Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 276(51): 48300-8, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11602597

ABSTRACT

Connexin 43 (Cx43) hexameric hemichannels, recently demonstrated to mediate NAD(+) transport, functionally interact in the plasma membrane of several cells with the ectoenzyme CD38 that converts NAD(+) to the universal calcium mobilizer cyclic ADP-ribose (cADPR). Here we demonstrate that functional uncoupling between CD38 and Cx43 in CD38-transfected 3T3 murine fibroblasts is paralleled by decreased [Ca(2+)](i) levels as a result of reduced intracellular conversion of NAD(+) to cADPR. A sharp inverse correlation emerged between [Ca(2+)](i) levels and NAD(+) transport (measured as influx into cells and as efflux therefrom), both in the CD38(+) cells (high [Ca(2+)](i), low transport) and in the CD38(-) fibroblasts (low [Ca(2+)](i), high transport). These differences were correlated with distinctive extents of Cx43 phosphorylation in the two cell populations, a lower phosphorylation with high NAD(+) transport (CD38(-) cells) and vice versa (CD38(+) cells). Conversion of NAD(+)-permeable Cx43 to the phosphorylated, NAD(+)-impermeable form occurs via Ca(2+)-stimulated protein kinase C (PKC). Thus, a self-regulatory loop emerged in CD38(+) fibroblasts whereby high [Ca(2+)](i) restricts further Ca(2+) mobilization by cADPR via PKC-mediated disruption of the Cx43-CD38 cross-talk. This mechanism may avoid: (i) leakage of NAD(+) from cells; (ii) depletion of intracellular NAD(+) by CD38; (iii) overproduction of intracellular cADPR resulting in potentially cytotoxic [Ca(2+)](i).


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Antigens, CD , Antigens, Differentiation/metabolism , Calcium/metabolism , Connexin 43/metabolism , NAD+ Nucleosidase/metabolism , NAD/metabolism , 3T3 Cells , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Base Sequence , Cyclic ADP-Ribose , DNA Primers , Enzyme Activation , Fibroblasts/enzymology , Fibroblasts/metabolism , Membrane Glycoproteins , Mice , Phosphorylation , Protein Kinase C/metabolism
2.
J Neurochem ; 78(3): 646-57, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11483668

ABSTRACT

Astrocytes possess different, efficient ways to generate complex changes in intracellular calcium concentrations, which allow them to communicate with each other and to interact with adjacent neuronal cells. Here we show that cultured hippocampal astrocytes coexpress the ectoenzyme CD38, directly involved in the metabolism of the calcium mobilizer cyclic ADP-ribose, and the NAD+ transporter connexin 43. We also demonstrate that hippocampal astrocytes can release NAD+ and respond to extracellular NAD+ or cyclic ADP-ribose with intracellular calcium increases, suggesting the existence of an autocrine cyclic ADP-ribose-mediated signalling. Cyclic ADP-ribose-induced calcium changes are in turn responsible for an increased glutamate and GABA release, this effect being completely inhibited by the cyclic ADP-ribose specific antagonist 8-NH2-cADPR. Furthermore, addition of NAD+ to astrocyte-neuron co-cultures results in a delayed intracellular calcium transient in neuronal cells, which is strongly but not completely inhibited by glutamate receptor blockers. These data indicate that an astrocyte-to-neuron calcium signalling can be triggered by the CD38/cADPR system, which, through the activation of intracellular calcium responses in astrocytes, is in turn responsible for the increased release of neuromodulators from glial cells.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Antigens, CD , Antigens, Differentiation/metabolism , Astrocytes/metabolism , Calcium Signaling/physiology , NAD+ Nucleosidase/metabolism , Neurotransmitter Agents/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/antagonists & inhibitors , Administration, Topical , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Differentiation/genetics , Cell Communication/physiology , Cells, Cultured , Coculture Techniques , Connexin 43/genetics , Connexin 43/metabolism , Cyclic ADP-Ribose , Glutamic Acid/metabolism , Glycyrrhetinic Acid/pharmacology , Hippocampus/cytology , Immunoblotting , Membrane Glycoproteins , Microscopy, Fluorescence , Models, Neurological , NAD/metabolism , NAD/pharmacology , NAD+ Nucleosidase/genetics , Neurons/metabolism , Rats , Spectrometry, Fluorescence , gamma-Aminobutyric Acid/metabolism
4.
Pediatr Res ; 49(4): 537-42, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11264438

