Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
FASEB J ; 34(9): 12565-12576, 2020 09.
Article in English | MEDLINE | ID: mdl-32717131

ABSTRACT

Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most potent Ca2+ mobilizing second messenger whose formation has remained elusive. In vitro, CD38-mediated NAADP synthesis requires an acidic pH and a nonphysiological concentration of nicotinic acid (NA). We discovered that CD38 catalyzes synthesis of NAADP by exchanging the nicotinamide moiety of nicotinamide adenine dinucleotide phosphate (NADP+ ) for the NA group of nicotinic acid adenine dinucleotide (NAAD) inside endolysosomes of interleukin 8 (IL8)-treated lymphokine-activated killer (LAK) cells. Upon IL8 stimulation, cytosolic NADP+ is transported to acidified endolysosomes via connexin 43 (Cx43) and gated by cAMP-EPAC-RAP1-PP2A signaling. CD38 then performs a base-exchange reaction with the donor NA group deriving from NAAD, produced by newly described endolysosomal activities of NA phosphoribosyltransferase (NAPRT) and NMN adenyltransferase (NMNAT) 3. Thus, the membrane organization of endolysosomal CD38, a signal-mediated transport system for NADP+ and luminal NAD+ biosynthetic enzymes integrate signals from a chemokine and cAMP to specify the spatiotemporal mobilization of Ca2+ to drive cell migration.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Calcium Signaling , Cell Movement , Interleukin-8/pharmacology , Killer Cells, Lymphokine-Activated/metabolism , Lysosomes/metabolism , Membrane Glycoproteins/metabolism , NADP/analogs & derivatives , Animals , Cells, Cultured , Killer Cells, Lymphokine-Activated/cytology , Mice , Mice, Inbred C57BL , NADP/metabolism
2.
FASEB J ; 33(3): 3404-3419, 2019 03.
Article in English | MEDLINE | ID: mdl-30452880

ABSTRACT

Reactive oxygen species (ROS) derived from NADPH oxidase (Nox) has been shown to activate ADP-ribosyl cyclase (ARC), which produces the Ca2+ mobilizing second messenger, cyclic ADP-ribose (cADPR). In the present study, we examined how ROS activates cluster of differentiation (CD)38, a mammalian prototype of ARC. CD38 exists in type II and III forms with opposing membrane orientation. This study showed the coexpression of type II and III CD38 in lymphokine-activated killer (LAK) cells. The catalytic site of the constitutively active type II CD38 faces the outside of the cell or the inside of early endosomes (EEs), whereas the basally inactive type III CD38 faces the cytosol. Type III CD38 interacted with Nox4/phosphorylated-p22phox (p-p22phox) in EEs of LAK cells upon IL-8 treatment. H2O2 derived from Nox4 activated type III CD38 by forming a disulfide bond between Cys164 and Cys177, resulting in increased cADPR formation. Our study identified the mechanism by which type III CD38 is activated in an immune cell (LAK), in which H2O2 generated by Nox4 oxidizes and activates type III CD38 to generate cADPR. These findings provide a novel model of cross-talk between ROS and Ca2+ signaling.-Park, D.-R., Nam, T.-S., Kim, Y.-W., Bae, Y. S., Kim, U.-H. Oxidative activation of type III CD38 by NADPH oxidase-derived hydrogen peroxide in Ca2+ signaling.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Hydrogen Peroxide/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Antigens, Differentiation/metabolism , Cell Line, Tumor , Cyclic ADP-Ribose/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress/physiology , Second Messenger Systems/physiology
3.
J Invest Dermatol ; 139(6): 1362-1372, 2019 06.
Article in English | MEDLINE | ID: mdl-30578820

ABSTRACT

UVB-induced skin damage is attributable to reactive oxygen species, which are triggered by intracellular Ca2+ signals. However, exactly how the reactive oxygen species are triggered by intracellular Ca2+ upon UVB irradiation remains obscure. Here, we show that UVB induces Ca2+ signals via sequential generation of the following Ca2+ messengers: inositol 1,4,5-trisphosphate, nicotinic acid adenine dinucleotide phosphate, and cyclic ADP-ribose. UVB induced H2O2 production through NADPH oxidase 4 activation, which is downstream to inositol 1,4,5-trisphosphate and nicotinic acid adenine dinucleotide phosphate. H2O2 derived from NADPH oxidase 4 activated CD38 to produce cyclic ADP-ribose. UVB first evoked the pannexin channel to release ATP, which acts on P2X7 receptor to generate inositol 1,4,5-trisphosphate. Inhibitors of these messengers, as well as antioxidants, blocked UVB-induced Ca2+ signals and IL-1ß secretion in keratinocytes. Furthermore, ablation of CD38 and NADPH oxidase 4 protected against UVB-induced inflammation and IL-1ß secretion in the murine epidermis. These results show that UVB induces IL-1ß secretion through cross-talk between Ca2+ and reactive oxygen species, providing insight towards potential targets against UVB-induced inflammation.


