genes, Reporter , Luminescent Measurements , Macrophage Activation , Melanoma/pathology , Mice, Transgenic , Muramidase/genetics , Promoter Regions, Genetic/genetics
2.
PLoS Genet ; 9(8): e1003728, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24009522

ABSTRACT

Genetic studies have identified a core set of transcription factors and target genes that control the development of the neocortex, the region of the human brain responsible for higher cognition. The specific regulatory interactions between these factors, many key upstream and downstream genes, and the enhancers that mediate all these interactions remain mostly uncharacterized. We perform p300 ChIP-seq to identify over 6,600 candidate enhancers active in the dorsal cerebral wall of embryonic day 14.5 (E14.5) mice. Over 95% of the peaks we measure are conserved to human. Eight of ten (80%) candidates tested using mouse transgenesis drive activity in restricted laminar patterns within the neocortex. GREAT based computational analysis reveals highly significant correlation with genes expressed at E14.5 in key areas for neocortex development, and allows the grouping of enhancers by known biological functions and pathways for further studies. We find that multiple genes are flanked by dozens of candidate enhancers each, including well-known key neocortical genes as well as suspected and novel genes. Nearly a quarter of our candidate enhancers are conserved well beyond mammals. Human and zebrafish regions orthologous to our candidate enhancers are shown to most often function in other aspects of central nervous system development. Finally, we find strong evidence that specific interspersed repeat families have contributed potentially key developmental enhancers via co-option. Our analysis expands the methodologies available for extracting the richness of information found in genome-wide functional maps.


Subject(s)
Enhancer Elements, Genetic , Evolution, Molecular , Neocortex/growth & development , Regulatory Sequences, Nucleic Acid/genetics , Animals , Base Sequence , Conserved Sequence/genetics , Gene Expression Regulation, Developmental , Humans , Mice , Neocortex/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Transcription Factors/genetics , Zebrafish/genetics , Zebrafish/growth & development
3.
Genome Res ; 23(5): 889-904, 2013 May.
Article in English | MEDLINE | ID: mdl-23382538

ABSTRACT

The human genome encodes 1500-2000 different transcription factors (TFs). ChIP-seq is revealing the global binding profiles of a fraction of TFs in a fraction of their biological contexts. These data show that the majority of TFs bind directly next to a large number of context-relevant target genes, that most binding is distal, and that binding is context specific. Because of the effort and cost involved, ChIP-seq is seldom used in search of novel TF function. Such exploration is instead done using expression perturbation and genetic screens. Here we propose a comprehensive computational framework for transcription factor function prediction. We curate 332 high-quality nonredundant TF binding motifs that represent all major DNA binding domains, and improve cross-species conserved binding site prediction to obtain 3.3 million conserved, mostly distal, binding site predictions. We combine these with 2.4 million facts about all human and mouse gene functions, in a novel statistical framework, in search of enrichments of particular motifs next to groups of target genes of particular functions. Rigorous parameter tuning and a harsh null are used to minimize false positives. Our novel PRISM (predicting regulatory information from single motifs) approach obtains 2543 TF function predictions in a large variety of contexts, at a false discovery rate of 16%. The predictions are highly enriched for validated TF roles, and 45 of 67 (67%) tested binding site regions in five different contexts act as enhancers in functionally matched cells.


Subject(s)
Binding Sites/genetics , Computational Biology , Software , Transcription Factors/genetics , Algorithms , Animals , Base Sequence , DNA-Binding Proteins/genetics , Genome , Humans , Mice , Protein Binding/genetics , Regulatory Sequences, Nucleic Acid
4.
Cell Rep ; 2(4): 817-23, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-23022484

ABSTRACT

Genotype-phenotype mapping is hampered by countless genomic changes between species. We introduce a computational "forward genomics" strategy that-given only an independently lost phenotype and whole genomes-matches genomic and phenotypic loss patterns to associate specific genomic regions with this phenotype. We conducted genome-wide screens for two metabolic phenotypes. First, our approach correctly matches the inactivated Gulo gene exactly with the species that lost the ability to synthesize vitamin C. Second, we attribute naturally low biliary phospholipid levels in guinea pigs and horses to the inactivated phospholipid transporter Abcb4. Human ABCB4 mutations also result in low phospholipid levels but lead to severe liver disease, suggesting compensatory mechanisms in guinea pig and horse. Our simulation studies, counts of independent changes in existing phenotype surveys, and the forthcoming availability of many new genomes all suggest that forward genomics can be applied to many phenotypes, including those relevant for human evolution and disease.


Subject(s)
Genomics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Ascorbic Acid/biosynthesis , Computational Biology , Exons , Gene Expression Profiling , Genotype , Guinea Pigs , Horses , Humans , Mice , Molecular Sequence Data , Mutation , Phenotype , Phospholipids/metabolism
5.
Nucleic Acids Res ; 40(22): 11463-76, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23042682

ABSTRACT

Conserved non-protein-coding DNA elements (CNEs) often encode cis-regulatory elements and are rarely lost during evolution. However, CNE losses that do occur can be associated with phenotypic changes, exemplified by pelvic spine loss in sticklebacks. Using a computational strategy to detect complete loss of CNEs in mammalian genomes while strictly controlling for artifacts, we find >600 CNEs that are independently lost in at least two mammalian lineages, including a spinal cord enhancer near GDF11. We observed several genomic regions where multiple independent CNE loss events happened; the most extreme is the DIAPH2 locus. We show that CNE losses often involve deletions and that CNE loss frequencies are non-uniform. Similar to less pleiotropic enhancers, we find that independently lost CNEs are shorter, slightly less constrained and evolutionarily younger than CNEs without detected losses. This suggests that independently lost CNEs are less pleiotropic and that pleiotropic constraints contribute to non-uniform CNE loss frequencies. We also detected 35 CNEs that are independently lost in the human lineage and in other mammals. Our study uncovers an interesting aspect of the evolution of functional DNA in mammalian genomes. Experiments are necessary to test if these independently lost CNEs are associated with parallel phenotype changes in mammals.


Subject(s)
Evolution, Molecular , Genome , Mammals/genetics , Regulatory Sequences, Nucleic Acid , Animals , Base Sequence , Conserved Sequence , DNA, Intergenic/chemistry , Enhancer Elements, Genetic , Genetic Pleiotropy , Genomics/methods , Humans , Primates , Sequence Deletion
6.
PLoS Genet ; 8(8): e1002852, 2012.
Article in English | MEDLINE | ID: mdl-22876195

ABSTRACT

The identification of homologies, whether morphological, molecular, or genetic, is fundamental to our understanding of common biological principles. Homologies bridging the great divide between deuterostomes and protostomes have served as the basis for current models of animal evolution and development. It is now appreciated that these two clades share a common developmental toolkit consisting of conserved transcription factors and signaling pathways. These patterning genes sometimes show common expression patterns and genetic interactions, suggesting the existence of similar or even conserved regulatory apparatus. However, previous studies have found no regulatory sequence conserved between deuterostomes and protostomes. Here we describe the first such enhancers, which we call bilaterian conserved regulatory elements (Bicores). Bicores show conservation of sequence and gene synteny. Sequence conservation of Bicores reflects conserved patterns of transcription factor binding sites. We predict that Bicores act as response elements to signaling pathways, and we show that Bicores are developmental enhancers that drive expression of transcriptional repressors in the vertebrate central nervous system. Although the small number of identified Bicores suggests extensive rewiring of cis-regulation between the protostome and deuterostome clades, additional Bicores may be revealed as our understanding of cis-regulatory logic and sample of bilaterian genomes continue to grow.


Subject(s)
Enhancer Elements, Genetic , Genome , Invertebrates/genetics , Transcription Factors/genetics , Vertebrates/genetics , Amino Acid Sequence , Animals , Binding Sites , Biological Evolution , Central Nervous System/embryology , Central Nervous System/metabolism , Conserved Sequence , Gene Expression Regulation, Developmental , Humans , Invertebrates/embryology , Invertebrates/metabolism , Molecular Sequence Data , Protein Binding , Sequence Alignment , Signal Transduction , Synteny , Transcription Factors/metabolism , Vertebrates/embryology , Vertebrates/metabolism
7.
Brain ; 134(Pt 6): 1777-89, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21616972

ABSTRACT

Stem cell transplantation promises new hope for the treatment of stroke although significant questions remain about how the grafted cells elicit their effects. One hypothesis is that transplanted stem cells enhance endogenous repair mechanisms activated after cerebral ischaemia. Recognizing that bilateral reorganization of surviving circuits is associated with recovery after stroke, we investigated the ability of transplanted human neural progenitor cells to enhance this structural plasticity. Our results show the first evidence that human neural progenitor cell treatment can significantly increase dendritic plasticity in both the ipsi- and contralesional cortex and this coincides with stem cell-induced functional recovery. Moreover, stem cell-grafted rats demonstrated increased corticocortical, corticostriatal, corticothalamic and corticospinal axonal rewiring from the contralesional side; with the transcallosal and corticospinal axonal sprouting correlating with functional recovery. Furthermore, we demonstrate that axonal transport, which is critical for both proper axonal function and axonal sprouting, is inhibited by stroke and that this is rescued by the stem cell treatment, thus identifying another novel potential mechanism of action of transplanted cells. Finally, we established in vitro co-culture assays in which these stem cells mimicked the effects observed in vivo. Through immunodepletion studies, we identified vascular endothelial growth factor, thrombospondins 1 and 2, and slit as mediators partially responsible for stem cell-induced effects on dendritic sprouting, axonal plasticity and axonal transport in vitro. Thus, we postulate that human neural progenitor cells aid recovery after stroke through secretion of factors that enhance brain repair and plasticity.


Subject(s)
Axonal Transport/physiology , Brain Ischemia/surgery , Cerebral Cortex/cytology , Neural Stem Cells , Neuronal Plasticity/physiology , Analysis of Variance , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Brain Infarction/etiology , Brain Infarction/pathology , Brain Ischemia/complications , Cell Survival , Cells, Cultured , Corpus Callosum/pathology , Dendrites/physiology , Dextrans/metabolism , Disease Models, Animal , Fetus , Gene Expression Regulation , Humans , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Psychomotor Performance/physiology , RNA, Messenger/metabolism , Rats , Rats, Nude , Rats, Sprague-Dawley , Time Factors , Vibrissae/innervation
8.
Nature ; 471(7337): 216-9, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21390129

ABSTRACT

Humans differ from other animals in many aspects of anatomy, physiology, and behaviour; however, the genotypic basis of most human-specific traits remains unknown. Recent whole-genome comparisons have made it possible to identify genes with elevated rates of amino acid change or divergent expression in humans, and non-coding sequences with accelerated base pair changes. Regulatory alterations may be particularly likely to produce phenotypic effects while preserving viability, and are known to underlie interesting evolutionary differences in other species. Here we identify molecular events particularly likely to produce significant regulatory changes in humans: complete deletion of sequences otherwise highly conserved between chimpanzees and other mammals. We confirm 510 such deletions in humans, which fall almost exclusively in non-coding regions and are enriched near genes involved in steroid hormone signalling and neural function. One deletion removes a sensory vibrissae and penile spine enhancer from the human androgen receptor (AR) gene, a molecular change correlated with anatomical loss of androgen-dependent sensory vibrissae and penile spines in the human lineage. Another deletion removes a forebrain subventricular zone enhancer near the tumour suppressor gene growth arrest and DNA-damage-inducible, gamma (GADD45G), a loss correlated with expansion of specific brain regions in humans. Deletions of tissue-specific enhancers may thus accompany both loss and gain traits in the human lineage, and provide specific examples of the kinds of regulatory alterations and inactivation events long proposed to have an important role in human evolutionary divergence.


Subject(s)
Biological Evolution , DNA/genetics , Genome, Human/genetics , Human Characteristics , Regulatory Sequences, Nucleic Acid/genetics , Sequence Deletion/genetics , Animals , Brain/anatomy & histology , Brain/metabolism , Chromosomes, Mammalian/genetics , Conserved Sequence/genetics , DNA, Intergenic/genetics , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Genes, Tumor Suppressor , Humans , Male , Mice , Organ Specificity , Pan troglodytes/genetics , Penis/anatomy & histology , Penis/metabolism , Species Specificity , Transgenes/genetics
9.
PLoS One ; 6(3): e17802, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21445347

ABSTRACT

Newborn neurons migrate from their birthplace to their final location to form a properly functioning nervous system. During these movements, young neurons must attach and subsequently detach from their substrate to facilitate migration, but little is known about the mechanisms cells use to release their attachments. We show that the machinery for clathrin-mediated endocytosis is positioned to regulate the distribution of adhesion proteins in a subcellular region just proximal to the neuronal cell body. Inhibiting clathrin or dynamin function impedes the movement of migrating neurons both in vitro and in vivo. Inhibiting dynamin function in vitro shifts the distribution of adhesion proteins to the rear of the cell. These results suggest that endocytosis may play a critical role in regulating substrate detachment to enable cell body translocation in migrating neurons.


Subject(s)
Cell Adhesion , Endocytosis , Neurons/metabolism , Clathrin/physiology , Dynamins/physiology , Electroporation , Humans , Immunohistochemistry , Microscopy, Electron
10.
Nat Biotechnol ; 28(5): 495-501, 2010 May.
Article in English | MEDLINE | ID: mdl-20436461

ABSTRACT

We developed the Genomic Regions Enrichment of Annotations Tool (GREAT) to analyze the functional significance of cis-regulatory regions identified by localized measurements of DNA binding events across an entire genome. Whereas previous methods took into account only binding proximal to genes, GREAT is able to properly incorporate distal binding sites and control for false positives using a binomial test over the input genomic regions. GREAT incorporates annotations from 20 ontologies and is available as a web application. Applying GREAT to data sets from chromatin immunoprecipitation coupled with massively parallel sequencing (ChIP-seq) of multiple transcription-associated factors, including SRF, NRSF, GABP, Stat3 and p300 in different developmental contexts, we recover many functions of these factors that are missed by existing gene-based tools, and we generate testable hypotheses. The utility of GREAT is not limited to ChIP-seq, as it could also be applied to open chromatin, localized epigenomic markers and similar functional data sets, as well as comparative genomics sets.


Subject(s)
Data Mining/methods , Genome , Genomics/methods , Regulatory Elements, Transcriptional , Software , Animals , Chromatin Immunoprecipitation , Databases, Genetic , E1A-Associated p300 Protein , Humans , Jurkat Cells , Mice , Protein Binding , Serum Response Factor
11.
Proc Natl Acad Sci U S A ; 102(38): 13652-7, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16174753

ABSTRACT

Discoveries from human and mouse genetics have identified cytoskeletal and signaling proteins that are essential for neuronal migration in the developing brain. To provide a meaningful context for these studies, we took an unbiased approach of correlative electron microscopy of neurons migrating through a three-dimensional matrix, and characterized the cytoskeletal events that occur as migrating neurons initiate saltatory forward movements of the cell nucleus. The formation of a cytoplasmic dilation in the proximal leading process precedes nuclear translocation. Cell nuclei translocate into these dilations in saltatory movements. Time-lapse imaging and pharmacological perturbation suggest that nucleokinesis requires stepwise or hierarchical interactions between microtubules, myosin II, and cell adhesion. We hypothesize that these interactions couple leading process extension to nuclear translocation during neuronal migration.


Subject(s)
Cell Movement/physiology , Cell Nucleus/physiology , Microtubules/metabolism , Myosin Type II/metabolism , Neurons/physiology , Animals , Brain/embryology , Brain/ultrastructure , Cell Adhesion/physiology , Cell Nucleus/ultrastructure , Cells, Cultured , Humans , Mice , Microscopy, Electron, Transmission , Microtubules/ultrastructure , Myosin Type II/ultrastructure , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley
12.
Neuron ; 44(2): 213-4, 2004 Oct 14.
Article in English | MEDLINE | ID: mdl-15473958

ABSTRACT

The development of the cerebral cortex is a highly orchestrated process of cell division and migration. In this issue of Neuron, Feng and Walsh and Shu et al. examine the roles of two related proteins, Nde1 (mNudE) and Ndel1 (NUDEL), in cortical development. These proteins play a crucial role in centrosome positioning, with Nde1 functioning mainly during progenitor cell divisions and Ndel1 functioning in neuronal migration.


Subject(s)
Carrier Proteins/physiology , Cerebral Cortex/embryology , Animals , Humans
13.
Neuron ; 41(2): 203-13, 2004 Jan 22.
Article in English | MEDLINE | ID: mdl-14741102

ABSTRACT

Doublecortin (Dcx) is a microtubule-associated protein that is mutated in X-linked lissencephaly (X-LIS), a neuronal migration disorder associated with epilepsy and mental retardation. Although Dcx can bind ubiquitously to microtubules in nonneuronal cells, Dcx is highly enriched in the leading processes of migrating neurons and the growth cone region of differentiating neurons. We present evidence that Dcx/microtubule interactions are negatively controlled by Protein Kinase A (PKA) and the MARK/PAR-1 family of protein kinases. In addition to a consensus MARK site, we identified a serine within a novel sequence that is crucial for the PKA- and MARK-dependent regulation of Dcx's microtubule binding activity in vitro. This serine is mutated in two families affected by X-LIS. Immunostaining neurons with an antibody that recognizes phosphorylated substrates of MARK supports the conclusion that Dcx localization and function are regulated at the leading edge of migrating cells by a balance of kinase and phosphatase activity.


Subject(s)
Cell Movement/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , Neurons/physiology , Neuropeptides/physiology , Phosphoric Monoester Hydrolases/metabolism , Animals , Cell Membrane/enzymology , Cell Membrane/metabolism , Cell Movement/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/physiology , Cytoskeleton/metabolism , Doublecortin Domain Proteins , Doublecortin Protein , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Microtubule-Associated Proteins/metabolism , Microtubules/drug effects , Mutation/physiology , Nerve Tissue Proteins/biosynthesis , Neurites/metabolism , Neurons/enzymology , Neurons/metabolism , Neuropeptides/metabolism , Okadaic Acid/pharmacology , Phosphorylation , Rats , Rats, Sprague-Dawley , Serine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...