ABSTRACT

Leukocyte adhesion deficiency type II is an autosomal recessive syndrome characterized by generalized reduction of L-fucose in glycoconjugates; the specific molecular defect is still undefined. The most important clinical symptoms include severe growth and mental retardation and severe immunodeficiency. Patients from two ethnic groups have been reported, i.e. Arab and Turkish. We have observed that GDP-L-fucose transport into Golgi vesicles was specifically impaired in an Arab patient, with a significant reduction of the V:(max) but no significant differences in the K:(m) from control and parents. GDP-L-fucose transport showed simple saturation kinetics in all samples. Transport of UDP-galactose, UDP-N:-acetylglucosamine, and CMP-sialic acid was comparable into vesicles from the Arab patient, parents, and control. These kinetic parameters probably account for the failure to obtain any clinical and biochemical response to fucose therapy in Arab patients. This contrasts both with the distinctive kinetic properties of GDP-L-fucose transport and with the success of fucose therapy, which have been recently reported in one patient of Turkish origin. Accordingly, the biochemical properties of GDP-L-fucose transport into the Golgi are consistent with different variants of leukocyte adhesion deficiency type II that are probably the result of different molecular defects.


Subject(s)
Fucose/therapeutic use , Golgi Apparatus/metabolism , Guanosine Diphosphate Fucose/metabolism , Leukocyte-Adhesion Deficiency Syndrome/drug therapy , Biological Transport , Leukocyte-Adhesion Deficiency Syndrome/metabolism
5.
J Biol Chem ; 276(24): 21642-8, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11274199

ABSTRACT

CD38 is a bifunctional ectoenzyme synthesizing from NAD(+) (ADP-ribosyl cyclase) and degrading (hydrolase) cyclic ADP-ribose (cADPR), a powerful universal calcium mobilizer from intracellular stores. Recently, hexameric connexin 43 (Cx43) hemichannels have been shown to release cytosolic NAD(+) from isolated murine fibroblasts (Bruzzone, S., Guida, L., Zocchi, E., Franco, L. and De Flora, A. (2001) FASEB J. 15, 10-12), making this dinucleotide available to the ectocellular active site of CD38. Here we investigated transwell co-cultures of CD38(+) (transfected) and CD38(-) 3T3 cells in order to establish the role of extracellular NAD(+) and cADPR on [Ca(2+)](i) levels and on proliferation of the CD38(-) target cells. CD38(+), but not CD38(-), feeder cells induced a [Ca(2+)](i) increase in the CD38(-) target cells which was comparable to that observed with extracellular cADPR alone and inhibitable by NAD(+)-glycohydrolase or by the cADPR antagonist 8-NH(2)-cADPR. Addition of recombinant ADP-ribosyl cyclase to the medium of CD38(-) feeders induced sustained [Ca(2+)](i) increases in CD38(-) target cells. Co-culture on CD38(+) feeders enhanced the proliferation of CD38(-) target cells over control values and significantly shortened the S phase of cell cycle. These results demonstrate a paracrine process based on Cx43-mediated release of NAD(+), its CD38-catalyzed conversion to extracellular cADPR, and influx of this nucleotide into responsive cells to increase [Ca(2+)](i) and stimulate cell proliferation.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Antigens, CD , Antigens, Differentiation/metabolism , Calcium/metabolism , Cell Division/physiology , NAD+ Nucleosidase/metabolism , NAD/metabolism , 3T3 Cells , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/pharmacology , Animals , Antigens, Differentiation/chemistry , Antigens, Differentiation/genetics , Binding Sites , Cell Membrane/metabolism , Coculture Techniques , Connexin 43/genetics , Connexin 43/physiology , Cyclic ADP-Ribose , Cytosol/metabolism , Kinetics , Membrane Glycoproteins , Mice , Models, Biological , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , NAD+ Nucleosidase/chemistry , NAD+ Nucleosidase/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
6.
Am J Physiol Lung Cell Mol Physiol ; 280(1): L98-L106, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11133499

ABSTRACT

Cyclic ADP-ribose (cADPR), a universal calcium releaser, is generated from NAD(+) by an ADP-ribosyl cyclase and is degraded to ADP-ribose by a cADPR hydrolase. In mammals, both activities are expressed as ectoenzymes by the transmembrane glycoprotein CD38. CD38 was identified in both epithelial cells and smooth myocytes isolated from bovine trachea. Intact tracheal smooth myocytes (TSMs) responded to extracellular cADPR (100 microM) with an increase in intracellular calcium concentration ([Ca(2+)](i)) both at baseline and after acetylcholine (ACh) stimulation. The nonhydrolyzable analog 3-deaza-cADPR (10 nM) elicited the same effects as cADPR, whereas the cADPR antagonist 8-NH(2)-cADPR (10 microM) inhibited both basal and ACh-stimulated [Ca(2+)](i) levels. Extracellular cADPR or 3-deaza-cADPR caused a significant increase of ACh-induced contraction in tracheal smooth muscle strips, whereas 8-NH(2)-cADPR decreased it. Tracheal mucosa strips, by releasing NAD(+), enhanced [Ca(2+)](i) in isolated TSMs, and this increase was abrogated by either NAD(+)-ase or 8-NH(2)-cADPR. These data suggest the existence of a paracrine mechanism whereby mucosa-released extracellular NAD(+) plays a hormonelike function and cADPR behaves as second messenger regulating calcium-related contractility in TSMs.


Subject(s)
Acetylcholine/pharmacology , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Antigens, CD , Bronchoconstriction/physiology , Cyclic ADP-Ribose/analogs & derivatives , Muscle, Smooth/physiology , Trachea/physiology , Vasodilator Agents/pharmacology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/pharmacology , Animals , Antigens, Differentiation/analysis , Bronchoconstriction/drug effects , Calcium/metabolism , Cattle , Extracellular Space/enzymology , In Vitro Techniques , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/enzymology , Muscle, Smooth/cytology , N-Glycosyl Hydrolases/metabolism , N-Glycosyl Hydrolases/pharmacology , NAD/metabolism , NAD+ Nucleosidase/analysis , Paracrine Communication/physiology , Receptor Cross-Talk/physiology , Respiratory Mucosa/chemistry , Respiratory Mucosa/enzymology , Trachea/cytology
7.
J Mol Biol ; 303(1): 77-91, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-11021971

ABSTRACT

GDP-4-keto-6-deoxy-d-mannose epimerase/reductase is a bifunctional enzyme responsible for the last step in the biosynthesis of GDP-l-fucose, the substrate of fucosyl transferases. Several cell-surface antigens, including the leukocyte Lewis system and cell-surface antigens in pathogenic bacteria, depend on the availability of GDP-l-fucose for their expression. Therefore, the enzyme is a potential target for therapy in pathological states depending on selectin-mediated cell-to-cell interactions. Previous crystallographic investigations have shown that GDP-4-keto-6-deoxy-d-mannose epimerase/reductase belongs to the short-chain dehydrogenase/reductase protein homology family. The enzyme active-site region is at the interface of an N-terminal NADPH-binding domain and a C-terminal domain, held to bind the substrate. The design, expression and functional characterization of seven site-specific mutant forms of GDP-4-keto-6-deoxy-d-mannose epimerase/reductase are reported here. In parallel, the crystal structures of the native holoenzyme and of three mutants (Ser107Ala, Tyr136Glu and Lys140Arg) have been investigated and refined at 1. 45-1.60 A resolution, based on synchrotron data (R-factors range between 12.6 % and 13.9 %). The refined protein models show that besides the active-site residues Ser107, Tyr136 and Lys140, whose mutations impair the overall enzymatic activity and may affect the coenzyme binding mode, side-chains capable of proton exchange, located around the expected substrate (GDP-4-keto-6-deoxy-d-mannose) binding pocket, are selectively required during the epimerization and reduction steps. Among these, Cys109 and His179 may play a primary role in proton exchange between the enzyme and the epimerization catalytic intermediates. Finally, the additional role of mutated active-site residues involved in substrate recognition and in enzyme stability has been analyzed.


Subject(s)
Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/metabolism , Escherichia coli Proteins , Escherichia coli/enzymology , Guanosine Diphosphate Mannose/analogs & derivatives , Ketone Oxidoreductases , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Mutagenesis, Site-Directed/genetics , Sugar Alcohol Dehydrogenases/chemistry , Sugar Alcohol Dehydrogenases/metabolism , Amino Acid Substitution/genetics , Binding Sites , Carbohydrate Epimerases/genetics , Catalysis , Chromatography, Thin Layer , Crystallography, X-Ray , Deoxy Sugars/analysis , Deoxy Sugars/metabolism , Enzyme Stability , Fucose/analogs & derivatives , Fucose/chemistry , Fucose/metabolism , Guanosine Diphosphate Mannose/chemistry , Guanosine Diphosphate Mannose/metabolism , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Hydrogen Bonding , Kinetics , Models, Molecular , Multienzyme Complexes/genetics , Mutation/genetics , NADP/metabolism , Protein Conformation , Protons , Structure-Activity Relationship , Substrate Specificity , Sugar Alcohol Dehydrogenases/genetics
9.
FASEB J ; 14(5): 680-90, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10744625