Subject(s)
Calcium Signaling/immunology , Epidermis/radiation effects , Interleukin-1beta/metabolism , Ultraviolet Rays/adverse effects , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Animals , Antioxidants/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/radiation effects , Carcinogenesis/immunology , Carcinogenesis/radiation effects , Cations, Divalent/metabolism , Cell Line , Epidermis/immunology , Epidermis/metabolism , Humans , Interleukin-1beta/immunology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Models, Animal , NADPH Oxidase 4/antagonists & inhibitors , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Photosensitivity Disorders/etiology , Photosensitivity Disorders/immunology , Primary Cell Culture , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Skin Aging/immunology , Skin Aging/radiation effects
4.
Cell Physiol Biochem ; 46(5): 2017-2030, 2018.
Article in English | MEDLINE | ID: mdl-29723871

ABSTRACT

BACKGROUND/AIMS: Cyclic ADP-ribose (cADPR) is a Ca2+ -mobilization messenger that acts on ryanodine-sensitive Ca2+ channels in the sarcoplasmic reticulum (SR) Ca2+ stores. Moreover, it has been proposed that cADPR serves an additional role in activating the sarcoendoplasmic reticulum Ca2+ -ATPase (SERCA) pump. The aim of this study was to determine the exact mechanism by which cADPR regulates SR Ca2+ stores in physiologically relevant systems. METHODS: We analyzed Ca2+ signals as well as the production of Ca2+ mobilizing messengers in the skeletal muscle cells of mice subjected to intensive exercise or in the SR fractions from skeletal muscle cells after ß-adrenergic receptor (ß-AR) stimulation. RESULTS: We show that cADPR enhances SERCA activity in skeletal muscle cells in response to ß-AR agonists, increasing SR Ca2+ uptake. We demonstrate that cADPR is generated by CD38, a cADPR-synthesizing enzyme, increasing muscle Ca2+ signals and contractile force during exercise. CD38 is upregulated by the cAMP response element-binding protein (CREB) transcription factor upon ß-AR stimuli and exercise. CD38 knockout (KO) mice show defects in their exercise and cADPR synthesis capabilities, lacking a ß-AR agonist-induced muscle contraction when compared to wild-type mice. The skeletal muscle of CD38 KO mice exhibits delayed cytosolic Ca2+ clearance and reduced SERCA activity upon exercise. CONCLUSION: These findings provide insight into the physiological adaptive mechanism by which the CD38- cADPR-SERCA signaling axis plays an essential role in muscle contraction under exercise, and define cADPR as an endogenous activator of SERCA in enhancing the SR Ca2+ load.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Adrenergic beta-Agonists/pharmacology , Cyclic ADP-Ribose/metabolism , Muscle Contraction/drug effects , Muscle, Skeletal/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction/drug effects , Animals , Cell Line , Male , Mice , Mice, Inbred C57BL , Receptors, Adrenergic, beta/metabolism
5.
BMC Neurosci ; 18(1): 35, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28340569

ABSTRACT

BACKGROUND: Recent rodent and human studies provide evidence in support of the fact that CD157, well known as bone marrow stromal cell antigen-1 (BST-1) and a risk factor in Parkinson's disease, also meaningfully acts in the brain as a neuroregulator and affects social behaviors. It has been shown that social behaviors are impaired in CD157 knockout mice without severe motor dysfunction and that CD157/BST1 gene single nucleotide polymorphisms are associated with autism spectrum disorder in humans. However, it is still necessary to determine how this molecule contributes to the brain's physiological and pathophysiological functions. METHODS: To gain fresh insights about the relationship between the presence of CD157 in the brain and its enzymatic activity, and aberrant social behavior, CD157 knockout mice of various ages were tested. RESULTS: CD157 immunoreactivity colocalized with nestin-positive cells and elements in the ventricular zones in E17 embryos. Brain CD157 mRNA levels were high in neonates but low in adults. Weak but distinct immunoreactivity was detected in several areas in the adult brain, including the amygdala. CD157 has little or no base exchange activity, but some ADP-ribosyl cyclase activity, indicating that CD157 formed cyclic ADP-ribose but much less nicotinic acid adenine dinucleotide phosphate, with both mobilizing Ca2+ from intracellular Ca2+ pools. Social avoidance in CD157 knockout mice was rescued by a single intraperitoneal injection of oxytocin. CONCLUSIONS: CD157 may play a role in the embryonic and adult nervous systems. The functional features of CD157 can be explained in part through the production of cyclic ADP-ribose rather than nicotinic acid adenine dinucleotide phosphate. Further experiments are required to elucidate how the embryonic expression of CD157 in neural stem cells contributes to behaviors in adults or to psychiatric symptoms.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Antigens, CD/metabolism , Brain/enzymology , Social Behavior , ADP-ribosyl Cyclase/genetics , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Animals , Animals, Newborn , Antigens, CD/genetics , Avoidance Learning/physiology , Brain/embryology , Brain/growth & development , Cyclic ADP-Ribose/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Models, Animal , NADP/analogs & derivatives , NADP/metabolism , Nestin/metabolism , RNA, Messenger/metabolism
6.
PLoS One ; 11(3): e0149125, 2016.
Article in English | MEDLINE | ID: mdl-26959359

ABSTRACT

Ca2+ signaling plays a fundamental role in cardiac hypertrophic remodeling, but the underlying mechanisms remain poorly understood. We investigated the role of Ca2+-mobilizing second messengers, NAADP and cADPR, in the cardiac hypertrophy induced by ß-adrenergic stimulation by isoproterenol. Isoproterenol induced an initial Ca2+ transients followed by sustained Ca2+ rises. Inhibition of the cADPR pathway with 8-Br-cADPR abolished only the sustained Ca2+ increase, whereas inhibition of the NAADP pathway with bafilomycin-A1 abolished both rapid and sustained phases of the isoproterenol-mediated signal, indicating that the Ca2+ signal is mediated by a sequential action of NAADP and cADPR. The sequential production of NAADP and cADPR was confirmed biochemically. The isoproterenol-mediated Ca2+ increase and cADPR production, but not NAADP production, were markedly reduced in cardiomyocytes obtained from CD38 knockout mice. CD38 knockout mice were rescued from chronic isoproterenol infusion-induced myocardial hypertrophy, interstitial fibrosis, and decrease in fractional shortening and ejection fraction. Thus, our findings indicate that ß-adrenergic stimulation contributes to the development of maladaptive cardiac hypertrophy via Ca2+ signaling mediated by NAADP-synthesizing enzyme and CD38 that produce NAADP and cADPR, respectively.


Subject(s)
Calcium Signaling/drug effects , Cardiomegaly/metabolism , Cyclic ADP-Ribose/pharmacology , NADP/analogs & derivatives , Receptors, Adrenergic, beta/metabolism , ADP-ribosyl Cyclase 1/metabolism , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/physiopathology , Isoproterenol , Male , Mice, Knockout , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NADP/pharmacology , Rats, Sprague-Dawley , Ultrasonography
7.
PLoS One ; 10(9): e0139110, 2015.
Article in English | MEDLINE | ID: mdl-26407101

ABSTRACT

Human sperm have to undergo a maturational process called capacitation in the female reproductive tract. Capacitation confers upon the sperm an ability to gain hypermotility and undergo acrosome reaction. Previous studies have suggested that seminal plasma proteins induce the capacitation of sperm in the female reproductive tract for the successful fertilization of the oocyte. However, the function of seminal plasma proteins in capacitation remains largely unclear. To the end, we found that soluble CD38 (sCD38) in seminal plasma increases the capacitation of sperm via specific interactions between sCD38 and the CD31 on the sperm. Upon the association of sCD38 with CD31, tyrosine kinase Src phosphorylates CD31, a process blocked by Src inhibitors. Shc, SHP-2, Grb2, and SOS, as well as Src kinase were found to associate with the phosphorylated CD31. The sCD38-induced phosphorylation of CD31 initiates a cascade reaction through the phosphorylation of Erk1/2, which results in the acrosome reaction, and sperm hypermotility. These processes were prevented by Src, Ras and MEK inhibitors. Taken together, these data indicate that the sCD38 present in seminal plasma plays a critical role in the capacitation of sperm.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Semen/metabolism , Sperm Capacitation , Acrosome Reaction , Humans , MAP Kinase Signaling System , Male , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Solubility , Sperm Motility
8.
J Biol Chem ; 289(23): 16362-73, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24759100

