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










Publication year range
1.
Dev Cell ; 57(20): 2426-2443.e6, 2022 10 24.
Article in English | MEDLINE | ID: mdl-36283392

ABSTRACT

Smooth muscle cells (SMCs) execute important physiological functions in numerous vital organ systems, including the vascular, gastrointestinal, respiratory, and urogenital tracts. SMC differ morphologically and functionally at these different anatomical locations, but the molecular underpinnings of the differences remain poorly understood. Here, using deep single-cell RNA sequencing combined with in situ gene and protein expression analysis in four murine organs-heart, aorta, lung, and colon-we identify a molecular basis for high-level differences among vascular, visceral, and airway SMC, as well as more subtle differences between, for example, SMC in elastic and muscular arteries and zonation of elastic artery SMC along the direction of blood flow. Arterial SMC exhibit extensive organotypic heterogeneity, whereas venous SMC are similar across organs. We further identify a specific SMC subtype within the pulmonary vasculature. This comparative SMC cross-organ resource offers insight into SMC subtypes and their specific functions.


Subject(s)
Muscle, Smooth, Vascular , Transcriptome , Mice , Animals , Muscle, Smooth, Vascular/metabolism , Transcriptome/genetics , Myocytes, Smooth Muscle/metabolism , Aorta , Cells, Cultured
3.
Nat Commun ; 11(1): 3953, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32769974

ABSTRACT

Many important cell types in adult vertebrates have a mesenchymal origin, including fibroblasts and vascular mural cells. Although their biological importance is undisputed, the level of mesenchymal cell heterogeneity within and between organs, while appreciated, has not been analyzed in detail. Here, we compare single-cell transcriptional profiles of fibroblasts and vascular mural cells across four murine muscular organs: heart, skeletal muscle, intestine and bladder. We reveal gene expression signatures that demarcate fibroblasts from mural cells and provide molecular signatures for cell subtype identification. We observe striking inter- and intra-organ heterogeneity amongst the fibroblasts, primarily reflecting differences in the expression of extracellular matrix components. Fibroblast subtypes localize to discrete anatomical positions offering novel predictions about physiological function(s) and regulatory signaling circuits. Our data shed new light on the diversity of poorly defined classes of cells and provide a foundation for improved understanding of their roles in physiological and pathological processes.


Subject(s)
Cell Differentiation , Fibroblasts/physiology , Mesenchymal Stem Cells/physiology , Myocytes, Smooth Muscle/physiology , Pericytes/physiology , Animals , Cell Separation , Coronary Vessels/cytology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Flow Cytometry , Intestines/blood supply , Intestines/cytology , Male , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/cytology , Muscle, Smooth, Vascular/cytology , Myocardium/cytology , Myocytes, Smooth Muscle/cytology , Pericytes/cytology , RNA-Seq , Single-Cell Analysis , Urinary Bladder/blood supply , Urinary Bladder/cytology
4.
Sci Rep ; 8(1): 17462, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30498224

ABSTRACT

Diabetes mellitus is associated with cognitive impairment and various central nervous system pathologies such as stroke, vascular dementia, or Alzheimer's disease. The exact pathophysiology of these conditions is poorly understood. Recent reports suggest that hyperglycemia causes cerebral microcirculation pathology and blood-brain barrier (BBB) dysfunction and leakage. The majority of these reports, however, are based on methods including in vitro BBB modeling or streptozotocin-induced diabetes in rodents, opening questions regarding the translation of the in vitro findings to the in vivo situation, and possible direct effects of streptozotocin on the brain vasculature. Here we used a genetic mouse model of hyperglycemia (Ins2AKITA) to address whether prolonged systemic hyperglycemia induces BBB dysfunction and leakage. We applied a variety of methodologies to carefully evaluate BBB function and cellular integrity in vivo, including the quantification and visualization of specific tracers and evaluation of transcriptional and morphological changes in the BBB and its supporting cellular components. These experiments did neither reveal altered BBB permeability nor morphological changes of the brain vasculature in hyperglycemic mice. We conclude that prolonged hyperglycemia does not lead to BBB dysfunction, and thus the cognitive impairment observed in diabetes may have other causes.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Hyperglycemia/metabolism , Hyperglycemia/pathology , Pericytes/metabolism , Pericytes/pathology , Animals , Cell Count , Disease Management , Disease Models, Animal , Gene Expression Profiling , Hyperglycemia/genetics , Immunohistochemistry , Male , Mice , Mice, Knockout , Microglia/metabolism
5.
Sci Data ; 5: 180160, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30129931

ABSTRACT

Vascular diseases are major causes of death, yet our understanding of the cellular constituents of blood vessels, including how differences in their gene expression profiles create diversity in vascular structure and function, is limited. In this paper, we describe a single-cell RNA sequencing (scRNA-seq) dataset that defines vascular and vessel-associated cell types and subtypes in mouse brain and lung. The dataset contains 3,436 single cell transcriptomes from mouse brain, which formed 15 distinct clusters corresponding to cell (sub)types, and another 1,504 single cell transcriptomes from mouse lung, which formed 17 cell clusters. In order to allow user-friendly access to our data, we constructed a searchable database (http://betsholtzlab.org/VascularSingleCells/database.html). Our dataset constitutes a comprehensive molecular atlas of vascular and vessel-associated cell types in the mouse brain and lung, and as such provides a strong foundation for future studies of vascular development and diseases.


Subject(s)
Blood Vessels , Brain/blood supply , Lung/blood supply , Transcriptome , Animals , Blood Vessels/cytology , Blood Vessels/physiology , Databases, Factual , Endothelial Cells/physiology , Mice , Myocytes, Smooth Muscle/physiology , Pericytes/physiology , Sequence Analysis, RNA , Single-Cell Analysis
6.
Nature ; 560(7716): E3, 2018 08.
Article in English | MEDLINE | ID: mdl-29925939

ABSTRACT

In Fig. 1b of this Article, 'Csf1r' was misspelt 'Csfr1'. In addition, in Extended Data Fig. 11b, owing to an error during figure formatting, the genes listed in the first column shifted down three rows below the first gene on the list, causing a mismatch between the gene names and their characteristics. These errors have been corrected online, and the original Extended Data Fig. 11b is provided as Supplementary Information to the accompanying Amendment.

7.
Nature ; 554(7693): 475-480, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29443965

ABSTRACT

Cerebrovascular disease is the third most common cause of death in developed countries, but our understanding of the cells that compose the cerebral vasculature is limited. Here, using vascular single-cell transcriptomics, we provide molecular definitions for the principal types of blood vascular and vessel-associated cells in the adult mouse brain. We uncover the transcriptional basis of the gradual phenotypic change (zonation) along the arteriovenous axis and reveal unexpected cell type differences: a seamless continuum for endothelial cells versus a punctuated continuum for mural cells. We also provide insight into pericyte organotypicity and define a population of perivascular fibroblast-like cells that are present on all vessel types except capillaries. Our work illustrates the power of single-cell transcriptomics to decode the higher organizational principles of a tissue and may provide the initial chapter in a molecular encyclopaedia of the mammalian vasculature.


Subject(s)
Blood Vessels/cytology , Brain/blood supply , Brain/cytology , Endothelial Cells/classification , Animals , Arteries/cytology , Arterioles/cytology , Capillaries/cytology , Female , Fibroblasts/classification , Male , Mice , Myocytes, Smooth Muscle/classification , Organ Specificity , Pericytes/classification , Single-Cell Analysis , Transcriptome , Veins/cytology
8.
J Cereb Blood Flow Metab ; 38(3): 456-468, 2018 03.
Article in English | MEDLINE | ID: mdl-28276839

ABSTRACT

The establishment of a fully functional blood vascular system requires elaborate angiogenic and vascular maturation events in order to fulfill organ-specific anatomical and physiological needs. Although vascular mural cells, i.e. pericytes and vascular smooth muscle cells, are known to play fundamental roles during these processes, their characteristics during vascular development remain incompletely understood. In this report, we utilized transgenic reporter mice in which mural cells are genetically labeled to examine developing vascular mural cells in the central nervous system (CNS). We found platelet-derived growth factor receptor ß gene ( Pdgfrb)-driven EGFP reporter expression as a suitable marker for vascular mural cells at the earliest stages of mouse brain vascularization. Furthermore, the combination of Pdgfrb and NG2 gene (Cspg4) driven reporter expression increased the specificity of brain vascular mural cell labeling at later stages. The expression of other known pericyte markers revealed time-, region- and marker-specific patterns, suggesting heterogeneity in mural cell maturation. We conclude that transgenic reporter mice provide an important tool to explore the development of CNS pericytes in health and disease.


Subject(s)
Blood Vessels/ultrastructure , Brain/cytology , Brain/growth & development , Genes, Reporter/genetics , Animals , Antigens/genetics , Blood Vessels/growth & development , Brain/ultrastructure , Cerebral Cortex/growth & development , Cerebral Cortex/ultrastructure , Embryonic Development , Female , Mice , Mice, Transgenic , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/ultrastructure , Pericytes/ultrastructure , Proteoglycans/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics
9.
Sci Rep ; 6: 35108, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27725773

ABSTRACT

Pericytes, the mural cells of blood microvessels, regulate microvascular development and function and have been implicated in many brain diseases. However, due to a paucity of defining markers, pericyte identification and functional characterization remain ambiguous and data interpretation problematic. In mice carrying two transgenic reporters, Pdgfrb-eGFP and NG2-DsRed, we found that double-positive cells were vascular mural cells, while the single reporters marked additional, but non-overlapping, neuroglial cells. Double-positive cells were isolated by fluorescence-activated cell sorting (FACS) and analyzed by RNA sequencing. To reveal defining patterns of mural cell transcripts, we compared the RNA sequencing data with data from four previously published studies. The meta-analysis provided a conservative catalogue of 260 brain mural cell-enriched gene transcripts. We validated pericyte-specific expression of two novel markers, vitronectin (Vtn) and interferon-induced transmembrane protein 1 (Ifitm1), using fluorescent in situ hybridization and immunohistochemistry. We further analyzed signaling pathways and interaction networks of the pericyte-enriched genes in silico. This work provides novel insight into the molecular composition of brain mural cells. The reported gene catalogue facilitates identification of brain pericytes by providing numerous new candidate marker genes and is a rich source for new hypotheses for future studies of brain mural cell physiology and pathophysiology.


Subject(s)
Brain/cytology , Gene Expression Profiling , Microvessels/cytology , Pericytes/physiology , Animals , Flow Cytometry , Mice , Sequence Analysis, RNA , Staining and Labeling
10.
Arterioscler Thromb Vasc Biol ; 35(2): 409-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25477343

ABSTRACT

OBJECTIVE: Vascular smooth muscle cells (VSMC) are important for contraction, blood flow distribution, and regulation of blood vessel diameter, but to what extent they contribute to the integrity of blood vessels and blood-brain barrier function is less well understood. In this report, we explored the impact of the loss of VSMC in the Notch3(-/-) mouse on blood vessel integrity in the central nervous system. APPROACH AND RESULTS: Notch3(-/-) mice showed focal disruptions of the blood-brain barrier demonstrated by extravasation of tracers accompanied by fibrin deposition in the retinal vasculature. This blood-brain barrier leakage was accompanied by a regionalized and patchy loss of VSMC, with VSMC gaps predominantly in arterial resistance vessels of larger caliber. The loss of VSMC appeared to be caused by progressive degeneration of VSMC resulting in a gradual loss of VSMC marker expression and a progressive acquisition of an aberrant VSMC phenotype closer to the gaps, followed by enhanced apoptosis and cellular disintegration in the gaps. Arterial VSMC were the only mural cell type that was morphologically affected, despite Notch3 also being expressed in pericytes. Transcriptome analysis of isolated brain microvessels revealed gene expression changes in Notch3(-/-) mice consistent with loss of arterial VSMC and presumably secondary transcriptional changes were observed in endothelial genes, which may explain the compromised vascular integrity. CONCLUSIONS: We demonstrate that Notch3 is important for survival of VSMC, and reveal a critical role for Notch3 and VSMC in blood vessel integrity and blood-brain barrier function in the mammalian vasculature.


Subject(s)
Blood-Brain Barrier/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Notch/metabolism , Actins/genetics , Actins/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Blood Vessels/metabolism , Blood-Brain Barrier/pathology , Capillary Permeability , Endothelial Cells/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genotype , Male , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Pericytes/metabolism , Phenotype , Receptor, Notch3 , Receptors, Notch/deficiency , Receptors, Notch/genetics , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction , Transcription, Genetic
11.
Nat Med ; 19(12): 1617-24, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24216753

ABSTRACT

Myofibroblasts are the major source of extracellular matrix components that accumulate during tissue fibrosis, and hepatic stellate cells (HSCs) are believed to be the major source of myofibroblasts in the liver. To date, robust systems to genetically manipulate these cells have not been developed. We report that Cre under control of the promoter of Pdgfrb (Pdgfrb-Cre) inactivates loxP-flanked genes in mouse HSCs with high efficiency. We used this system to delete the gene encoding α(v) integrin subunit because various α(v)-containing integrins have been suggested as central mediators of fibrosis in multiple organs. Such depletion protected mice from carbon tetrachloride-induced hepatic fibrosis, whereas global loss of ß3, ß5 or ß6 integrins or conditional loss of ß8 integrins in HSCs did not. We also found that Pdgfrb-Cre effectively targeted myofibroblasts in multiple organs, and depletion of the α(v) integrin subunit using this system was protective in other models of organ fibrosis, including pulmonary and renal fibrosis. Pharmacological blockade of α(v)-containing integrins by a small molecule (CWHM 12) attenuated both liver and lung fibrosis, including in a therapeutic manner. These data identify a core pathway that regulates fibrosis and suggest that pharmacological targeting of all α(v) integrins may have clinical utility in the treatment of patients with a broad range of fibrotic diseases.


Subject(s)
Integrin alphaV/metabolism , Kidney Diseases/genetics , Kidney/pathology , Liver Cirrhosis/genetics , Pulmonary Fibrosis/genetics , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Female , Fibrosis/genetics , Gene Targeting , Integrin alphaV/genetics , Kidney/metabolism , Kidney Diseases/pathology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myofibroblasts/metabolism , Myofibroblasts/pathology , Pulmonary Fibrosis/pathology , Signal Transduction/physiology
12.
Nat Genet ; 45(9): 1077-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913003

ABSTRACT

Calcifications in the basal ganglia are a common incidental finding and are sometimes inherited as an autosomal dominant trait (idiopathic basal ganglia calcification (IBGC)). Recently, mutations in the PDGFRB gene coding for the platelet-derived growth factor receptor ß (PDGF-Rß) were linked to IBGC. Here we identify six families of different ancestry with nonsense and missense mutations in the gene encoding PDGF-B, the main ligand for PDGF-Rß. We also show that mice carrying hypomorphic Pdgfb alleles develop brain calcifications that show age-related expansion. The occurrence of these calcium depositions depends on the loss of endothelial PDGF-B and correlates with the degree of pericyte and blood-brain barrier deficiency. Thus, our data present a clear link between Pdgfb mutations and brain calcifications in mice, as well as between PDGFB mutations and IBGC in humans.


Subject(s)
Basal Ganglia Diseases/genetics , Basal Ganglia Diseases/pathology , Calcinosis/genetics , Mutation , Proto-Oncogene Proteins c-sis/genetics , Amino Acid Substitution , Animals , Basal Ganglia Diseases/diagnosis , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Gene Order , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Pedigree , Tomography, X-Ray Computed
13.
Dev Cell ; 23(3): 587-99, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22975327

ABSTRACT

Angiogenesis, the process by which new blood vessels arise from preexisting ones, is critical for embryonic development and is an integral part of many disease processes. Recent studies have provided detailed information on how angiogenic sprouts initiate, elongate, and branch, but less is known about how these processes cease. Here, we show that S1PR1, a receptor for the blood-borne bioactive lipid sphingosine-1-phosphate (S1P), is critical for inhibition of angiogenesis and acquisition of vascular stability. Loss of S1PR1 leads to increased endothelial cell sprouting and the formation of ectopic vessel branches. Conversely, S1PR1 signaling inhibits angiogenic sprouting and enhances cell-to-cell adhesion. This correlates with inhibition of vascular endothelial growth factor-A (VEGF-A)-induced signaling and stabilization of vascular endothelial (VE)-cadherin localization at endothelial junctions. Our data suggest that S1PR1 signaling acts as a vascular-intrinsic stabilization mechanism, protecting developing blood vessels against aberrant angiogenic responses.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Neovascularization, Physiologic , Receptors, Lysosphingolipid/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Lysosphingolipid/deficiency , Sphingosine-1-Phosphate Receptors , Zebrafish
14.
Proc Natl Acad Sci U S A ; 108(51): 20479-84, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22143766

ABSTRACT

Lipoxygenases (LO) are a class of dioxygenases, which form hydroperoxy, hydroxy, and epoxy derivatives of arachidonic acid with distinct positional and stereochemical configurations. In man, there are two known types of 12-LO that are distinguished by their expression patterns and catalytic properties. The platelet 12S-LO plays a role in platelet aggregation and 12R-LO seems to be important for normal skin function. Using BLAST searches of the zebrafish (zf) genome we identified one candidate zf12-LO gene with 43% identity with human 12R-LO at the mRNA level and the deduced primary sequence carried the so called "Coffa" structural determinant (Gly residue) for R stereoselectivity of LOs. However, incubations of recombinant, purified, zf12-LO with arachidonic acid revealed exclusive formation of 12(S)-hydroperoxy-eicosatetraenoic acid. Further studies with immunohistochemistry showed prominent expression of zf12-LO in the cell nuclei of skin epithelium, the epithelial lining of the stomodeum, and the pharyngeal pouches in zf embryos. To probe its function, zf12-LO was subjected to targeted knock-down in zf embryos, resulting in the development of a severe phenotype, characterized by abnormal development of the brain, the eyes, and the tail as well as pericardial and yolk sac edema. Hence, we have identified a unique vertebrate 12S-LO that breaks the current structure-function paradigms for S and R stereo-specificity and with critical roles in normal embryonic development.


Subject(s)
Arachidonate 12-Lipoxygenase/chemistry , Gene Expression Regulation, Developmental , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonic Acid/chemistry , Blood Platelets/metabolism , Chromatography, High Pressure Liquid/methods , Chromatography, Liquid/methods , Cloning, Molecular , Developmental Biology/methods , Gene Expression Profiling , Immunohistochemistry/methods , Mass Spectrometry/methods , Microscopy, Fluorescence/methods , Phenotype , Stereoisomerism , Zebrafish
15.
J Am Soc Nephrol ; 22(11): 2037-46, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21949092

ABSTRACT

Unbiased transcriptome profiling and functional genomics approaches identified glucocorticoid-induced transcript 1 (GLCCI1) as being a transcript highly specific for the glomerulus, but its role in glomerular development and disease is unknown. Here, we report that mouse glomeruli express far greater amounts of Glcci1 protein compared with the rest of the kidney. RT-PCR and Western blotting demonstrated that mouse glomerular Glcci1 is approximately 60 kD and localizes to the cytoplasm of podocytes in mature glomeruli. In the fetal kidney, intense Glcci1 expression occurs at the capillary-loop stage of glomerular development. Using gene knockdown in zebrafish with morpholinos, morphants lacking Glcci1 function had collapsed glomeruli with foot-process effacement. Permeability studies of the glomerular filtration barrier in these zebrafish morphants demonstrated a disruption of the selective glomerular permeability filter. Taken together, these data suggest that Glcci1 promotes the normal development and maintenance of podocyte structure and function.


Subject(s)
Kidney Glomerulus/physiopathology , Podocytes/physiology , Pronephros/physiopathology , Proteinuria/metabolism , Proteinuria/physiopathology , Receptors, Glucocorticoid/deficiency , Animals , Cytoplasm/metabolism , Dexamethasone/pharmacology , Disease Models, Animal , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Glucocorticoids/pharmacology , Kidney Glomerulus/abnormalities , Male , Mice , Mice, Inbred ICR , Oligonucleotides, Antisense/pharmacology , Pronephros/abnormalities , Proteinuria/pathology , Rabbits , Receptors, Glucocorticoid/genetics , Zebrafish , Zebrafish Proteins
16.
Dev Biol ; 313(1): 455-64, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18062954

ABSTRACT

The coxsackie and adenovirus receptor (CAR) is a member of the immunoglobulin superfamily and a component of vertebrate tight junctions. CAR protein is widely expressed in fish and mammals in organs of epithelial origin suggesting possible functions in epithelial biology. In order to gain insight into its function, we knocked the CAR gene down in zebrafish using antisense morpholinos. We identified a requirement for CAR in the terminal differentiation of glomerular podocytes and pronephric tubular epithelia. Podocytes differentiate in CAR morphants but are not able to elaborate a regularly patterned architecture of foot processes. In the tubules, CAR was required for the apposition of plasma membranes from adjacent epithelial cells but did not appear to be necessary for the formation of tight junctions. Additionally, tubular epithelia lacking CAR were not able to elaborate apical brush border microvilli. These results establish a requirement for CAR in the terminal differentiation of renal glomerular and tubular cell types.


Subject(s)
Epithelial Cells/cytology , Kidney Glomerulus/embryology , Kidney Tubules/embryology , Receptors, Virus/metabolism , Zebrafish Proteins/metabolism , Animals , Cell Differentiation , Kidney Glomerulus/cytology , Kidney Tubules/cytology , Receptors, Virus/genetics , Zebrafish , Zebrafish Proteins/genetics
17.
Exp Cell Res ; 312(9): 1566-80, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16542650

ABSTRACT

The coxsackie- and adenovirus receptor (CAR) is a transmembrane protein belonging to the immunoglobulin superfamily. The function of CAR as a virus receptor has been extensively analyzed, while its physiological role and expression pattern in adult tissues have remained less clear. CAR associates with epithelial tight junctions in vitro and mediates cell-cell adhesion. Using a set of affinity-purified antibodies, we show that CAR is predominantly expressed in epithelial cells lining the body cavities in adult mice, where it specifically co-localizes with the tight junction components ZO-1 and occludin. Notably, CAR could not be detected in endothelial cells of the vasculature, including brain capillaries. CAR expression correlated positively with the maturity of tight junctions and inversely with permeability. With a few exceptions, the two known CAR isoforms were co-expressed in most epithelial cells analyzed. A CAR mutant lacking the intracellular tail over-expressed in transgenic mice was diffusely localized over the plasma membrane, showing the importance of this domain for correct subcellular localization in vivo. We conclude that CAR is localized to epithelial tight junctions in vivo where it may play a role in the regulation of epithelial permeability and tissue homeostasis.


Subject(s)
Epithelial Cells/chemistry , Homeostasis/physiology , Receptors, Virus/analysis , Tight Junctions/chemistry , Animals , Cell Line , Cell Membrane Permeability/physiology , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Epithelial Cells/cytology , Epithelial Cells/physiology , Fluorescent Antibody Technique , Gastrointestinal Tract/chemistry , Gastrointestinal Tract/cytology , Humans , Kidney/chemistry , Kidney/cytology , Liver/chemistry , Liver/cytology , Male , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Immunoelectron , Occludin , Phosphoproteins/analysis , Prostate/chemistry , Prostate/cytology , Receptors, Virus/genetics , Receptors, Virus/physiology , Respiratory System/chemistry , Respiratory System/cytology , Tight Junctions/physiology , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein
18.
Exp Cell Res ; 309(1): 110-20, 2005 Sep 10.
Article in English | MEDLINE | ID: mdl-15979067

ABSTRACT

The coxsackievirus and adenovirus receptor (CAR) is a cell surface protein that is proposed to be involved in cell-cell adhesion. Based on a yeast two-hybrid screen, co-immunoprecipitation and binding experiments, the intracellular tail of CAR was found to interact both in vivo and in vitro with the Ligand-of-Numb Protein-X2 (LNX2). The interacting domains between the two proteins were identified by truncation analyses and affinity chromatography. CAR and LNX2 protein expression in embryonic mouse tissues was analyzed by immunohistochemistry. The results suggest that CAR is a partner in a protein complex organized at specific subcellular sites by LNX2.


Subject(s)
Carrier Proteins/metabolism , Receptors, Virus/metabolism , Adenoviridae/physiology , Amino Acid Sequence , Animals , Binding Sites , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Embryo, Mammalian/metabolism , Enterovirus/physiology , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Tight Junctions/metabolism
19.
J Biol Chem ; 279(1): 796-804, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14573622

ABSTRACT

The CTX family is a growing group of type I transmembrane proteins within the immunoglobulin superfamily (IgSF). They localize to junctional complexes between endothelial and epithelial cells and seem to participate in cell-cell adhesion and transmigration of leukocytes. Here, we report the identification of a new member of the CTX family. This protein, which was designated CLMP (coxsackie- and adenovirus receptor-like membrane protein), is composed of 373 amino acids including an extracellular part containing a V- and a C2-type domain, a transmembrane region and a cytoplasmic tail. CLMP mRNA was detected in a variety of both human and mouse tissues and cell lines. The protein migrated with an Mr of around 48 on SDS-PAGE and was predominantly expressed in epithelial cells within different tissues. In cultured epithelial cells, CLMP was detected in areas of cell-cell contacts. When exogenously expressed in polarized MDCK cells, CLMP was restricted to the subapical area of the lateral cell surface, where it co-localized with the tight junction markers ZO-1 and occludin. Also endogenous CLMP showed association with tight junctions, as analyzed in polarized human CACO-2 cells. This suggested a role for CLMP in cell-cell adhesion and indeed, overexpressed CLMP induced aggregation of non-polarized CHO cells. Furthermore, CLMP-expressing MDCK cells showed significantly increased transepithelial resistance, indicating a role for CLMP in junctional barrier function. Thus, we conclude that CLMP is a novel cell-cell adhesion molecule and a new component of epithelial tight junctions. We also suggest, based on phylogenetic studies, that CLMP, CAR, ESAM, and BT-IgSF form a new group of proteins within the CTX family.


Subject(s)
Epithelial Cells/physiology , Membrane Proteins/physiology , Tight Junctions/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cloning, Molecular , Colonic Neoplasms , Conserved Sequence , Coxsackie and Adenovirus Receptor-Like Membrane Protein , DNA Primers , Databases, Nucleic Acid , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Phylogeny , Receptors, Virus , Recombinant Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Tumor Cells, Cultured
20.
J Biol Chem ; 278(9): 7439-44, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12468544

ABSTRACT

The Coxsackievirus and adenovirus receptor (CAR) functions as a virus receptor, but its primary biological function is unknown. A yeast two-hybrid screen was used to identify Ligand-of-Numb protein-X (LNX) as a binding partner to the intracellular tail of CAR. LNX harbors several protein-protein interacting domains, including four PDZ domains, and was previously shown to bind to and regulate the expression level of the cell-fate determinant Numb. CAR was able to bind LNX both in vivo and in vitro. Efficient binding to LNX required not only the consensus PDZ domain binding motif in the C terminus of CAR but also upstream sequences. The CAR binding region in LNX was mapped to the second PDZ domain. CAR and LNX were also shown to colocalize in vivo in mammalian cells. We speculate that CAR and LNX are part of a larger protein complex that might have important functions at discrete subcellular localizations in the cell.


Subject(s)
Carrier Proteins/chemistry , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Ubiquitin-Protein Ligases , Animals , Binding Sites , Blotting, Western , Carrier Proteins/metabolism , Cell Division , Cell Line , Coxsackie and Adenovirus Receptor-Like Membrane Protein , DNA, Complementary/metabolism , Fluorescent Antibody Technique, Indirect , Fungal Proteins/metabolism , Gene Library , Glutathione Transferase/metabolism , Humans , Mice , Plasmids/metabolism , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...