ABSTRACT

Cyclic ADP-ribose (cADPR) is a universal second messenger that regulates many calcium-related cellular events by releasing calcium from intracellular stores. Since these events include enhanced cell proliferation and since the bone marrow harbors both ectoenzymes that generate cADPR from NAD(+) (CD38 and BST-1), we investigated the effects of extracellular cADPR on human hemopoietic progenitors (HP). Exposure of HP to 100 microM cADPR for 24 h induced a significant increase in colony output (P<0.01) and colony size (P<0.003). A horizontal expansion of HP, as demonstrated by a markedly increased replating efficiency in semisolid medium (up to 700 times compared to controls), was also observed, indicating that cADPR priming can affect cell growth for multiple generations over several weeks after exposure. Influx of extracellular cADPR into the cells was demonstrated, and a causal relationship between the functional effects and the increase of intracellular free calcium concentration induced by cADPR on HP was established through the use of specific antagonists. Similar effects on HP were produced by nanomolar concentrations of the nonhydrolyzable cADPR analog 3-deaza-cADPR. These data demonstrate that extracellular cADPR behaves as a cytokine enhancing the proliferation of human HP, a finding that may have biomedical applications for the ex vivo expansion of hemopoietic cells.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Calcium/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Adenosine Diphosphate Ribose/antagonists & inhibitors , Adenosine Diphosphate Ribose/metabolism , Adenosine Diphosphate Ribose/pharmacology , Antigens, CD34/metabolism , Cell Division/drug effects , Cells, Cultured , Colony-Forming Units Assay , Cyclic ADP-Ribose , Cytarabine/pharmacology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Humans , Intracellular Fluid/metabolism , Second Messenger Systems
10.
FEBS Lett ; 456(3): 370-4, 1999 Aug 13.
Article in English | MEDLINE | ID: mdl-10462046

ABSTRACT

GDP-D-mannose-4,6-dehydratase (GMD) is the key enzyme in the 'de novo' pathway of GDP-L-fucose biosynthesis. The reported cDNA sequences for human GMD predict three forms of different length, whose 'in vivo' occurrence and molecular properties are completely undefined. Here, we report the expression in Escherichia coli and the properties of each native recombinant GMD form. Only the 42 kDa long GMD (L-GMD) and the 40.2 kDa (M-GMD) forms were recovered as soluble functional proteins, while the 38.7 kDa form, short GMD (S-GMD), lacking an N-terminal domain critical for dinucleotide binding, was inactive and formed a precipitate. Both L-GMD and M-GMD are homodimers and contain 1 mol of tightly bound NADP+. Their kinetic properties (Km, Kcat) are apparently identical and both forms are non-competitively feedback-inhibited by GDP-L-fucose to a similar extent. M-GMD is the predominant enzyme form expressed in several human cell lines. These data seem to suggest that modulation of the 'de novo' pathway of GDP-L-fucose biosynthesis involves mechanisms other than differential 'in vivo' expression of GMD forms.


Subject(s)
Hydro-Lyases/chemistry , Hydro-Lyases/metabolism , Amino Acid Sequence , Blotting, Western , Cell Line , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Glutathione Transferase/metabolism , Humans , Hydro-Lyases/antagonists & inhibitors , Isoenzymes/metabolism , Kinetics , Molecular Sequence Data , NADP/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
11.
J Acquir Immune Defic Syndr ; 21(2): 81-9, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10360798

ABSTRACT

Macrophages play a key role in AIDS pathogenesis and thus controlling infectivity and viral replication in these cells is a key issue in any antiretroviral therapy. In the present study, using a murine model of AIDS, we evaluated new therapeutic approaches specifically designed for the protection of macrophages. Based on previous observations, we took advantage of the unique ability of autologous erythrocytes to deliver drugs selectively to macrophages. The antiviral drugs selected were a new homodimer of AZT (AZTp2AZT) and reduced glutathione (GSH). The addition of an oral drug for the protection of lymphocytes (i.e., AZT) was also investigated. C57BL/6 mice infected with the retroviral complex LP-BM5 were treated with GSH-loaded erythrocytes, GSH-loaded erythrocytes plus oral AZT, or GSH/AZTp2AZT-loaded erythrocytes plus oral AZT. The treatments including AZT and erythrocytes loaded with GSH alone or with GSH plus AZTp2AZT provided similar results and were most effective in inhibiting the progression of MAIDS; they reduced splenomegaly, lymphadenopathy, and hypergammaglobulinemia by about 70%, 90% and 83%, respectively, when compared with infected animals at 10 weeks postinfection. Evaluation of BM5d proviral DNA content in infected organs revealed that both treatments were able to almost completely protect most infected animals. They were also able to normalize the blood lymphocyte phenotype and to restore the responses of T and B cells to mitogens significantly. Treatment with GSH-loaded erythrocytes alone did not provide significant results for most parameters investigated, but a marked reduction in proviral DNA content was obtained in infected organs, including the brain. The results reported in this paper confirm the important role of macrophages in retroviral infection and moreover prove that erythrocytes, by selectively protecting these cells, strongly affect MAIDS progression. Furthermore, the combination of GSH- or GSH/AZTp2AZT-loaded erythrocytes with an oral nucleoside analogue (AZT) for the protection of lymphocytes provides additive responses in all the parameters investigated.


Subject(s)
Anti-HIV Agents/administration & dosage , Erythrocytes , Macrophages/virology , Murine Acquired Immunodeficiency Syndrome/drug therapy , Animals , Anti-HIV Agents/therapeutic use , Brain/drug effects , Brain/virology , CD4-CD8 Ratio/drug effects , DNA, Viral/analysis , Dideoxynucleotides , Disease Progression , Drug Carriers , Drug Therapy, Combination , Female , Glutathione/administration & dosage , Glutathione/pharmacology , Glutathione/therapeutic use , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Liver/drug effects , Liver/immunology , Liver/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/virology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mitogens/pharmacology , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/pathology , Murine Acquired Immunodeficiency Syndrome/virology , Thymine Nucleotides/administration & dosage , Thymine Nucleotides/pharmacology , Thymine Nucleotides/therapeutic use , Zidovudine/administration & dosage , Zidovudine/pharmacology , Zidovudine/therapeutic use
12.
AIDS Res Hum Retroviruses ; 15(4): 345-53, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10082118

ABSTRACT

Disseminated infection with Mycobacterium avium complex (MAC) remains the most common serious bacterial infection in patients with advanced AIDS. The organisms that make up this complex are found ubiquitously in the environment, yet rarely cause disseminated disease in nonimmunocompromised human patients; on the contrary, up to 50% of patients with AIDS may ultimately develop the pathology. Hence, therapeutic strategies able to inhibit HIV and Mycobacterium replication are needed. Because of the rapid plasma elimination and toxicity of the most commonly used drugs, daily multiple-drug therapies must often be continued throughout life, frequently causing major side effects and, as a consequence, poor patient compliance. Therefore, alternative strategies that reduce the toxicity of the drugs and allow prolonged application intervals are sorely needed. Since erythrocytes (RBCs) can behave as bioreactors able to convert impermeant prodrugs to membrane-releasable active drugs, new compounds (AZTpEMB, AZTpEMBpAZT, and AZTp2EMB) consisting of both an antiretroviral and an antimicrobial drug were designed and synthesized. Among these, only AZTp2EMB was hydrolyzed by erythrocyte enzymes and could be encapsulated inside RBCs. AZTp2EMB-loaded RBCs slowly released AZT and EMB in culture medium, reducing its concentration by one-half about every 48 hr of incubation at 37 degrees C. Moreover, when AZTp2EMB-loaded erythrocytes were incubated for 6 days in the presence of human macrophages infected with Mycobacterium avium (M. avium) a marked bactericidal effect (>1 log) was observed. Thus, AZTp2EMB-loaded erythrocytes could be used as endogenous bioreactors for AZT and EMB delivery in the treatment of HIV and M. avium infection.


Subject(s)
Anti-HIV Agents/metabolism , Antitubercular Agents/metabolism , Erythrocytes/metabolism , Ethambutol/metabolism , Zidovudine/metabolism , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Bioreactors , Cells, Cultured , Dimerization , Ethambutol/chemical synthesis , Ethambutol/pharmacology , Humans , Molecular Structure , Mycobacterium avium/drug effects , Zidovudine/chemical synthesis , Zidovudine/pharmacology
13.
FASEB J ; 13(2): 273-83, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9973315

ABSTRACT

CD38, a transmembrane glycoprotein widely expressed in vertebrate cells, is a bifunctional ectoenzyme catalyzing the synthesis and hydrolysis of cyclic ADP-ribose (cADPR). cADPR is a universal second messenger that releases calcium from intracellular stores. Since cADPR is generated by CD38 at the outer surface of many cells, where it acts intracellularly, increasing attention is paid to addressing this topological paradox. Recently, we demonstrated that CD38 is a catalytically active, unidirectional transmembrane transporter of cADPR, which then reaches its receptor-operated intracellular calcium stores. Moreover, CD38 was reported to undergo a selective and extensive internalization through non clathrin-coated endocytotic vesicles upon incubating CD38(+) cells with either NAD+ or thiol compounds: these endocytotic vesicles can convert cytosolic NAD into cADPR despite an asymmetric unfavorable orientation that makes the active site of CD38 intravesicular. Here we demonstrate that the cADPR-generating activity of the endocytotic vesicles results in remarkable and sustained increases of intracellular free calcium concentration in different cells exposed to either NAD+, or GSH, or N-acetylcysteine. This effect of CD38-internalizing ligands on intracellular calcium levels was found to involve a two-step mechanism: 1) influx of cytosolic NAD+ into the endocytotic vesicles, mediated by a hitherto unrecognized dinucleotide transport system that is saturable, bidirectional, inhibitable by 8-N3-NAD+, and characterized by poor dinucleotide specificity, low affinity, and high efficiency; 2) intravesicular CD38-catalyzed conversion of NAD+ to cADPR, followed by outpumping of the cyclic nucleotide into the cytosol and subsequent release of calcium from thapsigargin-sensitive stores. This unknown intracellular trafficking of NAD+ and cADPR based on two distinctive and specific transmembrane carriers for either nucleotide can affect the intracellular calcium homeostasis in CD38(+) cells.


Subject(s)
Antigens, CD , Antigens, Differentiation/metabolism , Calcium/metabolism , NAD+ Nucleosidase/metabolism , NAD/metabolism , Signal Transduction , 3T3 Cells , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/metabolism , Animals , Biological Transport , HeLa Cells , Humans , Jurkat Cells , Ligands , Membrane Glycoproteins , Mice , Second Messenger Systems
14.
FASEB J ; 12(14): 1507-20, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9806760

ABSTRACT

CD38 is a type II transmembrane glycoprotein expressed in many vertebrate cells. It is a bifunctional ectoenzyme that catalyzes both the synthesis of Cyclic ADP-ribose (cADPR) from NAD+ and the degradation of cADPR to ADP-ribose by means of its ADP-ribosyl cyclase and cADPR-hydrolase activities, respectively. The cyclase also converts NGD+ to cyclic GDP-ribose (cGDPR), which is refractory to cADPR-hydrolase. cADPR, but not cGDPR, is a potent calcium mobilizer from intracellular stores. It has been demonstrated to be a new second messenger involved in the regulation of calcium homeostasis in many cell types, from plants to mammals. The number of physiological processes shown to be regulated by cADPR is steadily increasing. A topological paradox exists because ectocellularly generated cADPR acts intracellularly. Here we demonstrate that the catalytic functioning of CD38 is accompanied by a cADPR (cGDPR) -transporting activity across natural and artificial membranes. In resealed membranes from CD38(+) human erythrocytes, transport of catalytically generated cADPR or cGDPR was saturation dependent and occurred against a concentration gradient. Likewise, CD38-reconstituted proteoliposomes were active in concentrating NAD+ (NGD+) -derived cADPR (cGDPR) inside the vesicle compartment. Moreover, the cADPR-transporting activity in CD38 proteoliposomes prevented the hydrolase-catalyzed degradation to ADPR that occurs conversely with detergent-solubilized CD38, resulting in selective influx of cADPR. In the CD38 proteoliposomes, catalytically active CD38 exhibited monomeric, dimeric, and tetrameric structures. In CD38 sense- but not in antisense-transfected HeLa cells, externally added NAD+ resulted in significant, transient increases in cytosolic calcium. These data suggest that transmembrane juxtaposition of two or four CD38 monomers can generate a catalytically active channel for selective formation and influx of cADPR (cGDPR) to reach cADPR-responsive intracellular calcium stores.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Antigens, CD , Antigens, Differentiation/metabolism , Membrane Glycoproteins/metabolism , NAD+ Nucleosidase/metabolism , Second Messenger Systems , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/metabolism , Biological Transport , Catalysis , Cyclic ADP-Ribose , Erythrocyte Membrane/metabolism , Guanosine Diphosphate Sugars/metabolism , HeLa Cells , Humans , NAD/metabolism , Proteolipids/metabolism
15.
Acta Crystallogr D Biol Crystallogr ; 54(Pt 4): 684-6, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9761875

ABSTRACT

The GDP-4-keto-6-deoxy-D-mannose epimerase/reductase (GM_ER) isolated from E. coli has been overexpressed as a GST-fusion protein and purified to homogeneity. The enzyme, an NADP+(H)-binding homodimer of 70 kDa, is responsible for the production of GDP-L-fucose. GM_ER shows significant structural homology to the human erythrocyte protein FX, which is involved in blood-group glycoconjugate biosynthesis, displaying 3,5 epimerase/reductase activity on GDP-4-keto-6-deoxy-D-mannose. GM_ER has been crystallized in a trigonal crystalline form, containing one molecule per asymmetric unit, suitable for high-resolution crystallographic investigations.


Subject(s)
Bacterial Proteins/chemistry , Carbohydrate Epimerases/chemistry , Escherichia coli Proteins , Escherichia coli/enzymology , Ketone Oxidoreductases , Multienzyme Complexes , Sugar Alcohol Dehydrogenases/chemistry , Amino Acid Sequence , Bacterial Proteins/isolation & purification , Carbohydrate Epimerases/isolation & purification , Crystallization , Crystallography, X-Ray , Humans , Molecular Sequence Data , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Sequence Alignment , Sequence Homology, Amino Acid , Sugar Alcohol Dehydrogenases/isolation & purification
16.
FEBS Lett ; 433(3): 275-8, 1998 Aug 21.
Article in English | MEDLINE | ID: mdl-9744810

ABSTRACT

CD38, a type II transmembrane glycoprotein, behaves as a catalytically active transporter responsible for ectocellular generation of cyclic ADP-ribose (cADPR) from NAD+ and for subsequent influx of cADPR across membranes [Franco, L., Guida, L., Bruzzone, S., Zocchi, E., Usai, C. and De Flora, A. (1998) FASEB J. in press]. cADPR regulates intracellular calcium homeostasis by releasing calcium from responsive stores. The cADPR-transporting function of CD38 requires channel-generating oligomeric forms of the protein rather than the 46 kDa monomers that have been described so far in CD38+ cells. Here we demonstrate that CD38, both in reconstituted proteoliposomes and in CD38-transfected HeLa cells, is a mixture of catalytically active monomers, homodimers and homotetramers. A soluble recombinant form of CD38 corresponding to its ectocellular region proved to be monomeric. Thus, association of native CD38 with either artificial or natural membranes seems to result in a reversible juxtaposition of monomers suitable to cADPR-transporting activity.


Subject(s)
Antigens, CD , Antigens, Differentiation/metabolism , NAD+ Nucleosidase/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Antigens, Differentiation/chemistry , Antigens, Differentiation/genetics , Aplysia/enzymology , Cell Membrane/metabolism , Dimerization , HeLa Cells , Humans , Liposomes , Macromolecular Substances , Membrane Glycoproteins , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , NAD/metabolism , NAD+ Nucleosidase/chemistry , NAD+ Nucleosidase/genetics , Proteolipids/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection
17.
FEBS Lett ; 429(3): 274-8, 1998 Jun 16.
Article in English | MEDLINE | ID: mdl-9662431

ABSTRACT

Leukocyte adhesion deficiency type II (LAD II) is a rare genetic disease characterized by severe immunodeficiency which is related to defective expression in leukocytes of sialyl-Lewis X (SLeX), a fucosylated ligand for endothelial selectins. The molecular basis of LAD II is still unknown, but has been tentatively localized in the de novo pathway of GDP-L-fucose biosynthesis from GDP-D-mannose. Here, we demonstrate that in cell lysates from a LAD II patient, GDP-D-mannose-4,6-dehydratase (GMD), the first of the two enzymes of the pathway has a defective activity compared to control subjects. GMD in cell lysates from both parents showed intermediate activity levels. Cloning of GMD from patient and control lymphocytes ruled out any mutation affecting the amino acid GMD sequence and the purified recombinant proteins from both controls and the patient showed identical specific activities. Since the levels of immunoreactive GMD in cell lysates were comparable in the patient and in controls, the biochemical deficiency of intracellular GMD activity in LAD II seems to be due to mutation(s) affecting some still unidentified GMD-regulating protein.


Subject(s)
Hydro-Lyases/metabolism , Leukocyte-Adhesion Deficiency Syndrome/enzymology , Cloning, Molecular , Guanosine Diphosphate Fucose/biosynthesis , Guanosine Diphosphate Mannose/metabolism , Humans , Hydro-Lyases/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , Male , Oligosaccharides/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Sialyl Lewis X Antigen
18.
J Biol Chem ; 273(14): 8017-24, 1998 Apr 03.
Article in English | MEDLINE | ID: mdl-9525901

ABSTRACT

CD38 is a bifunctional ectoenzyme, predominantly expressed on hematopoietic cells during differentiation, that catalyzes the synthesis (cyclase) and the degradation (hydrolase) of cyclic ADP-ribose (cADPR), a powerful calcium mobilizer from intracellular stores. Due to the well established role of calcium levels in the regulation of apoptosis, proliferation, and differentiation, the CD38/cADPR system seems to be a likely candidate involved in the control of these fundamental processes. The ectocellular localization of the cyclase activity, however, contrasts with the intracellular site of action of cADPR. Here we demonstrate that ectocellular expression of human CD38 in CD38(-) HeLa and 3T3 cells results in intracellular CD38 substrate (NAD+ + NADH) consumption and product (cADPR) accumulation. Furthermore, a causal relationship is established between presence of intracellular cADPR, partial depletion of thapsigargin-sensitive calcium stores, increase in basal free cytoplasmic calcium concentration, and decrease of cell doubling time. The significant shortening of the S phase in CD38(+) HeLa cells, as compared with controls, demonstrates an effect of intracellular cADPR on the mammalian cell cycle.


Subject(s)
Antigens, CD , Antigens, Differentiation/biosynthesis , Calcium/metabolism , Cell Cycle/physiology , NAD+ Nucleosidase/biosynthesis , 3T3 Cells , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Biological Transport , Cell Membrane Permeability , DNA/analysis , HeLa Cells , Humans , Membrane Glycoproteins , Mice
19.
AIDS Res Hum Retroviruses ; 14(5): 435-44, 1998 Mar 20.
Article in English | MEDLINE | ID: mdl-9546803

ABSTRACT

Human herpesvirus (HSVs) are distributed worldwide and are among the most frequent causes of viral infection in HIV-1-immunocompromised patients. Hence, therapeutic strategies able to inhibit HSV-1 and HIV-1 replication are sorely needed. Until now, the most common therapies against HSV-1 and HIV-1 infectivity have been based on the administration of nucleoside analogs; however, to be active, these antiviral drugs must be converted to their triphosphorylated derivatives by viral and/or cellular kinases. At the cellular level, the main problems involved in the use of such drugs are their limited phosphorylation in some cells (e.g., antiretroviral drugs in macrophages) and the cytotoxic side effects of nucleoside analog triphosphates. To overcome these limitations, a new heterodinucleotide (AZTp2ACV) consisting of both an antiretroviral and an antiherpetic drug, bound by a pyrophosphate bridge, was designed and synthesized. The impermeant AZTp2ACV was encapsulated into autologous erythrocytes modified to increase their recognition and phagocytosis by human macrophages. Once inside macrophages, metabolic activation of the drug occurred. The addition of AZTp2ACV-loaded erythrocytes to human macrophages provided effective and almost complete in vitro protection from HIV-1 and HSV-1 replications, respectively. Therefore, AZTp2ACV acts as an efficient antiviral prodrug following selective targeting to macrophages by means of loaded erythrocytes.


Subject(s)
Acyclovir/pharmacology , Antiviral Agents/pharmacology , Erythrocytes , HIV-1/drug effects , Herpesvirus 1, Human/drug effects , Macrophages/virology , Virus Replication/drug effects , Zidovudine/pharmacology , Acyclovir/administration & dosage , Acyclovir/metabolism , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/metabolism , Chlorocebus aethiops , Chromatography, High Pressure Liquid , Cytopathogenic Effect, Viral , Drug Combinations , Erythrocytes/metabolism , HIV-1/growth & development , Herpesvirus 1, Human/growth & development , Humans , Macrophages/physiology , Phagocytosis , Vero Cells , Zidovudine/administration & dosage , Zidovudine/metabolism
20.
J Acquir Immune Defic Syndr Hum Retrovirol ; 17(3): 189-95, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9495216

ABSTRACT

A new antiretroviral drug (azidothymidine homodinucleotide, AZTp2AZT), designed for the protection of macrophages against retroviral infection, was evaluated in a murine retrovirus-induced immunodeficiency model of AIDS (MAIDS) alone and in combination with oral azidothymidine (AZT). C57BL/6 mice were infected with the retroviral complex LP-BM5 and treated for 3 months by weekly administrations of 15 nmol of AZTp2AZT encapsulated into autologous erythrocytes for macrophage protection. AZTp2AZT treatment was found to reduce lymphoadenopathy (48%), splenomegaly (26%), and BM5d proviral DNA content in lymph nodes, spleen, and brain of 37%, 40%, and 36%, respectively, compared with untreated animals. AZT administration in drinking water (0.25 mg/ml) was more effective than administration of AZTp2AZT encapsulated into erythrocytes in reducing lymphoadenopathy, splenomegaly, gammaglobulinemia, and proviral DNA content in lymph nodes, but it caused a reduction in erythrocyte count and hematocrit levels. Although combined treatments do not provide additive responses in the several parameters investigated, they were found to be much more effective in reducing the proviral DNA content in brain (67%) than were monotherapies. Furthermore, no apparent signs of hematotoxicity were observed. Thus, macrophage delivery of antiviral drugs may contribute to brain protection from retroviral infections by mechanisms other than those exerted by oral AZT administration.


Subject(s)
Anti-HIV Agents/therapeutic use , Murine Acquired Immunodeficiency Syndrome/drug therapy , Prodrugs/therapeutic use , Thymine Nucleotides/therapeutic use , Zidovudine/therapeutic use , Animals , Brain/virology , DNA, Viral/analysis , Dideoxynucleotides , Drug Therapy, Combination , Female , Flow Cytometry , Immunoglobulin G/blood , Injections, Intraperitoneal , Lymph Nodes/virology , Lymphatic Diseases/drug therapy , Lymphocyte Activation/drug effects , Lymphocyte Count/drug effects , Macrophages/virology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/immunology , Murine Acquired Immunodeficiency Syndrome/virology , Prodrugs/administration & dosage , Prodrugs/pharmacology , Spleen/virology , Splenomegaly/drug therapy , Thymine Nucleotides/administration & dosage , Thymine Nucleotides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL