Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Physiol ; 588(Pt 21): 4303-15, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20736420

ABSTRACT

Vagal afferent fibres innervating thoracic structures such as the respiratory tract and oesophagus are diverse, comprising several subtypes of functionally distinct C-fibres and A-fibres. Both morphological and functional studies of these nerve subtypes would be advanced by selective, effective and long-term transduction of vagal afferent neurons with viral vectors. Here we addressed the hypothesis that vagal sensory neurons can be transduced with adeno-associated virus (AAV) vectors in vivo, in a manner that would be useful for morphological assessment of nerve terminals, using enhanced green fluorescent protein (eGFP), as well as for the selective knock-down of specific genes of interest in a tissue-selective manner. We found that a direct microinjection of AAV vectors into the vagal nodose ganglia in vivo leads to selective, effective and long-lasting transduction of the vast majority of primary sensory vagal neurons without transduction of parasympathetic efferent neurons. The transduction of vagal neurons by pseudoserotype AAV2/8 vectors in vivo is sufficiently efficient such that it can be used to functionally silence TRPV1 gene expression using short hairpin RNA (shRNA). The eGFP encoded by AAV vectors is robustly transported to both the central and peripheral terminals of transduced vagal afferent neurons allowing for bright imaging of the nerve endings in living tissues and suitable for structure-function studies of vagal afferent nerve endings. Finally, the AAV2/8 vectors are efficiently taken up by the vagal nerve terminals in the visceral tissue and retrogradely transported to the cell body, allowing for tissue-specific transduction.


Subject(s)
Adenoviridae/genetics , Gene Silencing/physiology , Genetic Vectors , Neurons, Afferent/physiology , TRPV Cation Channels/metabolism , Animals , Animals, Genetically Modified , Green Fluorescent Proteins/metabolism , Guinea Pigs , Models, Animal , Nodose Ganglion/cytology , Nodose Ganglion/metabolism , Patch-Clamp Techniques , TRPV Cation Channels/genetics
2.
Transplant Proc ; 37(8): 3523-6, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16298649

ABSTRACT

In May 2003, at Indiana University, the standard cold preservation solution University of Wisconsin (UW) solution was replaced by histidine-tryptophan ketogluatarate (HTK) solution. Earlier, we presented our initial experience with HTK in pancreas preservation with an analysis of the first 10 pancreas transplants. Here we report updated results with HTK in pancreas transplantation over the past 18 months. Between May 2003 and March 2005, a total of 87 pancreas transplants were performed with 78 of these organs utilizing HTK. Seventy five patients received 78 organ transplants. Surgical procedures performed were: simultaneous kidney pancreas transplantation (n = 50, 64%), pancreas after kidney transplantation (n = 19, 24%), solitary pancreas transplantation (n = 9, 12%). Donor and recipient data were collected with primary outcomes as primary nonfunction and 30-day graft and patient survivals, and compared to the UW cohort from our original report. Donor and recipient demographics were similar. Mean follow-up time is 12 +/- 6 months. The mean cold ischemia time was 9 +/- 3 hours. There were no cases of primary graft nonfunction. Thirty-day and 1-year patient survivals were 99% and 93%. The 30-day and 1-year graft survivals were 96% and 93%. There were five grafts lost, including three within the first month (two venous and one arterial thrombosis). There was one case of chronic rejection and one noncompliance. All other patients were insulin-independent by discharge. Serum fasting blood glucose and serial amylase remained comparable at all intervals posttransplantation. Within this range of cold ischemia time, HTK appears to provide effective pancreas preservation.


Subject(s)
Pancreas Transplantation/physiology , Pancreas/cytology , Adult , Cause of Death , Female , Glucose , Humans , Male , Mannitol , Middle Aged , Organ Preservation Solutions , Pancreas Transplantation/mortality , Potassium Chloride , Procaine , Racial Groups , Retrospective Studies , Survival Analysis
3.
Transplant Proc ; 37(5): 2016-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15964327

ABSTRACT

INTRODUCTION: University of Wisconsin (UW) solution is the standard preservation solution for organ transplantation. Histidine-tryptophan ketogluatarate (HTK) solution has been used increasingly for kidney, pancreas, and liver transplantation. This study compared HTK and UW used during kidney procurement with subsequent pulsatile perfusion. METHODS: Between January and October 2003, 91 deceased renal and simultaneous kidney pancreas transplants were performed (UW, n = 41, and HTK, n = 50). There were no differences with regard to donor and recipient demographics or cold ischemia. RESULTS: Delayed graft function occurred in 3 (7%) of UW and 4 (8%) of HTK-preserved kidneys (P = NS). There were no significant differences between patient or graft survival. There was an anticipated difference between total preservative volumes used (HTK: 4.1 +/- 1.0 vs UW: 3.0 +/- 0.5; P < .005). CONCLUSION: UW and HTK appear to have similar efficacy in kidney preservation with pulsatile perfusion. HTK preservation solution can be used safely in conjunction with pulsatile preservation for cold storage of renal allografts.


Subject(s)
Kidney Transplantation/physiology , Kidney , Organ Preservation Solutions , Adenosine , Adult , Allopurinol , Female , Glucose , Glutathione , Humans , Insulin , Male , Mannitol , Middle Aged , Pancreas Transplantation , Perfusion/methods , Potassium Chloride , Procaine , Raffinose , Safety , Tissue Donors/statistics & numerical data , Transplantation, Homologous
4.
MMWR Suppl ; 53: 166-72, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15714647

ABSTRACT

INTRODUCTION: The Walter Reed Army Institute of Research used the Electronic Surveillance System for the Early Notification of Community-Based Epidemics (ESSENCE) to conduct population-based behavioral health surveillance among military-health-system beneficiaries. The study analyzed the effectiveness of using prescribing patterns of psychotropic medications to monitor changes in a community's behavioral health status. OBJECTIVES: The objectives of this study were to 1) determine the feasibility of tracking psychiatric illnesses by monitoring prescriptions for psychiatric medications; 2) assess how often psychiatric medications are prescribed for patients with no record of psychiatric illness; 3) determine at what types of clinics these medications are prescribed most often and what other diagnoses are attributed to these patients; and 4) analyze data for potential changes in the population's mental health after high-stress events. METHODS: Correlation analysis and calculations of sensitivity and specificity were used to determine how well prescription medications correlate with outpatient diagnoses and how well they serve as proxies for outpatient diagnoses. A descriptive analysis was conducted of the types of clinics (e.g., primary care, behavioral health, or other specialty clinics) treating patients and the associated percentage of concurrence between prescriptions and diagnostic codes. RESULTS: In military treatment facilities, a diagnosis of depression or anxiety correlated significantly (r = 0.82) with antidepressant or anxiolytic prescriptions. Sensitivity of prescriptions when compared with outpatient visits was 0.76, and specificity was 0.94. Among those patients who visited a primary care clinic either the day before or the same day as an antidepressant or anxiolytic prescription was filled, 60.1% did not receive a diagnosis of any mental health disorder. Behavioral health clinics had the highest correlation between diagnoses and prescriptions; specialty clinics had the lowest. CONCLUSIONS: Behavioral health trends in a population can be monitored by automated analysis of prescribing patterns alone. This method might be a rapid indicator of needed mental health interventions after acute stress-inducing events and be more sensitive than tracking diagnoses alone.


Subject(s)
Drug Utilization , Health Behavior , Mental Disorders/epidemiology , Population Surveillance/methods , Public Health Informatics/instrumentation , Disease Outbreaks/prevention & control , Humans , Life Change Events , Mental Disorders/drug therapy , Psychotropic Drugs/therapeutic use
5.
Neuropharmacology ; 43(6): 966-78, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12423666

ABSTRACT

We report here that human Ntera-2/D1 (NT-2) cells, an undifferentiated committed neuronal progenitor cell line, endogenously express a functional P2Y(1) receptor, while other P2Y subtypes, except perhaps P2Y(4), are not functionally expressed. Quantitative RT-PCR analysis showed that NT-2 cells abundantly express mRNA for P2Y(1) and P2Y(11) receptors, while P2Y(2) and P2Y(4) receptors were detected at considerably lower levels. Western blot analysis also demonstrated expression of P2Y(1) receptors and Galpha(q/11) subunits. Various nucleotides induced intracellular Ca(2+) mobilisation in NT-2 cells in a concentration-dependent manner with a rank order potency of 2-MeSADP > 2-MeSATP > ADP > ATP > UTP > ATPgammaS, a profile resembling that of human P2Y(1) receptors. Furthermore, P2Y(1) receptor-specific (A3P5P) and P2Y-selective (PPADS, suramin) antagonists inhibited adenine nucleotide-induced Ca(2+) responses in a concentration-dependent manner, consistent with expression of a P2Y(1) receptor. Moreover, of seven adenine nucleotides tested, only Bz-ATP and ATPgammaS elicited small increases in cAMP formation suggesting that few, if any, functional P2Y(11) receptors were expressed. P2Y(1) receptor-selective adenine nucleotides, including 2-MeSADP and ADP, also induced concentration-dependent phosphorylation and hence, activation of the extracellular-signal regulated protein kinases (ERK1/2). NT-2 cells, therefore, provide a useful neuronal-like cellular model for studying the precise signalling pathways and physiological responses mediated by a native P2Y(1) receptor.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Adenosine Triphosphate/analogs & derivatives , Membrane Proteins , Neurons/metabolism , Pyridoxal Phosphate/analogs & derivatives , Receptors, Purinergic P2/metabolism , Stem Cells/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP , Fluorescence , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Platelet Aggregation Inhibitors/pharmacology , Pyridoxal Phosphate/pharmacology , RNA, Messenger/analysis , Receptors, Purinergic P2/classification , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y12 , Receptors, Purinergic P2Y2 , Reverse Transcriptase Polymerase Chain Reaction/methods , Suramin/pharmacology , Thionucleotides/pharmacology , Uridine Triphosphate/pharmacology
6.
Mil Med ; 166(11): 931-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11725319

ABSTRACT

The purpose of this study was to construct and evaluate the psychometric properties of an instrument to estimate the level of individual readiness among U.S. Army nurses. This study constitutes phase II of congressionally sponsored research to establish the degree to which Army nurses are prepared for the expectations of deployment. An expert panel established the validity of the initial readiness questionnaire. Changes were then incorporated into the first Readiness Estimate and Deployability Index (READI) questionnaire. Internal consistency and test-retest techniques assessed multiple reliabilities from pilot administrations. The READI was refined based on the results. Analysis of field administrations of the revised READI to three separate groups of nurses replicated earlier reliability results. Principle component analyses appear to support the hypothesized dimensional structure underlying questionnaire attitude items. The READI produced psychometrically stable ratings and results with great utility for the Army and potential adaptation for other military services.


Subject(s)
Military Nursing , Nurses/psychology , Psychology, Military , Psychometrics , Surveys and Questionnaires , Attitude of Health Personnel , Female , Humans , Male , United States
7.
Biochim Biophys Acta ; 1521(1-3): 107-19, 2001 Oct 31.
Article in English | MEDLINE | ID: mdl-11690642

ABSTRACT

The diverse biological actions of extracellular nucleotides in tissues and cells are mediated by two distinct classes of P2 receptor, P2X and P2Y. The G protein-coupled P2Y receptors comprise at least six mammalian subtypes (P2Y(1,2,4,6,11,12)), all of which have been cloned from human tissues, as well as other species. The P2Y receptor subtypes differ in their pharmacological selectivity for various adenosine and uridine nucleotides, which overlap in some cases. Data concerning the mRNA expression patterns of five P2Y receptors (P2Y(1,2,4,6,11)) in different human tissues and cells are currently quite limited, while P2Y mRNA distribution in the human brain has not previously been studied. In this study, we have addressed this deficiency in receptor expression data by using a quantitative reverse transcription-polymerase chain reaction approach to measure the precise mRNA expression pattern of each P2Y receptor subtype in a number of human peripheral tissues and brain regions, from multiple individuals, as well as numerous human cell lines and primary cells. All five P2Y receptors exhibited widespread yet subtype-selective mRNA expression profiles throughout the human tissues, brain regions and cells used. Our extensive expression data indicate the many potentially important roles of P2Y receptors throughout the human body, and will help in elucidating the physiological function of each receptor subtype in a wide variety of human systems.


Subject(s)
Polymerase Chain Reaction/methods , RNA, Messenger/metabolism , Receptors, Purinergic P2/metabolism , Actins/analysis , Brain/metabolism , Cell Line , Cyclophilins/analysis , DNA Probes , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/analysis , Humans , Male , Protein Isoforms/analysis , Protein Isoforms/metabolism , RNA, Messenger/analysis , Receptors, Purinergic P2/analysis , Reverse Transcriptase Polymerase Chain Reaction
8.
Biochem J ; 359(Pt 3): 507-16, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11672424

ABSTRACT

GSH-dependent prostaglandin D(2) synthase (PGDS) enzymes represent the only vertebrate members of class Sigma glutathione S-transferases (GSTs) identified to date. Complementary DNA clones encoding the orthologous human and rat GSH-dependent PGDS (hPGDS and rPGDS, respectively) have been expressed in Escherichia coli, and the recombinant proteins isolated by affinity chromatography. The purified enzymes were both shown to catalyse specifically the isomerization of prostaglandin (PG) H(2) to PGD(2). Each transferase also exhibited GSH-conjugating and GSH-peroxidase activities. The ability of hPGDS to catalyse the conjugation of aryl halides and isothiocyanates with GSH was found to be less than that of the rat enzyme. Whilst there is no difference between the enzymes with respect to their K(m) values for 1-chloro-2,4-dinitrobenzene, marked differences were found to exist with respect to their K(m) for GSH (8 mM versus 0.3 mM for hPGDS and rPGDS, respectively). Using molecular modelling techniques, amino acid substitutions have been identified in the N-terminal domain of these enzymes that lie outside the proposed GSH-binding site, which may explain these catalytic differences. The tissue-specific expression of PGDS also varies significantly between human and rat; amongst the tissues examined, variation in expression between the two species was most apparent in spleen and bone marrow. Differences in catalytic properties and tissue-specific expression of hPGDS and rPGDS appears to reflect distinct physiological roles for class Sigma GST between species. The evolution of divergent functions for the hPGDS and rPGDS is discussed in the context of the orthologous enzyme from chicken.


Subject(s)
Glutathione Transferase/metabolism , Intramolecular Oxidoreductases/metabolism , Amino Acid Sequence , Animals , Catalysis , Glutathione Transferase/classification , Glutathione Transferase/genetics , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/genetics , Isoenzymes/metabolism , Lipocalins , Models, Molecular , Molecular Sequence Data , Organ Specificity , Protein Conformation , Protein Structure, Tertiary , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Sequence Alignment
9.
Brain Res Mol Brain Res ; 93(1): 70-80, 2001 Sep 10.
Article in English | MEDLINE | ID: mdl-11532340

ABSTRACT

Failure of several putative neuroprotectants in large multicentred clinical trials has re-focussed attention on the predictability of pre-clinical animal models of stroke. Model characterisation and relationship to heterogeneous patient sub-groups remains of paramount importance. Information gained from magnetic resonance imaging (MRI) signatures indicates that the Zea Longa model of rat middle cerebral artery occlusion may be more representative of slowly evolving infarcts. Understanding the molecular changes over several hours following cerebral ischaemia will allow detailed characterisation of the adaptive response to brain injury. Using a fully characterised model of Zea Longa middle cerebral artery occlusion we have used the representational difference analysis (RDA) subtractive hybridisation method to identify transcripts that accumulate in the ischaemic cortex. Along with a number of established ischaemia-induced gene products (including MCP-1, TIMP-1, hsp 70) we were also able to identify nine genes which have not previously been shown to accumulate following focal ischaemia (including SOCS-3, GADD45gamma, Xin).


Subject(s)
Brain Chemistry/genetics , Infarction, Middle Cerebral Artery/physiopathology , Nucleic Acid Hybridization/methods , Organic Chemicals , Animals , Antigens, Surface/genetics , Benzothiazoles , Cytokines/genetics , Diamines , Fluorescent Dyes , Gene Expression/physiology , Gene Library , Heat-Shock Proteins/genetics , Male , Polymerase Chain Reaction , Quinolines , Rats , Rats, Sprague-Dawley
10.
Immunology ; 103(3): 319-31, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11454061

ABSTRACT

The presentation of extremely low doses of antigen to T cells is enhanced by immunoglobulin E (IgE)-dependent antigen focusing to CD23, the low-affinity receptor for IgE, expressed on activated B cells. CD23 contains a C-type lectin domain in its extracellular sequence and a targeting signal for coated pits, required for endocytosis, in its cytoplasmic sequence. CD23 is non-covalently associated with the major histocompatibility complex class II antigen, human leucocyte antigen HLA-DR, on the surface of human B cells, but the fate of this complex following endocytosis is unknown. To answer this question we have labelled these proteins on the surface of RPMI 8866 B cells and traced their route through the cytoplasm. Endocytosis mediated by anti-CD23 antibodies (BU38 and MHM6) led to the loss of CD23 from the cells. Endocytosis mediated by an antibody to HLA-DR (CR3/43) or an antigen-IgE complex (NP-BSA-anti-NP IgE), however, led to recycling of the HLA-DR-CD23 complex to the cell surface on a time scale (3-6 hr) consistent with the recycling of HLA-DR in antigen presentation. Along the latter pathway CD23 label was observed in cytoplasmic organelles that resembled the 'compartments for peptide loading' or 'class II vesicles' described by previous authors. Two features of the recycling process may contribute to the efficiency of antigen presentation. Peptide exchange may be facilitated by the proximity of HLA-DR and antigen in peptide loading compartments of the endosomal network. The return of CD23 with HLA-DR to the cell surface may then help to stabilize specific B-cell-T-cell interactions, contributing to T-cell activation.


Subject(s)
B-Lymphocytes/immunology , Endocytosis/immunology , HLA-DR Antigens/metabolism , Receptors, IgE/metabolism , Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , B-Lymphocytes/ultrastructure , Cell Culture Techniques , Cytoplasmic Vesicles/immunology , Electrophoresis, Polyacrylamide Gel , Endosomes/immunology , Endosomes/ultrastructure , HLA-DR Antigens/immunology , Humans , Immunoglobulin E/immunology , Microscopy, Confocal , Microscopy, Electron , Receptors, IgE/immunology
11.
J Biol Chem ; 276(31): 28969-75, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11387329

ABSTRACT

A novel human G protein-coupled receptor named AXOR12, exhibiting 81% homology to the rat orphan receptor GPR54, was cloned from a human brain cDNA library. Heterologous expression of AXOR12 in mammalian cells permitted the identification of three surrogate agonist peptides, all with a common C-terminal amidated motif. High potency agonism, indicative of a cognate ligand, was evident from peptides derived from the gene KiSS-1, the expression of which prevents metastasis in melanoma cells. Quantitative reverse transcriptase-polymerase chain reaction was used to study the expression of AXOR12 and KiSS-1 in a variety of tissues. The highest levels of expression of AXOR12 mRNA were observed in brain, pituitary gland, and placenta. The highest levels of KiSS-1 gene expression were observed in placenta and brain. A polyclonal antibody raised to the C terminus of AXOR12 was generated and used to show localization of the receptor to neurons in the cerebellum, cerebral cortex, and brainstem. The biological significance of these expression patterns and the nature of the putative cognate ligand for AXOR12 are discussed.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Proteins/genetics , Proteins/metabolism , Receptors, Cell Surface/genetics , Amino Acid Sequence , Animals , Brain/metabolism , CHO Cells , Cricetinae , Female , Genes, Tumor Suppressor , Humans , Kinetics , Kisspeptins , Ligands , Melanoma/genetics , Molecular Sequence Data , Nephropidae , Neurons/metabolism , Organ Specificity , Peptide Fragments/pharmacology , Pituitary Gland/metabolism , Placenta/metabolism , Pregnancy , Proteins/chemistry , Rats , Receptors, Cell Surface/chemistry , Receptors, G-Protein-Coupled , Receptors, Kisspeptin-1 , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sea Anemones , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Tumor Suppressor Proteins
12.
J Biol Chem ; 276(23): 20125-9, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11274220

ABSTRACT

Melanin-concentrating hormone (MCH) is involved in the regulation of feeding and energy homeostasis. Recently, a 353-amino acid splice variant form of the human orphan receptor SLC-1 () (hereafter referred to as MCH(1)) was identified as an MCH receptor. This report describes the cloning and functional characterization of a novel second human MCH receptor, which we designate MCH(2), initially identified in a genomic survey sequence as being homologous to MCH(1) receptors. Using this sequence, a full-length cDNA was generated with an open reading frame of 1023 base pairs, encoding a polypeptide of 340 amino acids, with 38% identity to MCH(1) and with many of the structural features conserved in G protein-coupled receptors. This newly discovered receptor belongs to class 1 (rhodopsin-like) of the G protein-coupled receptor superfamily. HEK293 cells transfected with MCH(2) receptors responded to nanomolar concentrations of MCH with an increase in intracellular Ca(2+) levels and increased cellular extrusion of protons. In addition, fluorescently labeled MCH bound with nanomolar affinity to these cells. The tissue localization of MCH(2) receptor mRNA, as determined by quantitative reverse transcription-polymerase chain reaction, was similar to that of MCH(1) in that both receptors are expressed predominantly in the brain. The discovery of a novel MCH receptor represents a new potential drug target and will allow the further elucidation of MCH-mediated responses.


Subject(s)
Hypothalamic Hormones/metabolism , Melanins/metabolism , Pituitary Hormones/metabolism , Receptors, Pituitary Hormone/genetics , Amino Acid Sequence , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary , Humans , Molecular Sequence Data , Receptors, G-Protein-Coupled , Receptors, Pituitary Hormone/chemistry , Receptors, Pituitary Hormone/metabolism , Sequence Homology, Amino Acid
13.
Brain Res ; 892(1): 94-101, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11172753

ABSTRACT

We have cloned and functionally expressed the human orthologue of the mouse TRAAK gene. When cDNA for hTRAAK is expressed in either Xenopus oocytes or HEK293 cells it forms a K(+)-selective conductance and hyperpolarises the resting membrane potential. Quantitative mRNA expression analysis using Taqman revealed that hTRAAK mRNA is predominantly present in the central nervous system where it exhibits a regionally diverse pattern of expression. Like the related channel TREK-1, the activity of TRAAK was potentiated by arachidonic acid. The neuroprotective agent sipatrigine (10 microM) inhibited both hTREK-1 (73.3+/-4.4%) and hTRAAK (45.1+/-11.2%) in a reversible, voltage-independent manner. Inhibition of both channels was dose-dependent and for TREK-1 occurred with an IC(50) of 4 microM. The related compound lamotrigine, which is a better anticonvulsant but weaker neuroprotective agent than sipatrigine, was a far less effective antagonist of both channels, producing <10% inhibition at a concentration of 10 microM.


Subject(s)
Brain/physiology , Neuroprotective Agents/pharmacology , Piperazines/pharmacology , Potassium Channels, Tandem Pore Domain , Potassium Channels/physiology , Pyrimidines/pharmacology , Amino Acid Sequence , Animals , Cell Line , Female , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Molecular Sequence Data , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Oocytes/drug effects , Oocytes/physiology , Potassium Channel Blockers , Potassium Channels/chemistry , Potassium Channels/genetics , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Xenopus laevis
14.
Osteoarthritis Cartilage ; 9(8): 727-37, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11795992

ABSTRACT

OBJECTIVE: To characterize the expression pattern of clusterin in adult human normal and osteoarthritic cartilage. METHODS: Clusterin mRNA expression in adult human normal and osteoarthritic cartilage was investigated by analysis of cDNA libraries, TaqMan quantitative RT-PCR, microarray and in situ hybridization. RESULTS: Sequence analysis of ESTs from adult human normal and osteoarthritic cartilage cDNA libraries demonstrated that the abundance of clusterin in these libraries was equivalent to genes which have been more commonly associated with cartilage. To examine tissue distribution, TaqMan Quantitative PCR analysis was performed using RNA from a panel of individual normal tissues. Clusterin was expressed at significant levels in cartilage, brain, liver, and pancreas. The expression of clusterin mRNA was up-regulated in early osteoarthritic vs normal cartilage when analysed by microarray analysis. Using in situ hybridization, chondrocytes of normal cartilage expressed moderate levels of clusterin. Upper mid-zone chondrocytes in cartilage with early stages of osteoarthritic disease expressed high levels of clusterin mRNA. In advanced osteoarthritic cartilage, the overall expression of clusterin was reduced. CONCLUSION: The induction of clusterin has been associated with a variety of disease states where it appears to provide a cytoprotective effect. The increased expression of clusterin mRNA in the early stages of osteoarthritis (OA) may reflect an attempt by the chondrocytes to protect and repair the tissue. In contrast, the decrease in clusterin mRNA in the advanced osteoarthritic cartilage accompanies the final degenerative stages of the disease. An understanding of the expression of clusterin in osteoarthritis may allow consideration of this protein as a marker for cartilage changes in this chronic degenerative condition.


Subject(s)
Cartilage, Articular/metabolism , Glycoproteins/metabolism , Molecular Chaperones/metabolism , Osteoarthritis/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Clusterin , Female , Gene Library , Humans , In Situ Hybridization/methods , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Proteoglycans/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
15.
Mol Cell Neurosci ; 16(5): 609-19, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11083922

ABSTRACT

Sequential proteolytic processing of the Amyloid Precursor Protein (APP) by beta- and gamma-secretases generates the 4-kDa amyloid (A beta) peptide, a key component of the amyloid plaques seen in Alzheimer's disease (AD). We and others have recently reported the identification and characterisation of an aspartic proteinase, Asp2 (BACE), as beta-secretase. Here we describe the characterization of a second highly related aspartic proteinase, Asp1 as a second beta-secretase candidate. Asp1 is expressed in brain as detected at the mRNA level and at the protein level. Transient expression of Asp1 in APP-expressing cells results in an increase in the level of beta-secretase-derived soluble APP and the corresponding carboxy-terminal fragment. Paradoxically there is a decrease in the level of soluble A beta secreted from the cells. Asp1 colocalizes with APP in the Golgi/endoplasmic reticulum compartments of cultured cells. Asp1, when expressed as an Fc fusion protein (Asp1-Fc), has the N-terminal sequence ALEP..., indicating that it has lost the prodomain. Asp1-Fc exhibits beta-secretase activity by cleaving both wild-type and Swedish variant (KM/NL) APP peptides at the beta-secretase site.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/analysis , Amyloid beta-Protein Precursor/chemistry , Animals , Aspartic Acid Endopeptidases/chemistry , Binding Sites/physiology , COS Cells , Cloning, Molecular , Endopeptidases , Female , Glycoproteins/analysis , Humans , Male , Membrane Proteins/analysis , Molecular Sequence Data , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
16.
Brain Res Mol Brain Res ; 82(1-2): 74-83, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-11042359

ABSTRACT

We have isolated, by degenerate PCR, a complementary DNA encoding a novel two pore domain potassium channel. This is the 7th functional member of the human tandem pore domain potassium channel family to be reported. It has an open reading frame of 1.125 kb and encodes a 374 amino acid protein which shows 62% identity to the human TASK-1 gene: identity to other human members of the family is 31-35% at the amino acid level. We believe this gene to be human TASK-3, the ortholog of the recently reported rat TASK-3 gene: amino acid identity between the two is 74%. 'Taqman' mRNA analysis demonstrated a very specific tissue distribution pattern, showing human TASK-3 mRNA to be localised largely in the cerebellum, in contrast rat TASK-3 was reported to be widely distributed. We have shown by radiation hybrid mapping that human TASK-3 can be assigned to chromosome 8q24.3. Human TASK-3 was demonstrated to endow Xenopus oocytes with a negative resting membrane potential through the presence of a large K(+) selective conductance. TASK-3 is inhibited by extracellular acidosis with a mid-point of inhibition around pH 6. 5, supporting the predictions from the sequence data that this is a third human TASK (TWIK-related acid sensitive K(+) channel) gene.


Subject(s)
Cerebellum/metabolism , Chromosomes, Human, Pair 8 , Evoked Potentials/physiology , Nerve Tissue Proteins , Potassium Channels, Tandem Pore Domain , Potassium Channels/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , Genetic Variation , Humans , Membrane Potentials/physiology , Molecular Sequence Data , Oocytes/physiology , Phylogeny , Polymerase Chain Reaction , Potassium Channels/chemistry , Potassium Channels/physiology , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
17.
Biochem Biophys Res Commun ; 273(3): 805-10, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10891327

ABSTRACT

We have identified a cDNA, designated HOFNH30, which encodes a 354 amino acid G-protein-coupled receptor (GPCR). This receptor has 96% amino acid identity to the Jurkat-T cell-derived EDG7 and could be a splice variant. RT-PCR analysis demonstrated that HOFNH30 mRNA is expressed in placenta whereas EDG7 mRNA shows highest expression in prostate. The HOFNH30 gene is localized to human chromosome 1p22. 3-1p31.1. When HOFNH30 was expressed in RBL-2H3 cells, LPA and phosphatidic acid (PA) induced a calcium mobilization response with EC(50) values of 13 nM and 3 microM, respectively. LPA also induced phosphorylation of mitogen-activated protein kinase (p42(MAPK) and p44(MAPK)) in HOFNH30-transfected but not vector-transfected RBL-2H3 cells. In the present study, we have identified a novel variant from the EDG receptor family, a GPCR for which LPA is a high-affinity endogenous ligand.


Subject(s)
GTP-Binding Proteins/metabolism , Lysophospholipids/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Amino Acid Sequence , Base Sequence , Calcium/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 1 , Cloning, Molecular , Enzyme Activation , Humans , Jurkat Cells , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phosphorylation , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Lysophosphatidic Acid , Sequence Homology, Amino Acid , Tumor Cells, Cultured
18.
J Allergy Clin Immunol ; 105(6 Pt 1): 1093-100, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10856141

ABSTRACT

BACKGROUND: Eosinophils, basophils, and mast cells are believed to be the central tenet cells in allergic conditions including allergic rhinitis, asthma, and eczema. The molecular mechanisms underlying the recruitment of these cells to sites of allergic inflammation are poorly understood. OBJECTIVES: Our aim was to identify a common adhesion molecule that could potentially be responsible for mediating the recruitment of the allergic cell types to the lungs and other sites of allergy. METHODS: We have cloned a sialoadhesin molecule from a human eosinophil library with the use of expressed sequence tag technology and characterized its expression on allergic cells by the use of flow cytometry and specific mAbs. RESULTS: With the use of expressed sequence tag sequencing, we have identified a novel siglec molecule, SAF-2. SAF-2 has homology with other sialoadhesin family members (CD33 and siglec-5) and belongs to a subgroup of the Ig superfamily. SAF-2 is a 431-amino acid protein composed of 3 Ig domains with a 358-amino acid extracellular domain and a 47-amino acid tail. SAF-2 is highly restricted to eosinophils, basophils, and mast cells. Antibodies to SAF-2 do not modulate Ca(++) mobilization or chemotaxis of human eosinophils induced by eotaxin. CONCLUSION: SAF-2 is a highly restricted sialoadhesin molecule, which may be useful in the detection and/or modulation of allergic cells.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Antigens, Surface/biosynthesis , Basophils/metabolism , Eosinophils/metabolism , Hypersensitivity/pathology , Lectins , Mast Cells/metabolism , Antigens, CD/genetics , Antigens, CD/physiology , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/physiology , Antigens, Surface/genetics , Antigens, Surface/physiology , Erythrocytes/metabolism , Gene Expression , Humans , N-Acetylneuraminic Acid/pharmacology , RNA, Messenger/genetics , Sequence Homology, Amino Acid
19.
J Biol Chem ; 275(34): 25965-71, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10851242

ABSTRACT

Opiate tolerance and dependence are major clinical and social problems. The anti-opiate neuropeptides FF and AF (NPFF and NPAF) have been implicated in pain modulation as well as in opioid tolerance and may play a critical role in this process, although their mechanism of action has remained unknown. Here we describe a cDNA encoding a novel neuropeptide Y-like human orphan G protein-coupled receptor (GPCR), referred to as HLWAR77 for which NPAF and NPFF have high affinity. Cells transiently or stably expressing HLWAR77 bind and respond in a concentration-dependent manner to NPAF and NPFF and are also weakly activated by FMRF-amide (Phe-Met-Arg-Phe-amide) and a variety of related peptides. The high affinity and potency of human NPFF and human NPAF for HLWAR77 strongly suggest that these are the cognate ligands for this receptor. Expression of HLWAR77 was demonstrated in brain regions associated with opiate activity, consistent with the pain-modulating activity of these peptides, whereas the expression in adipose tissue suggests other physiological and pathophysiological activities for FMRF-amide neuropeptides. The discovery that the anti-opiate neuropeptides are the endogenous ligands for HLWAR77 will aid in defining the physiological role(s) of these ligands and facilitate the identification of receptor agonists and antagonists.


Subject(s)
Neuropeptides/metabolism , Oligopeptides/metabolism , Receptors, Neuropeptide/metabolism , Amino Acid Sequence , Arrestins/metabolism , Base Sequence , Calcium/metabolism , Cell Line , FMRFamide/pharmacology , Humans , Ligands , Molecular Sequence Data , Receptors, Neuropeptide/genetics , beta-Arrestins
20.
J Biol Chem ; 275(27): 20247-50, 2000 Jul 07.
Article in English | MEDLINE | ID: mdl-10811630

ABSTRACT

Neuromedins are a family of peptides best known for their contractile activity on smooth muscle preparations. The biological mechanism of action of neuromedin U remains unknown, despite the fact that the peptide was first isolated in 1985. Here we show that neuromedin U potently activates the orphan G protein-coupled receptor FM3, with subnanomolar potency, when FM3 is transiently expressed in human HEK-293 cells. Neuromedins B, C, K, and N are all inactive at this receptor. Quantitative reverse transcriptase-polymerase chain reaction analysis of neuromedin U expression in a range of human tissues showed that the peptide is highly expressed in the intestine, pituitary, and bone marrow, with lower levels of expression seen in stomach, adipose tissue, lymphocytes, spleen, and the cortex. Similar analysis of FM3 expression showed that the receptor is widely expressed in human tissue with highest levels seen in adipose tissue, intestine, spleen, and lymphocytes, suggesting that neuromedin U may have a wide range of presently undetermined physiological effects. The discovery that neuromedin U is an endogenous agonist for FM3 will significantly aid the study of the full physiological role of this peptide.


Subject(s)
GTP-Binding Proteins/metabolism , Membrane Proteins , Neuropeptides/pharmacology , Receptors, Cell Surface/agonists , Receptors, Neurotransmitter , Calcium/metabolism , Cell Line , Cloning, Molecular , Gene Expression Regulation , Humans , Inositol Phosphates/metabolism , Neuropeptides/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...