ABSTRACT

NAD glycohydrolases (NADases) catalyze the hydrolysis of NAD to ADP-ribose and nicotinamide. Although many members of the NADase family, including ADP-ribosyltransferases, have been cloned and characterized, the structure and function of NADases with pure hydrolytic activity remain to be elucidated. Here, we report the structural and functional characterization of a novel NADase from rabbit reticulocytes. The novel NADase is a glycosylated, glycosylphosphatidylinositol-anchored cell surface protein exclusively expressed in reticulocytes. shRNA-mediated knockdown of the NADase in bone marrow cells resulted in a reduction of erythroid colony formation and an increase in NAD level. Furthermore, treatment of bone marrow cells with NAD, nicotinamide, or nicotinamide riboside, which induce an increase in NAD content, resulted in a significant decrease in erythroid progenitors. These results indicate that the novel NADase may play a critical role in regulating erythropoiesis of hematopoietic stem cells by modulating intracellular NAD.


Subject(s)
Erythropoiesis , Hematopoietic Stem Cells/metabolism , NAD+ Nucleosidase/metabolism , NAD/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , DNA, Complementary , Glycosylation , HEK293 Cells , Humans , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis, Site-Directed , NAD+ Nucleosidase/chemistry , NAD+ Nucleosidase/genetics , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship
9.
Sci Signal ; 4(173): ra31, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21586728

ABSTRACT

Progesterone-induced calcium ion (Ca2+) signals in the neck region of sperm play a pivotal role in promoting sperm motility. Here, we show that a long-lasting Ca2+ signal required for sperm motility in response to progesterone depends on their pH-dependent fusion with prostasomes, which are small vesicles secreted by the prostate. We found that prostasome fusion led to the transfer of progesterone receptors, cyclic adenosine diphosphoribose (cADPR)-synthesizing enzymes, ryanodine receptors (RyRs), and other Ca2+ signaling tools from prostasomes to the sperm neck. Progesterone-induced sperm motility relied on cADPR-mediated Ca2+ mobilization through RyR located on acidic Ca2+ stores, followed by Ca2+ entry through store-operated channels. Treatment of prostasome-fused sperm with a cADPR antagonist or fusion with prostasomes in which type 2 RyR was depleted resulted in low fertilization rates, reduced sperm motility, or both. Thus, we conclude that sperm motility depends on the acquisition of Ca2+ signaling tools from prostasomes.


Subject(s)
Calcium Signaling , Progesterone/pharmacology , Prostate/metabolism , Sperm Motility/drug effects , Humans , Hydrogen-Ion Concentration , Male
10.
J Biol Chem ; 286(15): 12952-8, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21339289

ABSTRACT

CD38, a multifunctional enzyme that catalyzes the synthesis of intracellular Ca(2+) messengers, cyclic ADP-ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP), is known to be expressed on platelets. However, the role of CD38 in platelets remains unclear. Our present results show that treatment of platelets with thrombin results in a rapid and sustained Ca(2+) signal, resulting from a coordinated interplay of Ca(2+)-mobilizing messengers, inositol 1,4,5-trisphosphate, cADPR, and NAADP. By dissecting the signaling pathway using various agents, we delineated that cADPR and NAADP are sequentially produced through CD38 internalization by protein kinase C via myosin heavy chain IIA following phospholipase C activation in thrombin-induced platelets. An inositol 1,4,5-trisphosphate receptor antagonist blocked the thrombin-induced formation of cADPR and NAADP as well as Ca(2+) signals. An indispensable response of platelets relying on cytosolic calcium is the surface exposure of phosphatidylserine (PS), which implicates platelet procoagulant activity. Scrutinizing this parameter reveals that CD38(+/+) platelets fully express PS on the surface when stimulated with thrombin, whereas this response was decreased on CD38(-/-) platelets. Similarly, PS exposure and Ca(2+) signals were attenuated when platelets were incubated with 8-bromo-cADPR, bafilomycin A1, and a PKC inhibitor. Furthermore, in vivo, CD38-deficient mice exhibited longer bleeding times and unstable formation of thrombus than wild type mice. These results demonstrate that CD38 plays an essential role in thrombin-induced procoagulant activity of platelets and hemostasis via Ca(2+) signaling mediated by its products, cADPR and NAADP.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Blood Platelets/enzymology , Calcium Signaling/physiology , Hemostasis/physiology , Membrane Glycoproteins/metabolism , Thrombin/metabolism , ADP-ribosyl Cyclase 1/genetics , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cyclic ADP-Ribose/genetics , Cyclic ADP-Ribose/metabolism , Enzyme Inhibitors/pharmacology , Hemostasis/drug effects , Hemostatics/metabolism , Hemostatics/pharmacology , Inositol 1,4,5-Trisphosphate/genetics , Inositol 1,4,5-Trisphosphate/metabolism , Macrolides/pharmacology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , NADP/analogs & derivatives , NADP/genetics , NADP/metabolism , Phosphatidylserines/metabolism , Phosphatidylserines/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Thrombin/pharmacology
11.
J Biol Chem ; 285(28): 21877-87, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20442403

ABSTRACT

We have previously demonstrated that cyclic ADP-ribose (cADPR) is a calcium signaling messenger in interleukin 8 (IL-8)-induced lymphokine-activated killer (LAK) cells. In this study we examined the possibility that IL-8 activates CD38 to produce another messenger, nicotinic acid adenine dinucleotide phosphate (NAADP), in LAK cells, and we showed that IL-8 induced NAADP formation after cADPR production. These calcium signaling messengers were not produced when LAK cells prepared from CD38 knock-out mice were treated with IL-8, indicating that the synthesis of both NAADP and cADPR is catalyzed by CD38 in LAK cells. Application of cADPR to LAK cells induced NAADP production, whereas NAADP failed to increase intracellular cADPR levels, confirming that the production of cADPR precedes that of NAADP in IL-8-treated LAK cells. Moreover, NAADP increased intracellular Ca(2+) signaling as well as cell migration, which was completely blocked by bafilomycin A1, suggesting that NAADP is generated in lysosome-related organelles after cADPR production. IL-8 or exogenous cADPR, but not NAADP, increased intracellular cAMP levels. cGMP analog, 8-(4-chlorophenylthio)-guanosine 3',5'-cyclic monophosphate, increased both cADPR and NAADP production, whereas the cAMP analog, 8-(4-chlorophenylthio)-cAMP, increased only NAADP production, suggesting that cAMP is essential for IL-8-induced NAADP formation. Furthermore, activation of Rap1, a downstream molecule of Epac, was required for IL-8-induced NAADP formation in LAK cells. Taken together, our data suggest that IL-8-induced NAADP production is mediated by CD38 activation through the actions of cAMP/Epac/protein kinase A/Rap1 in LAK cells and that NAADP plays a key role in Ca(2+) signaling of IL-8-induced LAK cell migration.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Calcium Signaling , Cyclic ADP-Ribose/metabolism , Interleukin-8/metabolism , Killer Cells, Lymphokine-Activated/cytology , NADP/analogs & derivatives , Animals , Calcium/metabolism , Cell Movement , Humans , Mice , Mice, Transgenic , NADP/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , rap1 GTP-Binding Proteins/metabolism
12.
J Biol Chem ; 282(8): 5653-60, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17182620

ABSTRACT

Activation of CD38 in lymphokine-activated killer (LAK) cells involves interleukin-8 (IL8)-mediated protein kinase G (PKG) activation and results in an increase in the sustained intracellular Ca(2+) concentration ([Ca(2+)](i)), cADP-ribose, and LAK cell migration. However, direct phosphorylation or activation of CD38 by PKG has not been observed in vitro. In this study, we examined the molecular mechanism of PKG-mediated activation of CD38. Nonmuscle myosin heavy chain IIA (MHCIIA) was identified as a CD38-associated protein upon IL8 stimulation. The IL8-induced association of MHCIIA with CD38 was dependent on PKG-mediated phosphorylation of MHCIIA. Supporting these observations, IL8- or cell-permeable cGMP analog-induced formation of cADP-ribose, increase in [Ca(2+)](i), and migration of LAK cells were inhibited by treatment with the MHCIIA inhibitor blebbistatin. Binding studies using purified proteins revealed that the association of MHCIIA with CD38 occurred through Lck, a tyrosine kinase. Moreover, these three molecules co-immunoprecipitated upon IL8 stimulation of LAK cells. IL8 treatment of LAK cells resulted in internalization of CD38, which co-localized with MHCIIA and Lck, and blebbistatin blocked internalization of CD38. These findings demonstrate that the association of phospho-MHCIIA with Lck and CD38 is a critical step in the internalization and activation of CD38.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Calcium Signaling/physiology , Killer Cells, Lymphokine-Activated/enzymology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Nonmuscle Myosin Type IIA/metabolism , Protein Processing, Post-Translational/physiology , Calcium/metabolism , Calcium Signaling/drug effects , Cyclic ADP-Ribose/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Interleukin-8/pharmacology , Jurkat Cells , Killer Cells, Lymphokine-Activated/cytology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/physiology , Protein Processing, Post-Translational/drug effects
13.
Exp Mol Med ; 38(6): 718-26, 2006 Dec 31.
Article in English | MEDLINE | ID: mdl-17202848

ABSTRACT

ADP-ribosyl cyclase (ADPR-cyclase) produces a Ca2+-mobilizing second messenger, cyclic ADP- ribose (cADPR), from beta-NAD+. A prototype of mammalian ADPR-cyclases is a lymphocyte antigen CD38. Accumulating evidence indicates that ADPR-cyclases other than CD38 are expressed in various cells and organs. In this study, we discovered a small molecule inhibitor of kidney ADPR-cyclase. This compound inhibited kidney ADPR-cyclase activity but not CD38, spleen, heart or brain ADPR-cyclase activity in vitro. Characterization of the compound in a cell-based system revealed that an extracellular calcium-sensing receptor (CaSR)- mediated cADPR production and a later long-lasting increase in intracellular Ca2+ concentration ([Ca2+]i) in mouse mesangial cells were inhibited by the pre-treatment with this compound. In contrast, the compound did not block CD3/TCR-induced cADPR production and the increase of [Ca2+]i in Jurkat T cells, which express CD38 exclusively. The long-lasting Ca2+ signal generated by both receptors was inhibited by pre-treatment with an antagonistic cADPR derivative, 8-Br-cADPR, indicating that the Ca2+ signal is mediated by the ADPR-cyclase metabolite, cADPR. Moreover, among structurally similar compounds tested, the compound inhibited most potently the cADPR production and Ca2+ signal induced by CaSR. These findings provide evidence for existence of a distinct ADPR-cyclase in the kidney and basis for the development of tissue specific inhibitors.


Subject(s)
ADP-ribosyl Cyclase/antagonists & inhibitors , ADP-ribosyl Cyclase/metabolism , Azo Compounds/pharmacology , Calcium Signaling , Cyclic ADP-Ribose/metabolism , Enzyme Inhibitors/pharmacology , Kidney/enzymology , Animals , Azo Compounds/chemistry , Cell Line , Enzyme Inhibitors/chemistry , Humans , Mice , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/metabolism
14.
Biochem Biophys Res Commun ; 330(4): 1290-8, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15823583

ABSTRACT

ADP-ribosyl cyclase (ADPR-cyclase) produces a Ca(2+)-mobilizing second messenger cyclic ADP-ribose (cADPR) from beta-NAD(+). In this study, we examined the molecular basis of which beta-adrenergic receptor (betaAR) stimulation induces cADPR formation and characterized cardiac ADPR-cyclase. The results revealed that isoproterenol-mediated increase of [Ca(2+)](i) in rat cardiomyocytes was blocked by pretreatment with a cADPR antagonistic derivative 8-Br-cADPR, a PKA inhibitor H89 or high concentration of ryanodine. Moreover, incubation of ventricular lysates with isoproterenol, forskolin or cAMP resulted in activation of ADPR-cyclase that was inhibited by pretreatment with H89. Supporting the observations, the cADPR antagonist and H89 blocked 8-CPT-cAMP, a cell-permeant cAMP analog-induced increase in [Ca(2+)](i) but not cGMP-mediated increase. Characterization of partially purified cardiac ADPR-cyclase showed a molecular mass of approximately 42 kDa and no cross-activity with CD38 antibodies, and the enzyme activity was inhibited by Zn(2+) but not dithiothreitol. Microinjection of the enzyme into rat cardiomyocytes increased the level of [Ca(2+)](i) in a concentration-dependent manner. The enzyme-mediated increase of [Ca(2+)](i) was blocked by the cADPR antagonist. These findings suggest that betaAR-mediated regulation of [Ca(2+)](i) in rat cardiomyocytes is primed by activation of cardiac ADPR-cyclase via cAMP/PKA signaling and that cardiac ADPR-cyclase differs from CD38 in biochemical and immunological properties.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Calcium/metabolism , Cyclic AMP/physiology , Dithiothreitol/pharmacology , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta/physiology , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic GMP/physiology , Enzyme Activation , Rats , Rats, Sprague-Dawley , Signal Transduction , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL