Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 18(7): 762-770, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28504698

ABSTRACT

Trafficking of tissue dendritic cells (DCs) via lymph is critical for the generation of cellular immune responses in draining lymph nodes (LNs). In the current study we found that DCs docked to the basolateral surface of lymphatic vessels and transited to the lumen through hyaluronan-mediated interactions with the lymph-specific endothelial receptor LYVE-1, in dynamic transmigratory-cup-like structures. Furthermore, we show that targeted deletion of the gene Lyve1, antibody blockade or depletion of the DC hyaluronan coat not only delayed lymphatic trafficking of dermal DCs but also blunted their capacity to prime CD8+ T cell responses in skin-draining LNs. Our findings uncovered a previously unknown function for LYVE-1 and show that transit through the lymphatic network is initiated by the recognition of leukocyte-derived hyaluronan.


Subject(s)
Dendritic Cells/immunology , Endothelial Cells/metabolism , Glycoproteins/genetics , Hyaluronic Acid/metabolism , Lymphatic Vessels/metabolism , Vesicular Transport Proteins/metabolism , Animals , Cell Movement/immunology , Dendritic Cells/metabolism , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/metabolism , Flow Cytometry , Glycoproteins/metabolism , Humans , Immunity, Cellular/immunology , Lymph Nodes/immunology , Membrane Transport Proteins , Mice , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
2.
Hum Mol Genet ; 29(21): 3516-3531, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33105479

ABSTRACT

Neurodevelopmental disorder with microcephaly, hypotonia and variable brain anomalies (NMIHBA) is an autosomal recessive neurodevelopmental and neurodegenerative disorder characterized by global developmental delay and severe intellectual disability. Microcephaly, progressive cortical atrophy, cerebellar hypoplasia and delayed myelination are neurological hallmarks in affected individuals. NMIHBA is caused by biallelic variants in PRUNE1 encoding prune exopolyphosphatase 1. We provide in-depth clinical description of two affected siblings harboring compound heterozygous variant alleles, c.383G > A (p.Arg128Gln), c.520G > T (p.Gly174*) in PRUNE1. To gain insights into disease biology, we biochemically characterized missense variants within the conserved N-terminal aspartic acid-histidine-histidine (DHH) motif and provide evidence that they result in the destabilization of protein structure and/or loss of exopolyphosphatase activity. Genetic ablation of Prune1 results in midgestational lethality in mice, associated with perturbations to embryonic growth and vascular development. Our findings suggest that NMIHBA results from hypomorphic variant alleles in humans and underscore the potential key role of PRUNE1 exopolyphoshatase activity in neurodevelopment.


Subject(s)
Acid Anhydride Hydrolases/deficiency , Intellectual Disability/pathology , Microcephaly/pathology , Muscle Hypotonia/pathology , Mutation , Neurodevelopmental Disorders/pathology , Phosphoric Monoester Hydrolases/genetics , Alleles , Animals , Child, Preschool , Female , Humans , Infant , Intellectual Disability/etiology , Intellectual Disability/metabolism , Male , Mice , Microcephaly/etiology , Microcephaly/metabolism , Muscle Hypotonia/etiology , Muscle Hypotonia/metabolism , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Pedigree , Phenotype
3.
Mol Ther ; 29(12): 3512-3524, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34400331

ABSTRACT

Lysosomal diseases are a class of genetic disorders predominantly caused by loss of lysosomal hydrolases, leading to lysosomal and cellular dysfunction. Enzyme replacement therapy (ERT), where recombinant enzyme is given intravenously, internalized by cells, and trafficked to the lysosome, has been applied to treat several lysosomal diseases. However, current ERT regimens do not correct disease phenotypes in all affected organs because the biodistribution of enzyme uptake does not match that of the affected cells that require the enzyme. We present here targeted ERT, an approach that utilizes antibody-enzyme fusion proteins to target the enzyme to specific cell types. The antibody moiety recognizes transmembrane proteins involved in lysosomal trafficking and that are also preferentially expressed in those cells most affected in disease. Using Pompe disease (PD) as an example, we show that targeted ERT is superior to ERT in treating the skeletal muscle phenotypes of PD mice both as a protein replacement therapeutic and as a gene therapy.


Subject(s)
Glycogen Storage Disease Type II , Lysosomal Storage Diseases , Animals , Enzyme Replacement Therapy , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/genetics , Hydrolases/metabolism , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/genetics , Lysosomes/metabolism , Mice , Tissue Distribution , alpha-Glucosidases/genetics
4.
Angiogenesis ; 23(2): 179-192, 2020 05.
Article in English | MEDLINE | ID: mdl-31754927

ABSTRACT

Angiogenesis is largely driven by motile endothelial tip-cells capable of invading avascular tissue domains and enabling new vessel formation. Highly responsive to Vascular Endothelial Growth-Factor-A (VEGFA), endothelial tip-cells also suppress angiogenic sprouting in adjacent stalk cells, and thus have been a primary therapeutic focus in addressing neovascular pathologies. Surprisingly, however, there remains a paucity of specific endothelial tip-cell markers. Here, we employ transcriptional profiling and a lacZ reporter allele to identify Kcne3 as an early and selective endothelial tip-cell marker in multiple angiogenic contexts. In development, Kcne3 expression initiates during early phases of angiogenesis (E9) and remains specific to endothelial tip-cells, often adjacent to regions expressing VEGFA. Consistently, Kcne3 activation is highly responsive to exogenous VEGFA but maintains tip-cell specificity throughout normal retinal angiogenesis. We also demonstrate endothelial tip-cell selectivity of Kcne3 in several injury and tumor models. Together, our data show that Kcne3 is a unique marker of sprouting angiogenic tip-cells and offers new opportunities for investigating and targeting this cell type.


Subject(s)
Endothelial Cells/physiology , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Potassium Channels, Voltage-Gated/genetics , Vascular Endothelial Growth Factor A/physiology , Animals , Animals, Newborn , Cell Differentiation/genetics , Cells, Cultured , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Embryo, Mammalian , Endothelial Cells/pathology , Female , Gene Expression Regulation, Developmental/drug effects , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Morphogenesis/genetics , Neovascularization, Pathologic/metabolism , Pregnancy , Retina/metabolism , Retina/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology
5.
Biol Reprod ; 100(3): 686-696, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30289441

ABSTRACT

The Adisintegrin and metalloprotease domain-containing (ADAM) family of proteins is involved in cell adhesion, migration, proteolysis, and signaling. Many ADAMs are required for reproduction; however, the role of Adam6 has remained largely unknown. In the course of humanizing the mouse immunoglobulin heavy chain (IgH) locus, we generated Adam6-deficient mice that demonstrate severe subfertility. We decided to elucidate the role of ADAM6 in fertility and explore the underlying mechanisms. Despite normal sperm development and motility, Adam6-deficient mice display diminished male fertility, have abnormal sperm adhesion, and most importantly cannot transition from uterus to oviduct. To test whether ADAM6 is required for sperm's binding to extracellular matrix (ECM) components, we used a panel of ECM components and showed that unlike normal sperm, Adam6-deficient sperm cannot bind fibronectin, laminin, and tenascin. Reintroduction of Adam6 into these deficient mice repaired sperm interaction with ECM, restored male fertility, and corrected the sperm transport deficit. Together, our data suggest that ADAM6, either alone or in complex with other proteins, aids sperm transport through the female reproductive tract by providing a temporary site of attachment of sperm to ECM components prior to ascent into the oviduct.


Subject(s)
ADAM Proteins/metabolism , Infertility, Male/genetics , Sperm Motility/physiology , Spermatozoa/physiology , ADAM Proteins/genetics , Animals , Female , Male , Mice , Mice, Knockout , Oviducts , Sperm Motility/genetics
6.
Immunity ; 30(1): 67-79, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19110448

ABSTRACT

Notch1 signaling is required for T cell development and has been implicated in fate decisions in the thymus. We showed that Notch1 deletion in progenitor T cells (pro-T cells) revealed their latent developmental potential toward becoming conventional and plasmacytoid dendritic cells. In addition, Notch1 deletion in pro-T cells resulted in large numbers of thymic B cells, previously explained by T-to-B cell fate conversion. Single-cell genotyping showed, however, that the majority of these thymic B cells arose from Notch1-sufficient cells by a cell-extrinsic pathway. Fate switching nevertheless exists for a subset of thymic B cells originating from Notch1-deleted pro-T cells. Chimeric mice lacking the Notch ligand delta-like 4 (Dll4) in thymus epithelium revealed an essential role for Dll4 in T cell development. Thus, Notch1-Dll4 signaling fortifies T cell commitment by suppressing non-T cell lineage potential in pro-T cells, and normal Notch1-driven T cell development repels excessive B cells in the thymus.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Deletion , Receptor, Notch1/genetics , T-Lymphocytes/immunology , Thymus Gland/cytology , Animals , Cell Lineage , Flow Cytometry , Mice , Mice, Inbred C57BL , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Proc Natl Acad Sci U S A ; 112(6): 1845-9, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25624481

ABSTRACT

G protein-coupled receptor 17 (GPR17) was recently reported to be a Foxo1 target in agouti-related peptide (AGRP) neurons. Intracerebroventricular injection of GPR17 agonists induced food intake, whereas administration of an antagonist to the receptor reduced feeding. These data lead to the conclusion that pharmacological modulation of GPR17 has therapeutic potential to treat obesity. Here we report that mice deficient in Gpr17 (Gpr17(-/-)) have similar food intake and body weight compared with their wild-type littermates. Gpr17(-/-) mice have normal hypothalamic Agrp mRNA expression, AGRP plasma levels, and metabolic rate. GPR17 deficiency in mice did not affect glucose homeostasis or prevent fat-induced insulin resistance. These data do not support a role for GPR17 in the control of food intake, body weight, or glycemic control.


Subject(s)
Eating/genetics , Glucose/metabolism , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Agouti-Related Protein/metabolism , Analysis of Variance , Animals , Base Sequence , Body Composition/drug effects , Energy Metabolism/genetics , Energy Metabolism/physiology , Mice , Mice, Knockout , Molecular Sequence Data , Neurons/metabolism , Sequence Analysis, RNA , Time Factors , X-Ray Microtomography
8.
Proc Natl Acad Sci U S A ; 110(34): E3179-88, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918385

ABSTRACT

Conditional mutagenesis is becoming a method of choice for studying gene function, but constructing conditional alleles is often laborious, limited by target gene structure, and at times, prone to incomplete conditional ablation. To address these issues, we developed a technology termed conditionals by inversion (COIN). Before activation, COINs contain an inverted module (COIN module) that lies inertly within the antisense strand of a resident gene. When inverted into the sense strand by a site-specific recombinase, the COIN module causes termination of the target gene's transcription and simultaneously provides a reporter for tracking this event. COIN modules can be inserted into natural introns (intronic COINs) or directly into coding exons as part of an artificial intron (exonic COINs), greatly simplifying allele design and increasing flexibility over previous conditional KO approaches. Detailed analysis of over 20 COIN alleles establishes the reliability of the method and its broad applicability to any gene, regardless of exon-intron structure. Our extensive testing provides rules that help ensure success of this approach and also explains why other currently available conditional approaches often fail to function optimally. Finally, the ability to split exons using the COIN's artificial intron opens up engineering modalities for the generation of multifunctional alleles.


Subject(s)
Alleles , Gene Silencing , Genetic Engineering/methods , Mutagenesis, Insertional/methods , Sequence Inversion/genetics , DNA Nucleotidyltransferases/metabolism
9.
Dev Biol ; 383(1): 90-105, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23994639

ABSTRACT

WNT signaling is critical in most aspects of skeletal development and homeostasis, and antagonists of WNT signaling are emerging as key regulatory proteins with great promise as therapeutic agents for bone disorders. Here we show that Sost and its paralog Sostdc1 emerged through ancestral genome duplication and their expression patterns have diverged to delineate non-overlapping domains in most organ systems including musculoskeletal, cardiovascular, nervous, digestive, reproductive and respiratory. In the developing limb, Sost and Sostdc1 display dynamic expression patterns with Sost being restricted to the distal ectoderm and Sostdc1 to the proximal ectoderm and the mesenchyme. While Sostdc1(-/-) mice lack any obvious limb or skeletal defects, Sost(-/-) mice recapitulate the hand defects described for Sclerosteosis patients. However, elevated WNT signaling in Sost(-/-); Sostdc1(-/-) mice causes misregulation of SHH signaling, ectopic activation of Sox9 in the digit 1 field and preaxial polydactyly in a Gli1- and Gli3-dependent manner. In addition, we show that the syndactyly documented in Sclerosteosis is present in both Sost(-/-) and Sost(-/-); Sostdc1(-/-) mice, and is driven by misregulation of Fgf8 in the AER, a region lacking Sost and Sostdc1 expression. This study highlights the complexity of WNT signaling in skeletal biology and disease and emphasizes how redundant mechanism and non-cell autonomous effects can synergize to unveil new intricate phenotypes caused by elevated WNT signaling.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Ectoderm/embryology , Extremities/embryology , Gene Expression Regulation, Developmental/physiology , Glycoproteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Wnt Signaling Pathway/physiology , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Computational Biology , Ectoderm/metabolism , Evolution, Molecular , Glycoproteins/genetics , Hedgehog Proteins/metabolism , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Microarray Analysis , SOX9 Transcription Factor/metabolism , Zinc Finger Protein Gli3
10.
J Cell Sci ; 125(Pt 21): 5159-67, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22899709

ABSTRACT

Blood vessels deliver oxygen, nutrients, hormones and immunity factors throughout the body. To perform these vital functions, vascular cords branch, lumenize and interconnect. Yet, little is known about the cellular, molecular and physiological mechanisms that control how circulatory networks form and interconnect. Specifically, how circulatory networks merge by interconnecting 'in parallel' along their boundaries remains unexplored. To examine this process we studied the formation and functional maturation of the plexus that forms between the dorsal longitudinal anastomotic vessels (DLAVs) in the zebrafish. We find that the migration and proliferation of endothelial cells within the DLAVs and their segmental (Se) vessel precursors drives DLAV plexus formation. Remarkably, the presence of Se vessels containing only endothelial cells of the arterial lineage is sufficient for DLAV plexus morphogenesis, suggesting that endothelial cells from the venous lineage make a dispensable or null contribution to this process. The discovery of a circuit that integrates the inputs of circulatory flow and vascular endothelial growth factor (VEGF) signaling to modulate aortic arch angiogenesis, together with the expression of components of this circuit in the trunk vasculature, prompted us to investigate the role of these inputs and their relationship during DLAV plexus formation. We find that circulatory flow and VEGF signaling make additive contributions to DLAV plexus morphogenesis, rather than acting as essential inputs with equivalent contributions as they do during aortic arch angiogenesis. Our observations underscore the existence of context-dependent differences in the integration of physiological stimuli and signaling cascades during vascular development.


Subject(s)
Arteriovenous Anastomosis/embryology , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/metabolism , Animals , Arteriovenous Anastomosis/cytology , Cell Movement , Cell Proliferation , Endothelial Cells/physiology , Mice , Morphogenesis , Torso/blood supply , Torso/embryology , Vascular Endothelial Growth Factor A/physiology , Zebrafish
11.
Proc Natl Acad Sci U S A ; 108(7): 2807-12, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282641

ABSTRACT

The vasculature of the CNS is structurally and functionally distinct from that of other organ systems and is particularly prone to developmental abnormalities and hemorrhage. Although other embryonic tissues undergo primary vascularization, the developing nervous system is unique in that it is secondarily vascularized by sprouting angiogenesis from a surrounding perineural plexus. This sprouting angiogenesis requires the TGF-ß and Wnt pathways because ablation of these pathways results in aberrant sprouting and hemorrhage. We have genetically deleted Gpr124, a member of the large family of long N-terminal group B G protein-coupled receptors, few members of which have identified ligands or well-defined biologic functions in mammals. We show that, in the developing CNS, Gpr124 is specifically expressed in the vasculature and is absolutely required for proper angiogenic sprouting into the developing neural tube. Embryos lacking Gpr124 exhibit vascular defects characterized by delayed vascular penetration, formation of pathological glomeruloid tufts within the CNS, and hemorrhage. In addition, they display defects in palate and lung development, two processes in which TGF-ß and/or Wnt pathways also play important roles. We also show that TGF-ß stimulates Gpr124 expression, and ablation of Gpr124 results in perturbed TGF-ß pathway activation, suggesting roles for Gpr124 in modulating TGF-ß signaling. These results represent a unique function attributed to a long N-terminal group B-type G protein-coupled receptor in a mammalian system.


Subject(s)
Central Nervous System/blood supply , Central Nervous System/embryology , Neovascularization, Physiologic/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Embryo, Mammalian , Genetic Engineering , Histological Techniques , Immunohistochemistry , In Situ Hybridization , Lung/embryology , Lung/metabolism , Mice , Microarray Analysis , Palate/embryology , Palate/metabolism , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/physiology , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism
12.
Nat Med ; 12(2): 235-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16462802

ABSTRACT

The angiopoietins Ang-1 and Ang-2 have been identified as ligands of the receptor tyrosine kinase Tie-2 (refs. 1,2). Paracrine Ang-1-mediated activation of Tie-2 acts as a regulator of vessel maturation and vascular quiescence. In turn, the antagonistic ligand Ang-2 acts by an autocrine mechanism and is stored in endothelial Weibel-Palade bodies from where it can be rapidly released upon stimulation. The rapid release of Ang-2 implies functions of the angiopoietin-Tie system beyond its established role during vascular morphogenesis as a regulator of rapid vascular responses. Here we show that mice deficient in Ang-2 (encoded by the gene Angpt2) cannot elicit an inflammatory response in thioglycollate-induced or Staphylococcus aureus-induced peritonitis, or in the dorsal skinfold chamber model. Recombinant Ang-2 restores the inflammation defect in Angpt2(-/-) mice. Intravital microscopy showed normal TNF-alpha-induced leukocyte rolling in the vasculature of Angpt2(-/-)mice, but rolling cells did not firmly adhere to activated endothelium. Cellular experiments showed that Ang-2 promotes adhesion by sensitizing endothelial cells toward TNF-alpha and modulating TNF-alpha-induced expression of endothelial cell adhesion molecules. Together, these findings identify Ang-2 as an autocrine regulator of endothelial cell inflammatory responses. Ang-2 thereby acts as a switch of vascular responsiveness exerting a permissive role for the activities of proinflammatory cytokines.


Subject(s)
Angiopoietin-2/physiology , Inflammation/etiology , Tumor Necrosis Factor-alpha/physiology , Angiopoietin-1/physiology , Angiopoietin-2/deficiency , Angiopoietin-2/genetics , Animals , Cytokines/physiology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/physiology , Mice , Mice, Knockout , Models, Biological , Neovascularization, Pathologic , Signal Transduction
13.
Nature ; 449(7160): 346-50, 2007 Sep 20.
Article in English | MEDLINE | ID: mdl-17882221

ABSTRACT

Adult mammalian testis is a source of pluripotent stem cells. However, the lack of specific surface markers has hampered identification and tracking of the unrecognized subset of germ cells that gives rise to multipotent cells. Although embryonic-like cells can be derived from adult testis cultures after only several weeks in vitro, it is not known whether adult self-renewing spermatogonia in long-term culture can generate such stem cells as well. Here, we show that highly proliferative adult spermatogonial progenitor cells (SPCs) can be efficiently obtained by cultivation on mitotically inactivated testicular feeders containing CD34+ stromal cells. SPCs exhibit testicular repopulating activity in vivo and maintain the ability in long-term culture to give rise to multipotent adult spermatogonial-derived stem cells (MASCs). Furthermore, both SPCs and MASCs express GPR125, an orphan adhesion-type G-protein-coupled receptor. In knock-in mice bearing a GPR125-beta-galactosidase (LacZ) fusion protein under control of the native Gpr125 promoter (GPR125-LacZ), expression in the testis was detected exclusively in spermatogonia and not in differentiated germ cells. Primary GPR125-LacZ SPC lines retained GPR125 expression, underwent clonal expansion, maintained the phenotype of germline stem cells, and reconstituted spermatogenesis in busulphan-treated mice. Long-term cultures of GPR125+ SPCs (GSPCs) also converted into GPR125+ MASC colonies. GPR125+ MASCs generated derivatives of the three germ layers and contributed to chimaeric embryos, with concomitant downregulation of GPR125 during differentiation into GPR125- cells. MASCs also differentiated into contractile cardiac tissue in vitro and formed functional blood vessels in vivo. Molecular bookmarking by GPR125 in the adult mouse and, ultimately, in the human testis could enrich for a population of SPCs for derivation of GPR125+ MASCs, which may be employed for genetic manipulation, tissue regeneration and revascularization of ischaemic organs.


Subject(s)
Adult Stem Cells/cytology , Multipotent Stem Cells/cytology , Receptors, G-Protein-Coupled/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Adult Stem Cells/metabolism , Aging , Animals , Blood Vessels/cytology , Busulfan , Cell Differentiation , Cell Line , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/metabolism , Myocardium/cytology , Regeneration , Testis/cytology , Testis/metabolism
14.
eNeuro ; 10(11)2023 Nov.
Article in English | MEDLINE | ID: mdl-37852780

ABSTRACT

It has been suggested that stochasticity acts in the formation of topographically ordered maps in the visual system through the opposing chemoaffinity and neural activity forces acting on the innervating nerve fibers being held in an unstable equilibrium. Evidence comes from the Islet2-EphA3 knock-in mouse, in which ∼50% of the retinal ganglion cells, distributed across the retina, acquire the EphA3 receptor, thus having an enhanced density of EphA which specifies retinotopic order along the rostrocaudal (RC) axis of the colliculus. Sampling EphA3 knock-in maps in heterozygotes at different positions along the mediolateral (ML) extent of the colliculus had found single 1D maps [as in wild types (WTs)], double maps (as in homozygous knock-ins) or both single and double maps. We constructed full 2D maps from the same mouse dataset. We found either single maps or maps where the visual field projects rostrally, with a part-projection more caudally to form a double map, the extent and location of this duplication varying considerably. Contrary to previous analyses, there was no strict demarcation between heterozygous and homozygous maps. These maps were replicated in a computational model where, as the level of EphA3 was increased, there was a smooth transition from single to double maps. Our results suggest that the diversity in these retinotopic maps has its origin in a variability over the retina in the effective amount of EphA3, such as through variability in gene expression or the proportion of EphA3+ retinal ganglion cells, rather than the result of competing mechanisms acting at the colliculus.


Subject(s)
Superior Colliculi , Visual Pathways , Mice , Animals , Receptor, EphA3/genetics , Receptor, EphA3/metabolism , Superior Colliculi/metabolism , Visual Pathways/physiology , Retina/metabolism , Retinal Ganglion Cells/metabolism
15.
Nature ; 444(7122): 1032-7, 2006 Dec 21.
Article in English | MEDLINE | ID: mdl-17183313

ABSTRACT

Tumour growth requires accompanying expansion of the host vasculature, with tumour progression often correlated with vascular density. Vascular endothelial growth factor (VEGF) is the best-characterized inducer of tumour angiogenesis. We report that VEGF dynamically regulates tumour endothelial expression of Delta-like ligand 4 (Dll4), which was previously shown to be absolutely required for normal embryonic vascular development. To define Dll4 function in tumour angiogenesis, we manipulated this pathway in murine tumour models using several approaches. Here we show that blockade resulted in markedly increased tumour vascularity, associated with enhanced angiogenic sprouting and branching. Paradoxically, this increased vascularity was non-productive-as shown by poor perfusion and increased hypoxia, and most importantly, by decreased tumour growth-even for tumours resistant to anti-VEGF therapy. Thus, VEGF-induced Dll4 acts as a negative regulator of tumour angiogenesis; its blockade results in a striking uncoupling of tumour growth from vessel density, presenting a novel therapeutic approach even for tumours resistant to anti-VEGF therapies.


Subject(s)
Membrane Proteins/metabolism , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic , Adenoviridae/genetics , Animals , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation , Genes, Reporter/genetics , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Rats , Receptors, Notch/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
16.
Proc Natl Acad Sci U S A ; 106(52): 22399-404, 2009 Dec 29.
Article in English | MEDLINE | ID: mdl-20018779

ABSTRACT

Inhibiting angiogenesis has become an effective approach for treating cancer and other diseases. However, our understanding of signaling pathways in tumor angiogenesis has been limited by the embryonic lethality of many gene knockouts. To overcome this limitation, we used the plasticity of embryonic stem (ES) cells to develop a unique approach to study tumor angiogenesis. Murine ES cells can be readily manipulated genetically; in addition, ES cells implanted subcutaneously in mice develop into tumors that contain a variety of cell types (teratomas). We show that ES cells differentiate into bona fide endothelial cells within the teratoma, and that these ES-derived endothelial cells form part of the functional tumor vasculature. Using this powerful and flexible system, the Angiopoietin/Tie2 system is shown to have a key role in the regulation of tumor vessel size. Endothelial differentiation in the ES teratoma model allows gene-targeting methods to be used in the study of tumor angiogenesis.


Subject(s)
Embryonic Stem Cells/enzymology , Embryonic Stem Cells/pathology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/enzymology , Neovascularization, Pathologic , Receptor Protein-Tyrosine Kinases/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/physiology , Angiopoietins/antagonists & inhibitors , Animals , Cell Differentiation , Cell Line , Disease Models, Animal , Endothelial Cells/enzymology , Endothelial Cells/pathology , Mice , Mice, SCID , Neoplasms, Experimental/etiology , Receptor, TIE-2 , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Teratoma/blood supply , Teratoma/enzymology , Teratoma/etiology , Vascular Endothelial Growth Factor Receptor-2/physiology
17.
J Clin Invest ; 118(6): 2111-20, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18497886

ABSTRACT

Colon cancer stem cells are believed to originate from a rare population of putative CD133+ intestinal stem cells. Recent publications suggest that a small subset of colon cancer cells expresses CD133, and that only these CD133+ cancer cells are capable of tumor initiation. However, the precise contribution of CD133+ tumor-initiating cells in mediating colon cancer metastasis remains unknown. Therefore, to temporally and spatially track the expression of CD133 in adult mice and during tumorigenesis, we generated a knockin lacZ reporter mouse (CD133lacZ/+), in which the expression of lacZ is driven by the endogenous CD133 promoters. Using this model and immunostaining, we discovered that CD133 expression in colon is not restricted to stem cells; on the contrary, CD133 is ubiquitously expressed on differentiated colonic epithelium in both adult mice and humans. Using Il10-/-CD133lacZ mice, in which chronic inflammation in colon leads to adenocarcinomas, we demonstrated that CD133 is expressed on a full gamut of colonic tumor cells, which express epithelial cell adhesion molecule (EpCAM). Similarly, CD133 is widely expressed by human primary colon cancer epithelial cells, whereas the CD133- population is composed mostly of stromal and inflammatory cells. Conversely, CD133 expression does not identify the entire population of epithelial and tumor-initiating cells in human metastatic colon cancer. Indeed, both CD133+ and CD133- metastatic tumor subpopulations formed colonospheres in in vitro cultures and were capable of long-term tumorigenesis in a NOD/SCID serial xenotransplantation model. Moreover, metastatic CD133- cells form more aggressive tumors and express typical phenotypic markers of cancer-initiating cells, including CD44 (CD44+CD24-), whereas the CD133+ fraction is composed of CD44lowCD24+ cells. Collectively, our data suggest that CD133 expression is not restricted to intestinal stem or cancer-initiating cells, and during the metastatic transition, CD133+ tumor cells might give rise to the more aggressive CD133(- )subset, which is also capable of tumor initiation in NOD/SCID mice.


Subject(s)
Antigens, CD/biosynthesis , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glycoproteins/biosynthesis , Stem Cells/metabolism , AC133 Antigen , Animals , Epithelial Cells/metabolism , Inflammation , Mice , Mice, SCID , Mice, Transgenic , Models, Biological , Models, Genetic , Neoplasm Metastasis , Peptides , Phenotype , Promoter Regions, Genetic , Stem Cells/cytology
18.
Circ Res ; 105(2): 201-8, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19542015

ABSTRACT

Blood vessel formation is controlled by the balance between pro- and antiangiogenic pathways. Although much is known about the factors that drive sprouting of neovessels, the factors that stabilize and pattern neovessels are undefined. The expression of angiomodulin (AGM), a vascular endothelial growth factor (VEGF)-A binding protein, was increased in the vasculature of several human tumors as compared to normal tissue, raising the hypothesis that AGM may modulate VEGF-A-dependent vascular patterning. To elucidate the expression pattern of AGM, we developed an AGM knockin reporter mouse (AGM(lacZ/+)), with which we demonstrate that AGM is predominantly expressed in the vasculature of developing embryos and adult organs. During physiological and pathological angiogenesis, AGM is upregulated in the angiogenic vasculature. Using the zebrafish model, we found that AGM is restricted to developing vasculature by 17 to 22 hours postfertilization. Blockade of AGM activity with morpholino oligomers results in prominent angiogenesis defects in vascular sprouting and remodeling. Concurrent knockdown of both AGM and VEGF-A results in synergistic angiogenesis defects. When VEGF-A is overexpressed, the compensatory induction of the VEGF-A receptor, VEGFR2/flk-1, is blocked by the simultaneous injection of AGM morpholino oligomers. These results demonstrate that the vascular-specific marker AGM modulates vascular remodeling in part by temporizing the proangiogenic effects of VEGF-A.


Subject(s)
Neoplasm Proteins/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Retinal Neovascularization/metabolism , Skin/blood supply , Vascular Endothelial Growth Factor A/metabolism , Zebrafish Proteins/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genotype , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morpholines/metabolism , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/genetics , Oligonucleotides, Antisense/metabolism , Phenotype , Promoter Regions, Genetic , Retinal Neovascularization/genetics , Retinal Neovascularization/physiopathology , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing , Zebrafish/embryology , Zebrafish Proteins/genetics
19.
Mol Cell Biol ; 27(2): 595-604, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17101772

ABSTRACT

The hyaluronan receptor LYVE-1 is expressed abundantly on the surfaces of lymphatic vessels and lymph node sinus endothelial cells from early development, where it has been suggested to function both in cell adhesion/transmigration and as a scavenger for hyaluronan turnover. To investigate the physiological role(s) of LYVE-1, we generated mice in which the gene for the receptor was inactivated by replacement with a beta-galactosidase reporter. LYVE-1(-/-) mice displayed an apparently normal phenotype, with no obvious alteration in lymphatic vessel ultrastructure or function and no apparent change in secondary lymphoid tissue structure or cellularity. In addition, the levels of hyaluronan in tissue and blood were unchanged. LYVE-1(-/-) mice also displayed normal trafficking of cutaneous CD11c(+) dendritic cells to draining lymph nodes via afferent lymphatics and normal resolution of oxazolone-induced skin inflammation. Finally, LYVE-1(-/-) mice supported normal growth of transplanted B16F10 melanomas and Lewis lung carcinomas. These results indicate that LYVE-1 is not obligatory for normal lymphatic development and function and suggest either the existence of compensatory receptors or a role more specific than that previously envisaged.


Subject(s)
Glycoproteins/physiology , Hyaluronic Acid/metabolism , Lymph Nodes/physiology , Lymphatic Vessels/physiology , Animals , CD11c Antigen/metabolism , Carcinoma, Lewis Lung/pathology , Cell Movement , Dendritic Cells/physiology , Dermatitis, Contact/etiology , Dermatitis, Contact/immunology , Glycoproteins/genetics , Hyaluronic Acid/blood , Inflammation/chemically induced , Inflammation/immunology , Lymph Nodes/cytology , Lymph Nodes/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Melanoma/pathology , Membrane Transport Proteins , Mice , Mice, Knockout , Neoplasm Transplantation , Oxazolone , beta-Galactosidase/genetics
20.
Nat Biotechnol ; 25(1): 91-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187059

ABSTRACT

A useful approach for exploring gene function involves generating mutant mice from genetically modified embryonic stem (ES) cells. Recent advances in genetic engineering of ES cells have shifted the bottleneck in this process to the generation of mice. Conventional injections of ES cells into blastocyst hosts produce F0 generation chimeras that are only partially derived from ES cells, requiring additional breeding to obtain mutant mice that can be phenotyped. The tetraploid complementation approach directly yields mice that are almost entirely derived from ES cells, but it is inefficient, works only with certain hybrid ES cell lines and suffers from nonspecific lethality and abnormalities, complicating phenotypic analyses. Here we show that laser-assisted injection of either inbred or hybrid ES cells into eight cell-stage embryos efficiently yields F0 generation mice that are fully ES cell-derived and healthy, exhibit 100% germline transmission and allow immediate phenotypic analysis, greatly accelerating gene function assignment.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Gene Targeting/methods , Laser Therapy/methods , Mice, Transgenic/genetics , Microinjections/methods , Stem Cell Transplantation/methods , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic/anatomy & histology , Mice, Transgenic/surgery , Microsurgery/methods , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL