Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Am J Pathol ; 188(5): 1276-1288, 2018 05.
Article in English | MEDLINE | ID: mdl-29458011

ABSTRACT

Tumor invasion is a critical first step in the organismic dissemination of cancer cells and the formation of metastasis in distant organs, the most important prognostic factor and the actual cause of death in most of the cancer patients. We report herein that the cell surface protein podoplanin (PDPN), a potent inducer of cancer cell invasion, is conspicuously expressed by the invasive front of squamous cell carcinomas (SCCs) of the cervix in patients and in the transgenic human papillomavirus/estrogen mouse model of cervical cancer. Laser capture microscopy combined with gene expression profiling reveals that the expression of interferon-responsive genes is up-regulated in PDPN-expressing cells at the tumor invasive front, which are exposed to CD45-positive inflammatory cells. Indeed, PDPN expression can be induced in cultured SCC cell lines by single or combined treatments with interferon-γ, transforming growth factor-ß, and/or tumor necrosis factor-α. Notably, shRNA-mediated ablation of either PDPN or STAT1 in A431 SCC cells repressed cancer cell invasion on s.c. transplantation into immunodeficient mice. The results highlight the induction of tumor cell invasion by the inflammatory cytokine-stimulated expression of PDPN in the outermost cell layers of cervical SCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cytokines/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Membrane Glycoproteins/metabolism , Neoplasm Invasiveness/genetics , Uterine Cervical Neoplasms/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Female , Humans , Membrane Glycoproteins/genetics , Mice , Neoplasm Invasiveness/pathology , Transcriptome , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
2.
PLoS One ; 10(4): e0124870, 2015.
Article in English | MEDLINE | ID: mdl-25901736

ABSTRACT

BACKGROUND: Medulloblastomas are malignant childhood brain tumors that arise due to the aberrant activity of developmental pathways during postnatal cerebellar development and in adult humans. Transcriptome analysis has identified four major medulloblastoma subgroups. One of them, the Sonic hedgehog (SHH) subgroup, is caused by aberrant Hedgehog signal transduction due to mutations in the Patched1 (PTCH1) receptor or downstream effectors. Mice carrying a Patched-1 null allele (Ptch1∆/+) are a good model to study the alterations underlying medulloblastoma development as a consequence of aberrant Hedgehog pathway activity. RESULTS: Transcriptome analysis of human medulloblastomas shows that SERPINE2, also called Protease Nexin-1 (PN-1) is overexpressed in most medulloblastomas, in particular in the SHH and WNT subgroups. As siRNA-mediated lowering of SERPINE2/PN-1 in human medulloblastoma DAOY cells reduces cell proliferation, we analyzed its potential involvement in medulloblastoma development using the Ptch1∆/+ mouse model. In Ptch1∆/+ mice, medulloblastomas arise as a consequence of aberrant Hedgehog pathway activity. Genetic reduction of Serpine2/Pn-1 interferes with medulloblastoma development in Ptch1∆/+ mice, as ~60% of the pre-neoplastic lesions (PNLs) fail to develop into medulloblastomas and remain as small cerebellar nodules. In particular the transcription factor Atoh1, whose expression is essential for development of SHH subgroup medulloblastomas is lost. Comparative molecular analysis reveals the distinct nature of the PNLs in young Ptch1∆/+Pn-1Δ/+ mice. The remaining wild-type Ptch1 allele escapes transcriptional silencing in most cases and the aberrant Hedgehog pathway activity is normalized. Furthermore, cell proliferation and the expression of the cell-cycle regulators Mycn and Cdk6 are significantly reduced in PNLs of Ptch1∆/+Pn-1Δ/+ mice. CONCLUSIONS: Our analysis provides genetic evidence that aberrant Serpine2/Pn-1 is required for proliferation of human and mouse medulloblastoma cells. In summary, our analysis shows that Serpine2/PN-1 boosts malignant progression of PNLs to medulloblastomas, in which the Hedgehog pathway is activated in a SHH ligand-independent manner.


Subject(s)
Disease Progression , Medulloblastoma/metabolism , Medulloblastoma/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Serpin E2/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cerebellum/pathology , Disease Models, Animal , Gene Silencing , Genotype , Hedgehog Proteins/metabolism , Humans , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/metabolism , Signal Transduction
3.
Proc Natl Acad Sci U S A ; 112(18): E2327-36, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25902518

ABSTRACT

Regeneration of fragmented Drosophila imaginal discs occurs in an epimorphic manner involving local cell proliferation at the wound site. After disc fragmentation, cells at the wound site activate a restoration program through wound healing, regenerative cell proliferation, and repatterning of the tissue. However, the interplay of signaling cascades driving these early reprogramming steps is not well-understood. Here, we profiled the transcriptome of regenerating cells in the early phase within 24 h after wounding. We found that JAK/STAT signaling becomes activated at the wound site and promotes regenerative cell proliferation in cooperation with Wingless (Wg) signaling. In addition, we showed that the expression of Drosophila insulin-like peptide 8 (dilp8), which encodes a paracrine peptide to delay the onset of pupariation, is controlled by JAK/STAT signaling in early regenerating discs. Our findings suggest that JAK/STAT signaling plays a pivotal role in coordinating regenerative disc growth with organismal developmental timing.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/physiology , Imaginal Discs/physiology , Intercellular Signaling Peptides and Proteins/metabolism , STAT Transcription Factors/metabolism , Wound Healing , Animals , Body Patterning , Cell Lineage , Cell Proliferation , Cluster Analysis , Gene Expression Regulation , Janus Kinases/metabolism , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , Regeneration , Signal Transduction , Transcription Factors/metabolism , Transcriptome
4.
Breast Cancer Res ; 16(3): R60, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24916766

ABSTRACT

INTRODUCTION: Stromal-epithelial interactions play a fundamental role in tissue homeostasis, controlling cell proliferation and differentiation. Not surprisingly, aberrant stromal-epithelial interactions contribute to malignancies. Studies of the cellular and molecular mechanisms underlying these interactions require ex vivo experimental model systems that recapitulate the complexity of human tissue without compromising the differentiation and proliferation potentials of human primary cells. METHODS: We isolated and characterized human breast epithelial and mesenchymal precursors from reduction mammoplasty tissue and tagged them with lentiviral vectors. We assembled heterotypic co-cultures and compared mesenchymal and epithelial cells to cells in corresponding monocultures by analyzing growth, differentiation potentials, and gene expression profiles. RESULTS: We show that heterotypic culture of non-immortalized human primary breast epithelial and mesenchymal precursors maintains their proliferation and differentiation potentials and constrains their growth. We further describe the gene expression profiles of stromal and epithelial cells in co-cultures and monocultures and show increased expression of the tumor growth factor beta (TGFß) family member inhibin beta A (INHBA) in mesenchymal cells grown as co-cultures compared with monocultures. Notably, overexpression of INHBA in mesenchymal cells increases colony formation potential of epithelial cells, suggesting that it contributes to the dynamic reciprocity between breast mesenchymal and epithelial cells. CONCLUSIONS: The described heterotypic co-culture system will prove useful for further characterization of the molecular mechanisms mediating interactions between human normal or neoplastic breast epithelial cells and the stroma, and will provide a framework to test the relevance of the ever-increasing number of oncogenomic alterations identified in human breast cancer.


Subject(s)
Breast/cytology , Epithelial Cells/cytology , Inhibin-beta Subunits/metabolism , Mesenchymal Stem Cells/cytology , Transforming Growth Factor beta/metabolism , Adipocytes/cytology , Adipose Tissue/cytology , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Collagen Type XI/biosynthesis , Female , Gene Expression Profiling , Humans , Inhibin-beta Subunits/biosynthesis , Integrin alpha Chains/biosynthesis , Matrix Metalloproteinase 13/biosynthesis , Proteoglycans/biosynthesis , Signal Transduction , Small Leucine-Rich Proteoglycans , Transforming Growth Factor beta/biosynthesis
5.
Genes Dev ; 26(9): 920-32, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22499591

ABSTRACT

In mammals, totipotent embryos are formed by fusion of highly differentiated gametes. Acquisition of totipotency concurs with chromatin remodeling of parental genomes, changes in the maternal transcriptome and proteome, and zygotic genome activation (ZGA). The inefficiency of reprogramming somatic nuclei in reproductive cloning suggests that intergenerational inheritance of germline chromatin contributes to developmental proficiency after natural conception. Here we show that Ring1 and Rnf2, components of Polycomb-repressive complex 1 (PRC1), serve redundant transcriptional functions during oogenesis that are essential for proper ZGA, replication and cell cycle progression in early embryos, and development beyond the two-cell stage. Exchange of chromosomes between control and Ring1/Rnf2-deficient metaphase II oocytes reveal cytoplasmic and chromosome-based contributions by PRC1 to embryonic development. Our results strongly support a model in which Polycomb acts in the female germline to establish developmental competence for the following generation by silencing differentiation-inducing genes and defining appropriate chromatin states.


Subject(s)
DNA-Binding Proteins/physiology , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Oogenesis/genetics , Repressor Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Blastocyst/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , DNA Replication , DNA-Binding Proteins/genetics , Female , GATA4 Transcription Factor/genetics , Meiosis/genetics , Mice , Mice, Mutant Strains , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Repressor Proteins/genetics , Transcription, Genetic , Ubiquitin-Protein Ligases/genetics , Zygote/metabolism
6.
Nat Neurosci ; 15(3): 487-95, S1-2, 2012 Jan 22.
Article in English | MEDLINE | ID: mdl-22267162

ABSTRACT

Brain circuits are assembled from a large variety of morphologically and functionally diverse cell types. It is not known how the intermingled cell types of an individual adult brain region differ in their expressed genomes. Here we describe an atlas of cell type transcriptomes in one brain region, the mouse retina. We found that each adult cell type expressed a specific set of genes, including a unique set of transcription factors, forming a 'barcode' for cell identity. Cell type transcriptomes carried enough information to categorize cells into morphological classes and types. Several genes that were specifically expressed in particular retinal circuit elements, such as inhibitory neuron types, are associated with eye diseases. The resource described here allows gene expression to be compared across adult retinal cell types, experimenting with specific transcription factors to differentiate stem or somatic cells to retinal cell types, and predicting cellular targets of newly discovered disease-associated genes.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurons/classification , Neurons/physiology , Retina/cytology , Retinal Diseases/genetics , Transcription Factors/metabolism , Animals , Cluster Analysis , Connexins/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Flow Cytometry , Gene Expression/genetics , Gene Library , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis/methods , Mutation/genetics , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/genetics , Receptors, Kainic Acid/genetics , Transcription Factors/genetics , Visual Pathways/metabolism , GluK2 Kainate Receptor
7.
Nat Neurosci ; 14(4): 495-504, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21358645

ABSTRACT

The extent to which individual neurons are interconnected selectively within brain circuits is an unresolved problem in neuroscience. Neurons can be organized into preferentially interconnected microcircuits, but whether this reflects genetically defined subpopulations is unclear. We found that the principal neurons in the main subdivisions of the hippocampus consist of distinct subpopulations that are generated during distinct time windows and that interconnect selectively across subdivisions. In two mouse lines in which transgene expression was driven by the neuron-specific Thy1 promoter, transgene expression allowed us to visualize distinct populations of principal neurons with unique and matched patterns of gene expression, shared distinct neurogenesis and synaptogenesis time windows, and selective connectivity at dentate gyrus-CA3 and CA3-CA1 synapses. Matched subpopulation marker genes and neuronal subtype markers mapped near clusters of olfactory receptor genes. The nonoverlapping matched timings of synaptogenesis accounted for the selective connectivities of these neurons in CA3. Therefore, the hippocampus contains parallel connectivity channels assembled from distinct principal neuron subpopulations through matched schedules of synaptogenesis.


Subject(s)
Cell Communication/physiology , Hippocampus/embryology , Hippocampus/growth & development , Neurons/physiology , Animals , Body Patterning/genetics , Cell Communication/genetics , Gene Expression Regulation, Developmental/genetics , Growth Cones/metabolism , Growth Cones/ultrastructure , Hippocampus/cytology , Mice , Mice, Knockout , Mice, Neurologic Mutants , Mice, Transgenic , Nerve Net/cytology , Nerve Net/physiology , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/growth & development , Neurons/cytology , Organ Culture Techniques
8.
Science ; 329(5990): 413-7, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20576849

ABSTRACT

Retinitis pigmentosa refers to a diverse group of hereditary diseases that lead to incurable blindness, affecting two million people worldwide. As a common pathology, rod photoreceptors die early, whereas light-insensitive, morphologically altered cone photoreceptors persist longer. It is unknown if these cones are accessible for therapeutic intervention. Here, we show that expression of archaebacterial halorhodopsin in light-insensitive cones can substitute for the native phototransduction cascade and restore light sensitivity in mouse models of retinitis pigmentosa. Resensitized photoreceptors activate all retinal cone pathways, drive sophisticated retinal circuit functions (including directional selectivity), activate cortical circuits, and mediate visually guided behaviors. Using human ex vivo retinas, we show that halorhodopsin can reactivate light-insensitive human photoreceptors. Finally, we identified blind patients with persisting, light-insensitive cones for potential halorhodopsin-based therapy.


Subject(s)
Genetic Therapy , Halorhodopsins/genetics , Halorhodopsins/metabolism , Retinal Cone Photoreceptor Cells/physiology , Retinitis Pigmentosa/therapy , Animals , Dependovirus/genetics , Disease Models, Animal , Evoked Potentials, Visual , Genetic Vectors , Halobacteriaceae/genetics , Humans , Light , Mice , Mice, Knockout , Promoter Regions, Genetic , Retina/physiology , Retinal Ganglion Cells/physiology , Retinitis Pigmentosa/physiopathology , Tissue Culture Techniques , Transfection , Vision, Ocular , Visual Pathways/physiology
9.
Nat Neurosci ; 12(5): 627-36, 2009 May.
Article in English | MEDLINE | ID: mdl-19330001

ABSTRACT

The mechanisms underlying disease manifestations in neurodegeneration remain unclear, but their understanding is critical to devising effective therapies. We carry out a longitudinal analysis in vivo of identified motoneurons selectively vulnerable (VUL) or resistant (RES) to motoneuron disease (amyotrophic lateral sclerosis, ALS) and show that subtype-selective endoplasmic reticulum (ER) stress responses influence disease manifestations. VUL motoneurons were selectively prone to ER stress and showed gradually upregulated ER stress markers from birth on in three mouse models of familial ALS (FALS). 25-30 days before the earliest denervations, ubiquitin signals increased in both VUL and RES motoneurons, but an unfolded protein response coupled with microglial activation was initiated selectively in VUL motoneurons. This transition was followed by selective axonal degeneration and spreading stress. The ER stress-protective agent salubrinal attenuated disease manifestations and delayed progression, whereas chronic enhancement of ER stress promoted disease. Thus, whereas all motoneurons are preferentially affected in ALS, ER stress responses in specific motoneuron subtypes influence the progressive manifestations of weakening and paralysis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Central Nervous System/metabolism , Endoplasmic Reticulum/metabolism , Motor Neurons/metabolism , Oxidative Stress/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Biomarkers/analysis , Biomarkers/metabolism , Central Nervous System/pathology , Central Nervous System/physiopathology , Cinnamates/pharmacology , Disease Models, Animal , Disease Progression , Endoplasmic Reticulum/genetics , Genetic Predisposition to Disease/genetics , Gliosis/genetics , Gliosis/metabolism , Gliosis/physiopathology , Mice , Mice, Neurologic Mutants , Microglia/metabolism , Motor Neurons/classification , Motor Neurons/pathology , Neuroprotective Agents/pharmacology , Phenotype , Protein Folding , Thiourea/analogs & derivatives , Thiourea/pharmacology , Ubiquitin/metabolism , Ubiquitination , Wallerian Degeneration/genetics , Wallerian Degeneration/metabolism , Wallerian Degeneration/physiopathology
10.
Nature ; 447(7145): 686-90, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17554302

ABSTRACT

Accumulation of DNA damage leading to adult stem cell exhaustion has been proposed to be a principal mechanism of ageing. Here we address this question by taking advantage of the highly specific role of DNA ligase IV in the repair of DNA double-strand breaks by non-homologous end-joining, and by the discovery of a unique mouse strain with a hypomorphic Lig4(Y288C) mutation. The Lig4(Y288C) mouse, identified by means of a mutagenesis screening programme, is a mouse model for human LIG4 syndrome, showing immunodeficiency and growth retardation. Diminished DNA double-strand break repair in the Lig4(Y288C) strain causes a progressive loss of haematopoietic stem cells and bone marrow cellularity during ageing, and severely impairs stem cell function in tissue culture and transplantation. The sensitivity of haematopoietic stem cells to non-homologous end-joining deficiency is therefore a key determinant of their ability to maintain themselves against physiological stress over time and to withstand culture and transplantation.


Subject(s)
Aging/physiology , DNA Repair , Hematopoietic Stem Cells/cytology , Animals , Cell Proliferation , Cellular Senescence/physiology , DNA Breaks, Double-Stranded , DNA Damage , DNA Ligase ATP , DNA Ligases/deficiency , DNA Ligases/genetics , DNA Ligases/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mutation, Missense/drug effects , Mutation, Missense/genetics , Syndrome
11.
Aging Cell ; 6(2): 139-53, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17274801

ABSTRACT

A number of diseases associated with specific tissue degeneration and premature aging have mutations in the nuclear envelope proteins A-type lamins or emerin. Those diseases with A-type lamin mutation are inclusively termed laminopathies. Due to various hypothetical roles of nuclear envelope proteins in genome function we investigated whether alterations to normal genomic behaviour are apparent in cells with mutations in A-type lamins and emerin. Even though the distributions of these proteins in proliferating laminopathy fibroblasts appear normal, there is abnormal nuclear positioning of both chromosome 18 and 13 territories, from the nuclear periphery to the interior. This genomic organization mimics that found in normal nonproliferating quiescent or senescent cells. This finding is supported by distributions of modified pRb in the laminopathy cells. All laminopathy cell lines tested and an X-linked Emery-Dreifuss muscular dystrophy cell line also demonstrate increased incidences of apoptosis. The most extreme cases of apoptosis occur in cells derived from diseases with mutations in the tail region of the LMNA gene, such as Dunningan-type familial partial lipodystrophy and mandibuloacral dysplasia, and this correlates with a significant level of micronucleation in these cells.


Subject(s)
Aging, Premature/genetics , Apoptosis , Fibroblasts/ultrastructure , Genome, Human , Lamin Type A/genetics , Membrane Proteins/genetics , Nuclear Proteins/genetics , Aging, Premature/pathology , Cell Line , Cell Proliferation , Humans , Lipodystrophy, Familial Partial/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/pathology , Nuclear Envelope/ultrastructure
12.
Proc Natl Acad Sci U S A ; 102(8): 2868-73, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15710876

ABSTRACT

Apc(Min) mice have provided an example of a locus (Modifier of Min1; Mom1) modifying adenoma numbers in the intestines of inbred strains. Linkage analysis located Mom1 on chromosome 4, and further investigation identified secretory phospholipase A2 (Pla2g2a) as a candidate gene. Because of unknown variation introduced by a single founding male mouse, our Min stock, although Pla2g2a(Mom1-s), was not on a pure C57BL/6J background and exhibited several polymorphic loci, including a region on chromosome 18 distal to Apc. Through selective breeding for homozygosity for distal chromosome 18 markers, six recombinant lines that presented with limited intraline variation in adenoma numbers were established. One line (V) showed a particularly severe phenotype (mean adenoma number +/- SEM, 370 +/- 21) compared with the other lines that recorded significantly lower means (3- to 5-fold; P < 10(-3), t test). Intercrosses between lines I and V showed suppression of the severe phenotype in the N1 generation. In N2 (and subsequent) backcrosses, tumor multiplicity depended on the origins of the WT and Min Apc alleles. Mice carrying both alleles from line V had a severe phenotype; others had mild disease very similar to line I (likelihood ratio statistic > 49.0; likelihood of odds > 10; P < 10(-5)). Frequency of allele loss at Apc was increased significantly in adenomas of mice with more severe disease. We propose that a modifier gene close to Apc or structural variation on chromosome 18 modifies polyp numbers in our mice, possibly by altering the frequency of WT Apc allele loss.


Subject(s)
Adenoma/genetics , Genes, APC , Intestinal Neoplasms/genetics , Alleles , Animals , Chromosome Mapping , Female , Genetic Linkage , Genetic Predisposition to Disease , Intestinal Polyps/genetics , Male , Mice , Mice, Inbred C57BL , Phenotype
13.
Cytometry A ; 62(2): 150-61, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15523603

ABSTRACT

BACKGROUND: Telomeres are specialized nucleoprotein structures at chromosome ends that undergo dynamic changes after each cell cycle. Understanding the mechanisms of telomere dynamics is critically dependent on the ability to accurately measure telomere length in a cell population of interest. Techniques such as Southern blot, which measures average telomere length, and quantitative fluorescence in situ hybridization (Q-FISH), which can estimate telomere length in individual chromosomes, are limited in their capacity to determine the distribution of cells with differing telomere lengths in a given cell population. METHODS: We employed flow-FISH to determine whether mouse and human cell lines exhibit subpopulations of cells with differing telomere lengths. RESULTS: Our analysis showed that at least one of four analyzed mouse cell lines had two subpopulations of cells with differing telomere lengths. Differences in telomere length between subpopulations of cells were significant, and we term this phenomenon TELEFLUCS (TElomere LEngth FLUctuations in Cell Subpopulations). We also observed TELEFLUCS in 1 of 19 analyzed human nonalternative lengthening of telomere cell lines and in 1 of 2 analyzed human alternative lengthening of telomere cell lines. The existence of cell subpopulations with differing telomere lengths was confirmed by Q-FISH. CONCLUSION: Our results underscore the importance of flow-FISH in telomere length analysis.


Subject(s)
Fibroblasts/cytology , Telomere , Animals , Cell Line, Tumor , Flow Cytometry , Humans , In Situ Hybridization, Fluorescence , Mice , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism
14.
J Biochem ; 134(5): 675-82, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14688233

ABSTRACT

The Sd(a) antigen is a carbohydrate determinant expressed on erythrocytes, the colonic mucosa and other tissues. This epitope, whose structure is Siaalpha2,3[GalNAcbeta1,4]Gal beta1,4GlcNAc, is synthesized by a beta1,4 N-acetylgalactosaminyltransferase (beta4GalNAc-T) that transfers a beta1,4-linked GalNAc to the galactose residue of an alpha2,3-sialylated chain. We have cloned from human colon carcinoma Caco2 cells a cDNA whose transfection in COS cells induces a GalNAc-T active on sialylated but not on asialylated fetuin and putatively represents the human Sd(a) beta4GalNAc-T. The cDNA predicts a 566 aa protein showing 66.6% and 39% identity with mouse CT beta4GalNAc-T and human GM2/GD2 synthase, respectively, with a typical type II glycosyltransferase organization, no potential N-glycosylation sites and a 67 aa cytoplasmic tail, which is probably the longest among the glycosyltransferases cloned to date. The gene maps in chromosome 17q23, and is composed of at least 11 exons. Exons 2-11 are homologous to exons 2-11 of the previously cloned CT beta4GalNAc-T from murine cytotoxic T lymphocytes while exons 1 of the two enzymes are totally different. The mRNA is expressed at a high level in differentiated Caco2 cells and in colonic mucosa and at a much lower level in lymphocytes and other colon cancer cell lines.


Subject(s)
Blood Group Antigens/biosynthesis , Cytoplasm/metabolism , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Oligosaccharides/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Blood Group Antigens/metabolism , COS Cells , Caco-2 Cells , Chromosomes, Human, Pair 17/genetics , Cloning, Molecular , DNA, Complementary/genetics , Exons/genetics , Gene Expression Regulation , Genomics , Humans , Introns/genetics , Mice , Molecular Sequence Data , Oligosaccharides/metabolism , Open Reading Frames/genetics , Physical Chromosome Mapping , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
J Morphol ; 240(2): 169-194, 1999 May.
Article in English | MEDLINE | ID: mdl-29847871

ABSTRACT

We compare the cranial morphology of four fish species with an increasing anguilliformism in the following order: Clarias gariepinus, Clariallabes melas, Gymnallabes typus, and Channallabes apus. The main anatomical-morphological disparities are the stepwise reduction of the skull roof along with the relative enlargement of the external jaw muscles, which occurred in each of them. Gymnallabes typus and C. apus lack a bony protection to cover the jaw muscles. The neurocranial bones of C. gariepinus, however, form a closed, broad roof, whereas the width of the neurocranium in C. melas is intermediate. Several features of the clariid heads, such as the size of the mouth and the bands of small teeth, may be regarded as adaptations for manipulating large food particles, which are even more pronounced in anguilliform clariids. The jaw musculature of G. typus is hypertrophied and attached on a higher coronoid process of the lower jaw, causing a larger adductive force. The hyomandibula interdigitates more strongly with the neurocranium and its dentition with longer teeth is posteriorly extended, closer to the lower jaw articulation. The anguilliform clariids also have their cranial muscles modified to enable a wider gape. The adductor mandibulae and the levator operculi extend more posteriorly, and the anterior attachment site of the protractor hyoidei dorsalis shifts toward the sagittal plane of the head. A phylogenetic analysis of the Clariidae, which is in progress, could check the validity of Boulenger's hypothesis that predecessors of the primitive fishes, such as Heterobranchus and most Clarias, would have evolved into progressively anguilliform clariids. J. Morphol. 240:169-194, 1999. © 1999 Wiley-Liss, Inc.

SELECTION OF CITATIONS
SEARCH DETAIL