Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Tissue Eng Regen Med ; 11(1): 109-120, 2017 01.
Article in English | MEDLINE | ID: mdl-24737702

ABSTRACT

Previously we demonstrated the superiority of small islets vs large islets in terms of function and survival after transplantation, and we generated reaggregated rat islets (pseudo-islets) of standardized small dimensions by the hanging-drop culture method (HDCM). The aim of this study was to generate human pseudo-islets by HDCM and to evaluate and compare the physiological properties of rat and human pseudo-islets. Isolated rat and human islets were dissociated into single cells and incubated for 6-14 days by HDCM. Newly formed pseudo-islets were analysed for dimensions, morphology, glucose-stimulated insulin secretion (GSIS) and total insulin content. The morphology of reaggregated human islets was similar to that of native islets, while rat pseudo-islets had a reduced content of α and δ cells. GSIS of small rat and human pseudo-islets (250 cells) was increased up to 4.0-fold (p < 0.01) and 2.5-fold (p < 0.001), respectively, when compared to their native counterparts. Human pseudo-islets showed a more pronounced first-phase insulin secretion as compared to intact islets. GSIS was inversely correlated to islet size, and small islets (250 cells) contained up to six-fold more insulin/cell than large islets (1500 cells). Tissue loss with this new technology could be reduced to 49.2 ± 1.5% in rat islets, as compared to the starting amount. With HDCM, pseudo-islets of standardized size with similar cellular composition and improved biological function can be generated, which compensates for tissue loss during production. Transplantation of small pseudo-islets may represent an attractive strategy to improve graft survival and function, due to better oxygen and nutrient supply during the phase of revascularization. Copyright © 2014 John Wiley & Sons, Ltd.


Subject(s)
Cell Culture Techniques/methods , Insulin/chemistry , Islets of Langerhans/cytology , Animals , Cell Aggregation , Cells, Cultured , Glucose/chemistry , Graft Survival , Gravitation , Humans , Insulin-Secreting Cells/cytology , Male , Oxygen/chemistry , Perfusion , Rats , Rats, Inbred Lew
2.
Diabetologia ; 51(1): 110-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17973096

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to compare the long-term outcomes--in terms of glucose control, renal function and procedure-related complications--of simultaneous islet-kidney (SIK) transplantation with those of simultaneous pancreas-kidney (SPK) transplantation in patients with type 1 diabetes mellitus. METHODS: HbA1c, need for insulin, GFR and complication rate were compared between 13 recipients of SIK and 25 recipients of SPK transplants at the same institution. The mean follow-up was 41 months. RESULTS: Two primary organ non-functions occurred in the SIK group. HbA1c did not differ at any time point during follow-up in the SIK group compared with the SPK group (mean during follow-up 6.3 vs 5.9%). Similarly, kidney function over time was not different between the two groups. A higher rate of insulin independence following SPK transplantation (after 1 year 96 vs 31% in the SIK group) was counterbalanced by a higher rate of serious adverse events (40% relaparotomies vs 0% in the SIK group). CONCLUSIONS/INTERPRETATION: The endogenous insulin production achieved by islet transplantation, combined with optimal insulin therapy, was sufficient for maintaining near-normal glucose levels. In terms of glucose control, islet transplantation provides results comparable to those achieved with pancreas transplantation. However, SPK results in a higher rate of insulin independence, albeit at the cost of more surgical complications. These results have led to a new paradigm in islet transplantation at our institution, where the primary goal is not insulin independence, but good glucose control and avoidance of severe hypoglycaemia.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/methods , Islets of Langerhans/metabolism , Kidney Transplantation/methods , Pancreas Transplantation/methods , Adult , Female , Follow-Up Studies , Glomerular Filtration Rate , Glycated Hemoglobin/metabolism , Humans , Insulin/metabolism , Insulin/therapeutic use , Insulin Secretion , Male , Middle Aged , Treatment Outcome
3.
Transplant Proc ; 39(6): 2018-20, 2007.
Article in English | MEDLINE | ID: mdl-17692680

ABSTRACT

INTRODUCTION: Rejection and hypoxia are the main factors that limit islet engraftment in the recipient liver in the immediate posttransplant period. Recently authors have reported a negative relationship of graft function and islet size, concluding that small islets are superior to large islets. Islets can be dissociated into single cells and reaggregated into so called "pseudoislets," which are functionally equivalent to intact islets but exhibit reduced immunogenicity. The aim of our study was develop a technique that enabled one to obtain pseudoislets of defined, preferably small, dimensions. MATERIALS AND METHODS: Islets were harvested from Lewis rats by the collagenase digestion procedure. After purification, the isolated islets were dissociated into single cells by trypsin digestion. Fractions with different cell numbers were seeded into single drops onto cell culture dishes, which were inverted and incubated for 5 to 8 days under cell culture conditions. Newly formed pseudoislets were analyzed for dimension, morphology, and cellular composition. RESULTS: The volume of reaggregated pseudoislets strongly correlated with the cell number (r(2) = .995). The average diameter of a 250-cell aggregate was 95 +/- 8 microm (mean +/- SD) compared with 122 +/- 46 microm of freshly isolated islets. Islet cell loss may be minimized by performing reaggregation in the presence of medium glucose (11 mmol/L) and the GLP-1 analogue Exendin-4. Morphology, cellular composition, and architecture of reaggregated islets were comparable to intact islets. CONCLUSION: The "hanging drop" culture method allowed us to obtain pseudoislets of standardized size and regular shape, which did not differ from intact islets in terms of cellular composition or architecture. Further investigations are required to minimize cell loss and test in vivo function of transplanted pseudoislets.


Subject(s)
Islets of Langerhans/anatomy & histology , Animals , Cell Aggregation , Glucagon/analysis , Graft Rejection/immunology , Immunohistochemistry , Insulin/analysis , Islets of Langerhans/cytology , Islets of Langerhans Transplantation/immunology , Rats , Somatostatin/analysis
4.
Exp Cell Res ; 303(1): 68-78, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15572028

ABSTRACT

Studies in vivo indicate that IRS2 plays an important role in maintaining functional beta-cell mass. To investigate if IRS2 autonomously affects beta-cells, we have studied proliferation, apoptosis, and beta-cell function in isolated rat and human islets after overexpression of IRS2 or IRS1. We found that beta-cell proliferation was significantly increased in rat islets overexpressing IRS2 while IRS1 was less effective. Moreover, proliferation of a beta-cell line, INS-1, was decreased after repression of Irs2 expression using RNA oligonucleotides. Overexpression of IRS2 in human islets significantly decreased apoptosis of beta-cells, induced by 33.3 mM D-glucose. However, IRS2 did not protect cultured rat islets against apoptosis in the presence of 0.5 mM palmitic acid. Overexpression of IRS2 in isolated rat islets significantly increased basal and D-glucose-stimulated insulin secretion as determined in perifusion experiments. Therefore, IRS2 is sufficient to induce proliferation in rat islets and to protect human beta-cells from D-glucose-induced apoptosis. In addition, IRS2 can improve beta-cell function. Our results indicate that IRS2 acts autonomously in beta-cells in maintenance and expansion of functional beta-cell mass in vivo.


Subject(s)
Apoptosis/drug effects , Cell Proliferation , Hyperglycemia/metabolism , Islets of Langerhans/metabolism , Phosphoproteins/metabolism , Animals , Cells, Cultured , Cloning, Molecular , Glucose/pharmacology , Humans , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Palmitic Acid/pharmacology , Phosphoproteins/genetics , Rats
5.
Eur J Neurosci ; 13(6): 1113-24, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11285009

ABSTRACT

Dystrophin and its autosomal homologue utrophin are coexpressed in muscle cells, and utrophin is functionally able to replace dystrophin in models of Duchenne muscular dystrophy. In brain, the two proteins are expressed differentially, suggesting distinct functional roles. Dystrophin is associated with postsynaptic GABA(A) receptors in hippocampus, cortex and cerebellum, whereas utrophin is present extrasynaptically, notably in large brainstem neurons. Here, the regulation of dystrophin and utrophin was investigated in a model of temporal lobe epilepsy. Adult mice were injected unilaterally with kainic acid into the dorsal hippocampus to induce loss of pyramidal cells and hypertrophy of dentate gyrus (DG) granule cells, as described (Suzuki, F., Junier, M.P., Guilhem, D., Sorensen, J.C. & Onteniente, B. (1995) Neuroscience, 64, 665--674.). These morphological changes were associated with an increase in postsynaptic GABA(A)-receptors in the ipsilateral DG, as demonstrated by a parallel increase in punctate immunoreactivity to GABA(A)-receptor alpha 2 subunit, gephyrin and dystrophin in the molecular layer. Thus, both dystrophin and gephyrin were involved in postsynaptic clustering of GABA(A) receptors. A transient induction of utrophin was seen at the onset of degeneration in CA1 and CA3 pyramidal cells and in the hilus. Most strikingly, however, utrophin immunoreactivity appeared in the granule cell layer of the DG and became very strong in hypertrophic granule cells 1--2 months post-kainate treatment. These results suggest that utrophin provides structural support of neuronal membranes, whereas dystrophin is a component of GABAergic synapses.


Subject(s)
Cytoskeletal Proteins/metabolism , Dystrophin/metabolism , Epilepsy, Temporal Lobe/physiopathology , Membrane Proteins/metabolism , Neuronal Plasticity/physiology , gamma-Aminobutyric Acid/physiology , Animals , Carrier Proteins/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Hippocampus/pathology , Male , Mice , Neurons/metabolism , Protein Isoforms/metabolism , Receptors, GABA-A/metabolism , Staining and Labeling , Up-Regulation , Utrophin
6.
J Comp Neurol ; 422(4): 594-611, 2000 Jul 10.
Article in English | MEDLINE | ID: mdl-10861528

ABSTRACT

The cellular distribution of utrophin, the autosomal homologue of dystrophin, was investigated in developing and adult rat and mouse brain by in situ hybridization and immunohistochemistry. Digoxigenin-labeled cRNA probes complementary to N-terminal, rod-domain, and C-terminal encoding sequences of utrophin were used to differentiate between full-length and short C-terminal isoforms. Largely overlapping distribution patterns were seen for the three probes in neurons of cerebral cortex, accessory olfactory bulb, and several sensory and motor brainstem nuclei as well as in blood vessels, pia mater, and choroid plexus. The C-terminal probe was detected in addition in the main olfactory bulb, striatum, thalamic reticular nucleus, and hypothalamus, suggesting a selective expression of G-utrophin in these neurons. Western blot analysis with isoform-specific antisera confirmed the expression of both full-length and G-utrophin in brain. Immunohistochemically, only full-length utrophin was detected in neurons, in close association with the plasma membrane. In addition, intense staining was seen in blood vessels, meninges, and choroid plexus, selectively localized in the basolateral membrane of immunopositive epithelial cells. The expression pattern of utrophin was already established at early postnatal stages and did not change thereafter. Double-labeling analysis revealed that utrophin and dystrophin are differentially expressed on the cellular and subcellular levels in juvenile and adult brain. Likewise, in mice lacking full-length dystrophin isoforms (mdx mice), no change in utrophin expression and distribution could be detected in brain, although utrophin was markedly up-regulated in muscle cells. These results suggest that utrophin and dystrophin are independently regulated and have distinct functional roles in CNS neurons.


Subject(s)
Brain/metabolism , Cytoskeletal Proteins/metabolism , Dystrophin/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Age Factors , Animals , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Utrophin
7.
J Cell Biochem ; 77(3): 418-31, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10760950

ABSTRACT

Dystrophin and utrophin are known to link the intracellular cytoskeleton to the extracellular matrix via a transmembraneous glycoprotein complex. Four short C-terminal isoforms (Dp71, Dp116, Dp140, and Dp260) are described for dystrophin and three for utrophin (Up71, Up113, and Up140). We describe here for the first time the existence of a 3.7-kb transcript and a 62-kDa protein in C6 glioma cells representing a short N-terminal isoform unique for utrophin (N-utrophin). More than 20 clones covering the entire coding region of utrophin were isolated from a rat C6 glioma cell cDNA library. Two clones were found to code for a protein with 539 amino acids. Its sequence is identical to that of the full-length utrophin, except for the last residue where Cys is replaced by Val. This isoform contains the actin binding domain (consisting of two calponin homology subdomains), followed by two spectrin-like repeats. A recombinant fragment corresponding to N-utrophin binds to F-actin in vitro with an equilibrium constant (affinity) K of 4.5 x 10(5) M(-1) and a stoichiometry of one fragment per around five actin monomers. Immunocytochemical staining of C6 glioma cells with antisera specific for different utrophin regions localised full-length utrophin in the submembraneous cortical actin layer as revealed by confocal microscopy. A distinct staining pattern for the N-utrophin was not detectable, although it was expected to localise at the actin stress fibers. It is assumed that it co-localises via the two spectrin-like repeats with the full-length utrophin at the cell membrane.


Subject(s)
Cytoskeletal Proteins/chemistry , Membrane Proteins/chemistry , Actins/metabolism , Animals , Blotting, Northern , Blotting, Western , Cloning, Molecular , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , DNA, Complementary/metabolism , Diaphragm/metabolism , Dystrophin/metabolism , Gene Library , Immunohistochemistry , Kidney/metabolism , Lung/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Microscopy, Confocal , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Rats , Recombinant Proteins/metabolism , Tumor Cells, Cultured , Utrophin
8.
Eur J Neurosci ; 11(12): 4457-62, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10594673

ABSTRACT

Dystrophin is selectively localized in the postsynaptic density of neurons in cerebral cortex, hippocampus and cerebellum. Here, we show by double-immunofluorescence staining that dystrophin is extensively colocalized with GABAA receptor subunit clusters in these brain regions. To determine the relevance of this observation, we investigated in mdx mice, which provide a model of Duchenne muscular dystrophy, whether the absence of dystrophin affects the synaptic clustering of GABAA receptors. A marked reduction in the number of clusters immunoreactive for the alpha1 and alpha2 subunits was observed in, respectively, cerebellum and hippocampus of mdx mice, but not in striatum, which is normally devoid of dystrophin. Furthermore, these alterations were not accompanied by a change in gephyrin staining, although gephyrin is colocalized with the majority of GABAA receptor clusters in these regions. These results indicate that dystrophin may play an important role in the clustering or stabilization of GABAA receptors in a subset of central inhibitory synapses. These deficits may underlie the cognitive impairment seen in Duchenne patients.


Subject(s)
Dystrophin/deficiency , Muscular Dystrophy, Animal/metabolism , Receptors, GABA-A/metabolism , Synapses/metabolism , Animals , Carrier Proteins/metabolism , Cerebellum/metabolism , Corpus Striatum/metabolism , Dystrophin/metabolism , Hippocampus/metabolism , Immunohistochemistry , Macromolecular Substances , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Neocortex/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution
9.
Mol Cell Neurosci ; 10(5/6): 229-42, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9618215

ABSTRACT

Utrophin is normally present exclusively in synaptic regions of skeletal muscle fibers, although it is expressed extrasynaptically in certain pathological situations, where it has been proposed to compensate for the absence of dystrophin in Duchenne muscular dystrophy patients and mdx mice. Recently there have been conflicting reports regarding the preferential expression of utrophin mRNA at the neuromuscular junction. Using in situ hybridization with RNA probes, we show a clear accumulation of autoradiographic labeling at more than 90% of neuromuscular junctions (identified by histochemical demonstration of cholinesterase activity). The intensity of this labeling is proportional to the number of junctional myonuclei in the section. Some clusters of labeling were found associated with nonmuscle nuclei (e.g., blood vessels, nerves), where utrophin is present. In addition, labeling for utrophin mRNA was associated with about 25% of extrajunctional myonuclei, where the protein is not present. The mean labeling per nucleus at junctional myonuclei was at least 10 times greater than at extrajunctional myonuclei. We discuss the possible regulatory mechanisms involved in the heterogeneous expression of utrophin mRNA in skeletal muscle. Copyright 1998 Academic Press.

10.
Cardiovasc Res ; 37(2): 381-404, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9614495

ABSTRACT

Cardiac hypertrophy is an adaptive response that normalizes wall stress and compensates for increased workload. It is accompanied by distinct qualitative and quantitative changes in the expression of protein isoforms concerning contractility, intracellular Ca(2+)-homeostasis and metabolism. Changes in the myosin subunit isoform expression improves contractility by an increase in force generation at a given Ca(2+)-concentration (increased Ca(2+)-sensitivity) and by improving the economy of the chemo-mechanical transduction process per amount of utilised ATP (increased duty ratio). In the human atrium this is achieved by partial replacement of the endogenous fast myosin by the ventricular slow-type heavy and light chains. In the hypertrophic human ventricle the slow-type beta-myosin heavy chains remain unchanged, but the ectopic expression of the atrial myosin essential light chain (ALC1) partially replaces the endogenous ventricular isoform (VLC1). The ventricular contractile apparatus with myosin containing ALC1 is characterised by faster cross-bridge kinetics, a higher Ca(2+)-sensitivity of force generation and an increased duty ratio. The mechanism for cross-bridge modulation relies on the extended Ala-Pro-rich N-terminus of the essential light chains of which the first eleven residues interact with the C-terminus of actin. A change in charge in this region between ALC1 and VLC1 explains their functional difference. The intracellular Ca(2+)-handling may be impaired in heart failure, resulting in either higher or lower cytosolic Ca(2+)-levels. Thus the state of the cardiomyocyte determines whether this hypertrophic adaptation remains beneficial or becomes detrimental during failure. Also discussed are the effects on contractility of long-term changes in isoform expression of other sarcomeric proteins. Positive and negative modulation of contractility by short-term phosphorylation reactions at multiple sites in the myosin regulatory light chain, troponin-I, troponin-T, alpha-tropomyosin and myosin binding protein-C are considered in detail.


Subject(s)
Calcium/metabolism , Cardiomegaly/metabolism , Myocardial Contraction , Myosin Light Chains/metabolism , Animals , Humans , Rats , Sarcomeres/metabolism
11.
Mol Cell Neurosci ; 10(5-6): 229-42, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9604203

ABSTRACT

Utrophin is normally present exclusively in synaptic regions of skeletal muscle fibers, although it is expressed extrasynaptically in certain pathological situations, where it has been proposed to compensate for the absence of dystrophin in Duchenne muscular dystrophy patients and mdx mice. Recently there have been conflicting reports regarding the preferential expression of utrophin mRNA at the neuromuscular junction. Using in situ hybridization with RNA probes, we show a clear accumulation of autoradiographic labeling at more than 90% of neuromuscular junctions (identified by histochemical demonstration of cholinesterase activity). The intensity of this labeling is proportional to the number of junctional myonuclei in the section. Some clusters of labeling were found associated with nonmuscle nuclei (e.g., blood vessels, nerves), where utrophin is present. In addition, labeling for utrophin mRNA was associated with about 25% of extrajunctional myonuclei, where the protein is not present. The mean labeling per nucleus at junctional myonuclei was at least 10 times greater than at extrajunctional myonuclei. We discuss the possible regulatory mechanisms involved in the heterogeneous expression of utrophin mRNA in skeletal muscle.


Subject(s)
Cytoskeletal Proteins/genetics , Membrane Proteins/genetics , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , RNA, Messenger/biosynthesis , Amino Acid Sequence , Animals , Autoradiography , Cell Nucleus/metabolism , Female , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , RNA, Messenger/analysis , Rats , Rats, Wistar , Utrophin
12.
Proc Natl Acad Sci U S A ; 92(9): 3697-701, 1995 Apr 25.
Article in English | MEDLINE | ID: mdl-7731967

ABSTRACT

The utrophin gene is closely related to the dystrophin gene in both sequence and genomic structure. The Duchenne muscular dystrophy (DMD) locus encodes three 14-kb dystrophin transcripts in addition to several smaller isoforms, one of which, Dp116, is specific to peripheral nerve. We describe here the corresponding 5.5-kb mRNA from the utrophin locus. This transcript, designated G-utrophin, is of particular interest because it is specifically expressed in the adult mouse brain and appears to be the predominant utrophin transcript in this tissue. G-utrophin is expressed in brain sites generally different from the regions expressing beta-dystroglycan. During mouse embryogenesis G-utrophin is also seen in the developing sensory ganglia. Our data confirm the close evolutionary relationships between the DMD and utrophin loci; however, the functions for the corresponding proteins probably differ.


Subject(s)
Brain/metabolism , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Dystrophin/genetics , Ganglia, Sensory/metabolism , Gene Expression , Membrane Proteins , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Consensus Sequence , DNA, Complementary , In Situ Hybridization , Mice , Molecular Sequence Data , Muscular Dystrophy, Animal/genetics , Open Reading Frames , Organ Specificity , RNA, Messenger/biosynthesis , Sequence Homology, Amino Acid , Transcription, Genetic , Utrophin
13.
Eur J Biochem ; 227(3): 617-28, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7867620

ABSTRACT

We have isolated and characterised the gene encoding the chicken axonal cell adhesion molecule axonin-1. This gene comprises 23 exons distributed over approximately 40 kb. Each of the six immunoglobulin-like domains and the four fibronectin-type-III-like domains of axonin-1 is encoded by two exons. The introns between two domains are exclusively phase I. Their exon/intron borders correspond to the domain borders of the protein, suggesting that the gene of axonin-1 had been generated by exon shuffling. Three transcripts with a length of 4.3 kb, 5 kb, and 8 kb are found, and we provide evidence that they result from alternative use of polyadenylation signals. In situ hybridization revealed co-localisation of these transcripts in time and space in the developing chicken retina. Several identical transcription initiation sites were found in retina, brain, and cerebellum by RNase protection assay and anchored polymerase chain reaction. By transfection of HeLa cells, rat PC-12 phaeochromocytoma cells, and chicken embryonic fibroblasts with serially truncated segments of the 5'-flanking region linked to a luciferase reporter gene, we have found that the sequence from -91 to +56 relative to the transcription initiation site is sufficient to promote efficient gene expression. Tissue-specific expression of the axonin-1 gene seems to be regulated in part by sequences more than 1 kb upstream of the transcription initiation site. As revealed by computer analysis, the sequence immediately upstream of exon 1 contains an AP-2 binding site, a tumor phorbol-ester-responsive element, and a homeodomain protein binding site, but no canonical TATA box. A second AP-2 binding site and a homeodomain protein binding site are located within exon 1.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Chickens/genetics , Promoter Regions, Genetic , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Chick Embryo , Chromosome Mapping , Cloning, Molecular , Contactin 2 , DNA Primers/genetics , Exons , Gene Expression , Genes, Reporter , HeLa Cells , Humans , Introns , Molecular Sequence Data , RNA, Messenger/genetics , Rats , Transcription, Genetic , Transfection
14.
Proc Natl Acad Sci U S A ; 91(18): 8307-13, 1994 Aug 30.
Article in English | MEDLINE | ID: mdl-8078878

ABSTRACT

Duchenne muscular dystrophy is a severe X chromosome-linked, muscle-wasting disease caused by lack of the protein dystrophin. The exact function of dystrophin remains to be determined. However, analysis of its interaction with a large oligomeric protein complex at the sarcolemma and the identification of a structurally related protein, utrophin, is leading to the characterization of candidate genes for other neuromuscular disorders.


Subject(s)
Cytoskeletal Proteins/physiology , Dystrophin/physiology , Membrane Glycoproteins/physiology , Membrane Proteins/physiology , Muscle Proteins/physiology , Muscles/physiology , Animals , Calcium-Binding Proteins , Chromosome Mapping , Dystroglycans , Genes , Humans , Macromolecular Substances , Neuromuscular Junction/physiology , Utrophin
15.
Eur J Biochem ; 211(1-2): 329-39, 1993 Jan 15.
Article in English | MEDLINE | ID: mdl-8425542

ABSTRACT

Axonal surface glycoproteins, composed of repeated immunoglobulin-like and fibronectin-type-III(FNIII)-like domains, mediate adhesion between axons or between axons and non-neuronal cells or extracellular matrix proteins. Several representatives of this group promote neurite outgrowth, when presented as substratum to neurons in culture, and have been implicated in axonal guidance mechanisms. TAG-1 and axonin-1 are presumptive species homologues of the rat and the chick, respectively; together with F11/F3, they form a subgroup of Ig/FNIII-like molecules containing a glycosyl-PtdIns membrane anchor. Recent reports on tumor suppressor genes encoding Ig-like and FNIII-like sequences prompted us to isolate the human homologue to TAG-1 and axonin-1. Polymerase chain reaction (PCR) primers were designed to regions conserved in both TAG-1 and axonin-1 using deoxyinosine at ambiguous positions. An expected 1000-bp fragment was obtained from cDNA derived from adult human cerebellum. Using this PCR fragment as a probe, several clones were isolated from a human fetal brain cDNA library. Nucleotide sequence analysis of a full-length clone, as expected, revealed a high degree of similarity to rat TAG-1 (91% identity) and chicken axonin-1 (75% identity) at the amino acid level. The encoded protein was then transiently expressed in monkey COS1 cells, and a stable mouse myeloma cell line was established expressing human TAG-1/axonin-1. The transfected COS1 and myeloma cells showed immunoreactivity on the cell surface with polyclonal anti-(chicken axonin-1) serum. On Western blots, the same antibodies recognized the recombinant protein migrating slightly slower on SDS/PAGE than chicken axonin-1. A comparison of chicken and human brain-tissue proteins by Western-blot analysis revealed a similar apparent molecular mass difference between the two species, which might be due to three additional N-glycosylation sites present on human TAG-1/axonin-1. Immunostaining of cryostat sections of embryonic retinas with polyclonal anti-(axonin-1) serum showed similar expression patterns in chicken and human samples at corresponding developmental stages. An additional shared feature of human TAG-1/axonin-1, rat TAG-1 and chick axonin-1 is their attachment to the cell membrane with a glycosyl-PtdIns anchor.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Membrane Glycoproteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Chick Embryo , Chickens , Chlorocebus aethiops , Cloning, Molecular , Contactin 2 , DNA/genetics , Growth Substances/genetics , Humans , In Vitro Techniques , Molecular Sequence Data , Neurites/ultrastructure , Oligodeoxyribonucleotides/chemistry , Polymerase Chain Reaction , Recombinant Proteins , Retina/metabolism , Sequence Alignment
16.
Eur J Biochem ; 204(2): 453-63, 1992 Mar 01.
Article in English | MEDLINE | ID: mdl-1311675

ABSTRACT

Axonin-1 is an axon-associated cell adhesion molecule (AxCAM) of the chicken, which promotes neurite outgrowth by interaction with the AxCAM L1(G4) of the neuritic membrane. Here we report the cloning and sequence determination of a cDNA encoding axonin-1. Peptides generated by enzymatic cleavage showed similarity to the AxCAM F11. Degenerated polymerase chain reaction (PCR) primers were designed and an axonin-1 fragment was amplified from mRNA of embryonic retina. Screening of a cDNA library from embryonic brain resulted in the isolation of a 4.0-kb cDNA insert with an open reading frame of 3108 nucleotides. The deduced polypeptide of 1036 amino acids includes a putative hydrophobic N-terminal signal sequence of 23 or 25 amino acids and a C-terminal hydrophobic sequence of 29 amino acids which is suggestive of sequences serving as signal for the attachment of a glycosyl-phosphatidylinositol (glycosyl-PtdIns) anchor. The putative mature form of axonin-1 comprises six immunoglobulin-like repeats, followed by four fibronectin-type III repeats. Axonin-1 exhibits 75% amino acid identity with the AxCAM TAG-1 of the rat, suggesting that it is the chicken homologue of TAG-1. Like TAG-1, axonin-1 is glycosyl-PtdIns-anchored to the neuronal membrane; in contrast to TAG-1, it does not exhibit an Arg-Gly-Asp sequence.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Chick Embryo , Contactin 2 , DNA/genetics , Glycosylation/drug effects , Immunoglobulins/genetics , Molecular Sequence Data , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoric Diester Hydrolases/metabolism , Plasmids , Polymerase Chain Reaction , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Tunicamycin/pharmacology , Vitreous Body/metabolism
17.
EMBO J ; 8(1): 55-63, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2714258

ABSTRACT

Using selective metabolic labelling in a compartmental cell culture system two proteins, denoted axonin-1 and axonin-2, were found to be secreted by axons of dorsal root ganglia neurons from chicken embryos. Based on its characteristic coordinates and spot morphology in two-dimensional gel electrophoresis, axonin-1 was detected in the cerebrospinal fluid and the vitreous fluid, axonin-1 was purified 476-fold to homogeneity by a four-step chromatographic procedure. The identity of the purified protein as axonin-1 was confirmed by immunological methods. Axonin-1 is a glycoprotein that subdivides into at least 16 immunologically similar isoelectric variants; their molecular weight range extends from 132 to 140 kd and their pI range from 5.3 to 6.2. In the vitreous fluid of the embryo, axonin-1 could first be detected on the embryonic day 5 and highest concentrations were measured during the second half of embryonic life; in the vitreous fluid of the adult chicken, concentrations were approximately 20 times lower. The early onset of secretion and the time course of expression suggest a role for axonin-1 in the development of the nervous system.


Subject(s)
Axons/metabolism , Glycoproteins/isolation & purification , Nerve Tissue Proteins/isolation & purification , Animals , Antibodies, Monoclonal/immunology , Cells, Cultured , Cerebrospinal Fluid/analysis , Chick Embryo , Ganglia, Spinal/cytology , Glycoproteins/metabolism , Isoelectric Focusing , Morphogenesis , Nerve Tissue Proteins/metabolism , Vitreous Body/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...