Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 329: 113252, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32087251

RESUMEN

SARM1 is the central executioner of pathological axon degeneration, promoting axonal demise in response to axotomy, traumatic brain injury, and neurotoxic chemotherapeutics that induce peripheral neuropathy. SARM1 is an injury-activated NAD+ cleavage enzyme, and this NADase activity is required for the pro-degenerative function of SARM1. At present, SARM1 function is assayed by either analysis of axonal loss, which is far downstream of SARM1 enzymatic activity, or via NAD+ levels, which are regulated by many competing pathways. Here we explored the utility of measuring cADPR, a product of SARM1-dependent cleavage of NAD+, as an in cell and in vivo biomarker of SARM1 enzymatic activity. We find that SARM1 is a major producer of cADPR in cultured dorsal root ganglion (DRG) neurons, sciatic nerve, and brain, demonstrating that SARM1 has basal activity in the absence of injury. Following injury, there is a dramatic SARM1-dependent increase in the levels of axonal cADPR that precedes morphological axon degeneration. In vivo, there is also a rapid and large injury-stimulated increase in cADPR in sciatic and optic nerves. The increase in cADPR after injury is proportional to SARM1 gene dosage, suggesting that SARM1 activity is the prime regulator of cADPR levels. The role of cADPR as an important calcium mobilizing agent prompted exploration of its functional contribution to axon degeneration. We used multiple bacterial and mammalian engineered enzymes to manipulate cADPR levels in neurons but found no changes in the time course of axonal degeneration, suggesting that cADPR is unlikely to be an important contributor to the degenerative mechanism. Using cADPR as a SARM1 biomarker, we find that SARM1 can be partially activated by a diverse array of mitochondrial toxins administered at doses that do not induce axon degeneration. Hence, the subcritical activation of SARM1 induced by mitochondrial dysfunction may contribute to the axonal vulnerability common to many neurodegenerative diseases. Finally, we assay levels of both nerve cADPR and plasma neurofilament light chain (NfL) following nerve injury in vivo, and demonstrate that both biomarkers are excellent readouts of SARM1 activity, with cADPR reporting the early molecular changes in the nerve and NfL reporting subsequent axonal breakdown. The identification and characterization of cADPR as a SARM1 biomarker will help identify neurodegenerative diseases in which SARM1 contributes to axonal loss and expedite target validation studies of SARM1-directed therapeutics.


Asunto(s)
Proteínas del Dominio Armadillo/metabolismo , Axones/metabolismo , ADP-Ribosa Cíclica/metabolismo , Proteínas del Citoesqueleto/metabolismo , Dosificación de Gen/fisiología , Degeneración Nerviosa/metabolismo , Animales , Proteínas del Dominio Armadillo/genética , Axones/patología , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , ADP-Ribosa Cíclica/genética , Proteínas del Citoesqueleto/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Nervio Ciático/metabolismo , Nervio Ciático/patología
2.
Biomed Res Int ; 2015: 965271, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26146638

RESUMEN

Angiopoietins are vascular factors essential for blood vessel assembly and correct organization and maturation. This study describes a novel calcium-dependent machinery activated through Angiopoietin-1/2-Tie receptor system in HUVECs monolayer. Both cytokines were found to elicit intracellular calcium mobilization. Targeting intracellular Ca(2+) signaling, antagonizing IP3 with 2-APB or cADPR with 8Br-cADPR, was found to modulate in vitro angiogenic responses to Angiopoietins in a specific way. 2-APB and 8Br-cADPR impaired the phosphorylation of AKT and FAK induced by Ang-1 and Ang-2. On the other hand, phosphorylation of ERK1/2 and p38, as well as cell proliferation, was not affected by either inhibitor. The ability of ECs to migrate following Angs stimulation, evaluated by "scratch assay," was reduced by either 2-APB or 8Br-cADPR following Ang-2 stimulation and only slightly affected by 2-APB in cells stimulated with Ang-1. These results identify a novel calcium-dependent machinery involved in the complex interplay regulating angiogenic processes showing that IP3- and cADPR-induced Ca(2+) release specifically regulates distinct Angs-mediated angiogenic steps.


Asunto(s)
Angiotensina II/metabolismo , Señalización del Calcio/genética , ADP-Ribosa Cíclica/genética , Neovascularización Fisiológica/genética , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Angiotensina II/genética , Calcio/metabolismo , Proliferación Celular/genética , ADP-Ribosa Cíclica/biosíntesis , Células Endoteliales de la Vena Umbilical Humana , Humanos , Receptor TIE-2/genética , Receptor TIE-2/metabolismo
3.
Biochem Soc Trans ; 43(3): 405-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26009183

RESUMEN

Group I metabotropic glutamate receptors (I-mGluRs) modulate numerous cellular functions such as specific membrane currents and neurotransmitter release linked to their ability to mobilize calcium from intracellular calcium stores. As such, most I-mGluR research to date has focused on the coupling of these receptors to phospholipase C (PLC)-dependent and inositol (1,4,5) trisphosphate (IP3)-mediated calcium release via activation of IP3 receptors located upon the sarco/endoplasmic reticulum. However, there are now numerous examples of PLC- and IP3-independent I-mGluR-evoked signals, which may instead be mediated by activation of ryanodine receptors (RyRs). A prime candidate for mediating this coupling between I-mGluR activation and RyR opening is cyclic ADP ribose (cADPR) and, indeed, several of these PLC-/IP3-independent I-mGluR-evoked calcium signals have now been shown to be mediated wholly or partly by cADPR-evoked activation of RyRs. The contribution of cADPR signalling to I-mGluR-mediated responses is relatively complex, dependent as it is on factors such as cell type, excitation state of the cell and location of I-mGluRs on the cell. However, these factors notwithstanding, I-mGluR-mediated cADPR signalling remains poorly characterized, with several key aspects yet to be fully elucidated such as (1) the range of stimuli which evoke cADPR production, (2) the specific molecular mechanism(s) coupling cADPR to RyR activation and (3) the contribution of cADPR-mediated responses to downstream outputs such as synaptic plasticity. Furthermore, it is possible that the cADPR pathway may play a role in diseases underpinned by dysregulated calcium homoeostasis such as Alzheimer's disease (AD).


Asunto(s)
Enfermedad de Alzheimer/genética , Señalización del Calcio/genética , ADP-Ribosa Cíclica/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Calcio/metabolismo , ADP-Ribosa Cíclica/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Glutamato Metabotrópico/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
4.
J Biol Chem ; 288(38): 27128-27137, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23940051

RESUMEN

Aberrant Ca(2+) signals within pancreatic acinar cells are an early and critical feature in acute pancreatitis, yet it is unclear how these signals are generated. An important mediator of the aberrant Ca(2+) signals due to bile acid exposure is the intracellular Ca(2+) channel ryanodine receptor. One putative activator of the ryanodine receptor is the nucleotide second messenger cyclic ADP-ribose (cADPR), which is generated by an ectoenzyme ADP-ribosyl cyclase, CD38. In this study, we examined the role of CD38 and cADPR in acinar cell Ca(2+) signals and acinar injury due to bile acids using pharmacologic inhibitors of CD38 and cADPR as well as mice deficient in Cd38 (Cd38(-/-)). Cytosolic Ca(2+) signals were imaged using live time-lapse confocal microscopy in freshly isolated mouse acinar cells during perifusion with the bile acid taurolithocholic acid 3-sulfate (TLCS; 500 µM). To focus on intracellular Ca(2+) release and to specifically exclude Ca(2+) influx, cells were perifused in Ca(2+)-free medium. Cell injury was assessed by lactate dehydrogenase leakage and propidium iodide uptake. Pretreatment with either nicotinamide (20 mM) or the cADPR antagonist 8-Br-cADPR (30 µM) abrogated TLCS-induced Ca(2+) signals and cell injury. TLCS-induced Ca(2+) release and cell injury were reduced by 30 and 95%, respectively, in Cd38-deficient acinar cells compared with wild-type cells (p < 0.05). Cd38-deficient mice were protected against a model of bile acid infusion pancreatitis. In summary, these data indicate that CD38-cADPR mediates bile acid-induced pancreatitis and acinar cell injury through aberrant intracellular Ca(2+) signaling.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Células Acinares/metabolismo , Ácidos y Sales Biliares/toxicidad , Señalización del Calcio/efectos de los fármacos , ADP-Ribosa Cíclica/metabolismo , Glicoproteínas de Membrana/metabolismo , Pancreatitis/metabolismo , ADP-Ribosil Ciclasa 1/genética , Células Acinares/patología , Animales , Calcio/metabolismo , Señalización del Calcio/genética , ADP-Ribosa Cíclica/genética , Humanos , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Pancreatitis/inducido químicamente , Pancreatitis/genética , Pancreatitis/patología
5.
Vet Anaesth Analg ; 40(5): 512-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23565906

RESUMEN

OBJECTIVE: To investigate the CD38/cADPR signaling pathway as possible underlying mechanism of the effects of medetomidine on insulin and glucose homeostasis. ANIMALS: Thirty-two C57BL/6 mice of both sexes. METHODS: Wild-type (WT) and CD38-knockout (CD38(-/-) ) mice received medetomidine (50 µg kg(-1) ) or a similar volume of 0.9% NaCl (control) by intraperitoneal (IP) injection (each group n = 8). The mice were euthanized 45 minutes later with sodium pentobarbital IP and blood was sampled via cardiac puncture. Insulin and glucose concentrations were measured by radioimmunoassay and by the oxygen rate method, respectively. Data were analyzed with anova and Bonferroni post hoc (5% significance) and are shown as mean ± SD. RESULTS: Plasma insulin and glucose concentrations were similar between WT and CD38(-/-) mice under control conditions. As compared to controls, medetomidine administration produced a statistically significant decrease in plasma insulin concentrations in the WT mice whereas the decrease in the CD38(-/-) mice was not statistically significant. Correspondingly, medetomidine caused a significantly greater increase in plasma glucose concentrations in the WT than in the CD38(-/-) mice. CONCLUSION: The CD38/cADPR signaling pathway may be one underlying mechanism of the glucose and insulin effects of the alpha-2 adrenergic receptor agonist medetomidine and likely other drugs of its class.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , ADP-Ribosa Cíclica/metabolismo , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Insulina/metabolismo , Medetomidina/farmacología , Glicoproteínas de Membrana/metabolismo , ADP-Ribosil Ciclasa 1/genética , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Animales , ADP-Ribosa Cíclica/genética , Femenino , Regulación de la Expresión Génica/fisiología , Homeostasis/fisiología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
6.
J Biol Chem ; 287(42): 35599-35611, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22908234

RESUMEN

Cyclic adenosine diphosphoribose (cADPR) is an endogenous Ca(2+) mobilizing messenger that is formed by ADP-ribosyl cyclases from nicotinamide adenine dinucleotide (NAD). The main ADP-ribosyl cyclase in mammals is CD38, a multi-functional enzyme and a type II membrane protein. Here we explored the role of CD38-cADPR-Ca(2+) in the cardiomyogenesis of mouse embryonic stem (ES) cells. We found that the mouse ES cells are responsive to cADPR and possess the key components of the cADPR signaling pathway. In vitro cardiomyocyte (CM) differentiation of mouse ES cells was initiated by embryoid body (EB) formation. Interestingly, beating cells appeared earlier and were more abundant in CD38 knockdown EBs than in control EBs. Real-time RT-PCR and Western blot analyses further showed that the expression of several cardiac markers, including GATA4, MEF2C, NKX2.5, and α-MLC, were increased markedly in CD38 knockdown EBs than those in control EBs. Similarly, FACS analysis showed that more cardiac Troponin T-positive CMs existed in CD38 knockdown or 8-Br-cADPR, a cADPR antagonist, treated EBs compared with that in control EBs. On the other hand, overexpression of CD38 in mouse ES cells significantly inhibited CM differentiation. Moreover, CD38 knockdown ES cell-derived CMs possess the functional properties characteristic of normal ES cell-derived CMs. Last, we showed that the CD38-cADPR pathway negatively modulated the FGF4-Erks1/2 cascade during CM differentiation of ES cells, and transiently inhibition of Erk1/2 blocked the enhanced effects of CD38 knockdown on the differentiation of CM from ES cells. Taken together, our data indicate that the CD38-cADPR-Ca(2+) signaling pathway antagonizes the CM differentiation of mouse ES cells.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Señalización del Calcio/fisiología , Diferenciación Celular/fisiología , ADP-Ribosa Cíclica/metabolismo , Células Madre Embrionarias/enzimología , Glicoproteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/enzimología , ADP-Ribosil Ciclasa 1/genética , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Línea Celular , ADP-Ribosa Cíclica/genética , Cuerpos Embrioides/citología , Cuerpos Embrioides/enzimología , Células Madre Embrionarias/citología , Factor 4 de Crecimiento de Fibroblastos/biosíntesis , Factor 4 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Glicoproteínas de Membrana/genética , Ratones , Proteínas Musculares/genética , Miocitos Cardíacos/citología
7.
J Biol Chem ; 287(37): 31003-14, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22829588

RESUMEN

Extracellular ß-nicotinamide adenine dinucleotide (NAD(+)) is anti-inflammatory. We hypothesized that NAD(+) would modulate the anti-inflammatory cytokine Transforming Growth Factor (TGF)-ß1. Indeed, NAD(+) led to increases in both active and latent cell-associated TGF-ß1 in RAW 264.7 mouse macrophages as well as in primary peritoneal macrophages isolated from both C3H/HeJ (TLR4-mutant) and C3H/HeOuJ (wild-type controls for C3H/HeJ) mice. NAD(+) acts partially via cyclic ADP-ribose (cADPR) and subsequent release of Ca(2+). Treatment of macrophages with the cADPR analog 3-deaza-cADPR or Ca(2+) ionophores recapitulated the effects of NAD(+) on TGF-ß1, whereas the cADPR antagonist 8-Br-cADPR, Ca(2+) chelation, and antagonism of L-type Ca(2+) channels suppressed these effects. The time and dose effects of NAD(+) on TGF-ß1 were complex and could be modeled both statistically and mathematically. Model-predicted levels of TGF-ß1 protein and mRNA were largely confirmed experimentally but also suggested the presence of other mechanisms of regulation of TGF-ß1 by NAD(+). Thus, in vitro and in silico evidence points to NAD(+) as a novel modulator of TGF-ß1.


Asunto(s)
ADP-Ribosa Cíclica/metabolismo , Macrófagos/metabolismo , Modelos Biológicos , NAD/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Calcio/metabolismo , Ionóforos de Calcio/farmacología , Línea Celular , ADP-Ribosa Cíclica/análogos & derivados , ADP-Ribosa Cíclica/genética , ADP-Ribosa Cíclica/farmacología , Macrófagos/citología , Ratones , Ratones Mutantes , NAD/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Crecimiento Transformador beta1/genética
8.
J Biol Chem ; 287(25): 21067-81, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22547068

RESUMEN

Intracellular NAD(+) levels ([NAD(+)](i)) are important in regulating human T lymphocyte survival, cytokine secretion, and the capacity to respond to antigenic stimuli. NAD(+)-derived Ca(2+)-mobilizing second messengers, produced by CD38, play a pivotal role in T cell activation. Here we demonstrate that [NAD(+)](i) modifications in T lymphocytes affect intracellular Ca(2+) homeostasis both in terms of mitogen-induced [Ca(2+)](i) increase and of endoplasmic reticulum Ca(2+) store replenishment. Lowering [NAD(+)](i) by FK866-mediated nicotinamide phosphoribosyltransferase inhibition decreased the mitogen-induced [Ca(2+)](i) rise in Jurkat cells and in activated T lymphocytes. Accordingly, the Ca(2+) content of thapsigargin-sensitive Ca(2+) stores was greatly reduced in these cells in the presence of FK866. When NAD(+) levels were increased by supplementing peripheral blood lymphocytes with the NAD(+) precursors nicotinamide, nicotinic acid, or nicotinamide mononucleotide, the Ca(2+) content of thapsigargin-sensitive Ca(2+) stores as well as cell responsiveness to mitogens in terms of [Ca(2+)](i) elevation were up-regulated. The use of specific siRNA showed that the changes of Ca(2+) homeostasis induced by NAD(+) precursors are mediated by CD38 and the consequent ADPR-mediated TRPM2 gating. Finally, the presence of NAD(+) precursors up-regulated important T cell functions, such as proliferation and IL-2 release in response to mitogens.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , ADP-Ribosa Cíclica/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Mitógenos/farmacología , NAD/metabolismo , Linfocitos T/metabolismo , Canales Catiónicos TRPM/metabolismo , Acrilamidas/farmacología , Señalización del Calcio/fisiología , Proliferación Celular/efectos de los fármacos , ADP-Ribosa Cíclica/genética , Citocinas/antagonistas & inhibidores , Citocinas/genética , Citocinas/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Interleucina-2/genética , Interleucina-2/metabolismo , Activación del Canal Iónico/fisiología , Células Jurkat , NAD/genética , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/genética , Nicotinamida Fosforribosiltransferasa/metabolismo , Piperidinas/farmacología , Linfocitos T/citología , Canales Catiónicos TRPM/genética , Tapsigargina/farmacología
9.
J Biol Chem ; 287(18): 14502-14, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22396532

RESUMEN

Phagocytosis is a crucial event in the immune system that allows cells to engulf and eliminate pathogens. This is mediated through the action of immunoglobulin (IgG)-opsonized microbes acting on Fcγ receptors (FcγR) on macrophages, which results in sustained levels of intracellular Ca(2+) through the mobilization of Ca(2+) second messengers. It is known that the ADP-ribosyl cyclase is responsible for the rise in Ca(2+) levels after FcγR activation. However, it is unclear whether and how CD38 is involved in FcγR-mediated phagocytosis. Here we show that CD38 is recruited to the forming phagosomes during phagocytosis of IgG-opsonized particles and produces cyclic-ADP-ribose, which acts on ER Ca(2+) stores, thus allowing an increase in FcγR activation-mediated phagocytosis. Ca(2+) data show that pretreatment of J774A.1 macrophages with 8-bromo-cADPR, ryanodine, blebbistatin, and various store-operated Ca(2+) inhibitors prevented the long-lasting Ca(2+) signal, which significantly reduced the number of ingested opsonized particles. Ex vivo data with macrophages extracted from CD38(-/-) mice also shows a reduced Ca(2+) signaling and phagocytic index. Furthermore, a significantly reduced phagocytic index of Mycobacterium bovis BCG was shown in macrophages from CD38(-/-) mice in vivo. This study suggests a crucial role of CD38 in FcγR-mediated phagocytosis through its recruitment to the phagosome and mobilization of cADPR-induced intracellular Ca(2+) and store-operated extracellular Ca(2+) influx.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Macrófagos Peritoneales/metabolismo , Glicoproteínas de Membrana/metabolismo , Fagocitosis/fisiología , Receptores de IgG/metabolismo , ADP-Ribosil Ciclasa 1/genética , Animales , Línea Celular , ADP-Ribosa Cíclica/genética , ADP-Ribosa Cíclica/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Mycobacterium bovis/metabolismo , Receptores de IgG/genética
10.
J Biol Chem ; 286(52): 44480-90, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22033928

RESUMEN

The ADP-ribosyl cyclase CD38 whose catalytic domain resides in outside of the cell surface produces the second messenger cyclic ADP-ribose (cADPR) from NAD(+). cADPR increases intracellular Ca(2+) through the intracellular ryanodine receptor/Ca(2+) release channel (RyR). It has been known that intracellular NAD(+) approaches ecto-CD38 via its export by connexin (Cx43) hemichannels, a component of gap junctions. However, it is unclear how cADPR extracellularly generated by ecto-CD38 approaches intracellular RyR although CD38 itself or nucleoside transporter has been proposed to import cADPR. Moreover, it has been unknown what physiological stimulation can trigger Cx43-mediated export of NAD(+). Here we demonstrate that Cx43 hemichannels, but not CD38, import cADPR to increase intracellular calcium through RyR. We also demonstrate that physiological stimulation such as Fcγ receptor (FcγR) ligation induces calcium mobilization through three sequential steps, Cx43-mediated NAD(+) export, CD38-mediated generation of cADPR and Cx43-mediated cADPR import in J774 cells. Protein kinase A (PKA) activation also induced calcium mobilization in the same way as FcγR stimulation. FcγR stimulation-induced calcium mobilization was blocked by PKA inhibition, indicating that PKA is a linker between FcγR stimulation and NAD(+)/cADPR transport. Cx43 knockdown blocked extracellular cADPR import and extracellular cADPR-induced calcium mobilization in J774 cells. Cx43 overexpression in Cx43-negative cells conferred extracellular cADPR-induced calcium mobilization by the mediation of cADPR import. Our data suggest that Cx43 has a dual function exporting NAD(+) and importing cADPR into the cell to activate intracellular calcium mobilization.


Asunto(s)
Calcio/metabolismo , Conexina 43/metabolismo , ADP-Ribosa Cíclica/metabolismo , NAD/metabolismo , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Transporte Biológico Activo/fisiología , Conexina 43/genética , ADP-Ribosa Cíclica/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células HeLa , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , NAD/genética , Receptores de IgG/genética , Receptores de IgG/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
11.
J Biol Chem ; 286(15): 12952-8, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21339289

RESUMEN

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.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Plaquetas/enzimología , Señalización del Calcio/fisiología , Hemostasis/fisiología , Glicoproteínas de Membrana/metabolismo , Trombina/metabolismo , ADP-Ribosil Ciclasa 1/genética , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , ADP-Ribosa Cíclica/genética , ADP-Ribosa Cíclica/metabolismo , Inhibidores Enzimáticos/farmacología , Hemostasis/efectos de los fármacos , Hemostáticos/metabolismo , Hemostáticos/farmacología , Inositol 1,4,5-Trifosfato/genética , Inositol 1,4,5-Trifosfato/metabolismo , Macrólidos/farmacología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , NADP/análogos & derivados , NADP/genética , NADP/metabolismo , Fosfatidilserinas/metabolismo , Fosfatidilserinas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Trombina/farmacología
12.
J Biol Chem ; 286(11): 9136-40, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21216967

RESUMEN

The mechanism by which cyclic adenosine diphosphate ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP) mobilize intracellular Ca(2+) stores remains controversial. It is open to question whether cADPR regulates ryanodine receptors (RyRs) directly, as originally proposed, or indirectly by promoting Ca(2+) uptake into the sarco/endoplasmic reticulum by sarco/endoplasmic reticulum Ca(2+)-ATPases. Conversely, although we have proposed that NAADP mobilizes endolysosomal Ca(2+) stores by activating two-pore domain channels (TPCs), others suggest that NAADP directly activates RyRs. We therefore assessed Ca(2+) signals evoked by intracellular dialysis from a patch pipette of cADPR and NAADP into HEK293 cells that stably overexpress either TPC1, TPC2, RyR1, or RyR3. No change in intracellular Ca(2+) concentration was triggered by cADPR in either wild-type HEK293 cells (which are devoid of RyRs) or in cells that stably overexpress TPC1 and TPC2, respectively. By contrast, a marked Ca(2+) transient was triggered by cADPR in HEK293 cells that stably expressed RyR1 and RyR3. The Ca(2+) transient was abolished following depletion of endoplasmic reticulum stores by thapsigargin and block of RyRs by dantrolene but not following depletion of acidic Ca(2+) stores by bafilomycin. By contrast, NAADP failed to evoke a Ca(2+) transient in HEK293 cells that expressed RyR1 or RyR3, but it induced robust Ca(2+) transients in cells that stably overexpressed TPC1 or TPC2 and in a manner that was blocked following depletion of acidic stores by bafilomycin. We conclude that cADPR triggers Ca(2+) release by activating RyRs but not TPCs, whereas NAADP activates TPCs but not RyRs.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , ADP-Ribosa Cíclica/metabolismo , Retículo Endoplásmico/metabolismo , NADP/análogos & derivados , Canales de Calcio/genética , ADP-Ribosa Cíclica/genética , Retículo Endoplásmico/genética , Células HEK293 , Humanos , NADP/genética , NADP/metabolismo
13.
J Biol Chem ; 284(43): 29335-42, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19696022

RESUMEN

Intracellular Ca(2+) mobilization plays an important role in a wide variety of cellular processes, and multiple second messengers are responsible for mediating intracellular Ca(2+) changes. Here we explored the role of one endogenous Ca(2+)-mobilizing nucleotide, cyclic adenosine diphosphoribose (cADPR), in the proliferation and differentiation of neurosecretory PC12 cells. We found that cADPR induced Ca(2+) release in PC12 cells and that CD38 is the main ADP-ribosyl cyclase responsible for the acetylcholine (ACh)-induced cADPR production in PC12 cells. In addition, the CD38/cADPR signaling pathway is shown to be required for the ACh-induced Ca(2+) increase and cell proliferation. Inhibition of the pathway, on the other hand, accelerated nerve growth factor (NGF)-induced neuronal differentiation in PC12 cells. Conversely, overexpression of CD38 increased cell proliferation but delayed NGF-induced differentiation. Our data indicate that cADPR plays a dichotomic role in regulating proliferation and neuronal differentiation of PC12 cells.


Asunto(s)
ADP-Ribosil Ciclasa 1/biosíntesis , Señalización del Calcio/fisiología , Calcio/metabolismo , Diferenciación Celular/fisiología , ADP-Ribosa Cíclica/metabolismo , Glicoproteínas de Membrana/biosíntesis , Factor de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , ADP-Ribosil Ciclasa/biosíntesis , ADP-Ribosil Ciclasa/genética , ADP-Ribosil Ciclasa 1/genética , Acetilcolina/metabolismo , Acetilcolina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Colinérgicos/metabolismo , Colinérgicos/farmacología , ADP-Ribosa Cíclica/genética , Glicoproteínas de Membrana/genética , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/farmacología , Neuronas/citología , Células PC12 , Ratas
14.
J Neuroimmune Pharmacol ; 3(3): 154-64, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18581239

RESUMEN

CD38 is a 45-kD ectoenzyme involved in the synthesis of potent calcium (Ca(2+))-mobilizing agents, cyclic adenosine diphosphate-ribose (cADPR), and nicotinic acid adenine dinucleotide phosphate (NAADP+). In HIV-1-infected patients, increased CD38 expression on CD8+ T cells is linked to immune system activation and progression of HIV-1 infection. However, the role of CD38 upregulation in astrocyte function and HIV-1-associated dementia (HAD-now called HAND: HIV-1-associated neurocognitive disorder) neuropathogenesis is unclear. To these ends, we used interleukin (IL)-1beta and HIV-1gp120 to activate primary human astrocytes and measured CD38 expression using real-time polymerase chain reaction and CD38 function by ADP-ribosyl cyclase activity. We also determined cADPR-mediated changes in single-cell intracellular Ca(2+) transients in activated astrocytes in presence or absence of ethylene glycol tetraacetic acid. CD38 levels were downregulated using CD38 small-interfering RNA (siRNA) and intracellular Ca(2+) concentration ([Ca(2+)](i)) was measured. We previously reported a approximately 20-fold rise in CD38 messenger RNA levels in IL-1beta-activated astrocytes. We extend this observation and report that HIV-1gp120 potentiated CD38 expression in a dose-dependent manner and also increased CD38 enzyme activity in control and IL-1beta-activated astrocytes. We demonstrate higher cADPR levels in IL-1beta-activated astrocytes with a corresponding rise in [Ca(2+)](i) upon cADPR application and its non-hydrolysable analog, 3-deaza-cADPR. In activated astrocytes, pre-treatment with the cADPR-specific antagonist 8-Br-cADPR and CD38 siRNA transfection returned elevated [Ca(2+)](i) to baseline, thus confirming a CD38-cADPR specific response. These data are important for unraveling the mechanisms underlying the role of astrocyte-CD38 in HAD and have broader implications in other inflammatory diseases involving astrocyte activation and CD38 dysregulation.


Asunto(s)
ADP-Ribosil Ciclasa 1/fisiología , Complejo SIDA Demencia/patología , Astrocitos/patología , Señalización del Calcio/fisiología , ADP-Ribosa Cíclica/fisiología , VIH-1/fisiología , Mediadores de Inflamación/fisiología , ADP-Ribosil Ciclasa 1/genética , Complejo SIDA Demencia/genética , Complejo SIDA Demencia/metabolismo , Astrocitos/metabolismo , Células Cultivadas , ADP-Ribosa Cíclica/genética , Feto , Proteína gp120 de Envoltorio del VIH/fisiología , Humanos , Líquido Intracelular/metabolismo , Neuronas/metabolismo , Neuronas/patología
15.
Biochemistry ; 45(39): 11867-78, 2006 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-17002287

RESUMEN

Schistosoma mansoni NAD(P)+ catabolizing enzyme (SmNACE) is a new member of the ADP-ribosyl cyclase family. In contrast to all the other enzymes that are involved in the production of metabolites that elicit Ca2+ mobilization, SmNACE is virtually unable to transform NAD+ into the second messenger cyclic ADP-ribose (cADPR). Sequence alignments revealed that one of four conserved residues within the active site of these enzymes was replaced in SmNACE by a histidine (His103) instead of the highly conserved tryptophan. To find out whether the inability of SmNACE to catalyze the canonical ADP-ribosyl cyclase reaction is linked to this change, we have replaced His103 with a tryptophan. The H103W mutation in SmNACE was indeed found to restore ADP-ribosyl cyclase activity as cADPR amounts for 7% of the reaction products (i.e., a value larger than observed for other members of this family such as CD38). Introduction of a Trp103 residue provides some of the binding characteristics of mammalian ADP-ribosyl cyclases such as increased affinity for Cibacron blue and slow-binding inhibition by araF-NAD+. Homology modeling of wild-type and H103W mutant three-dimensional structures, and docking of substrates within the active sites, provides new insight into the catalytic mechanism of SmNACE. Both residue side chains share similar roles in the nicotinamide-ribose bond cleavage step leading to an E.ADP-ribosyl reaction intermediate. They diverge, however, in the evolution of this intermediate; His103 provides a more polar environment favoring the accessibility to water and hydrolysis leading to ADP-ribose at the expense of the intramolecular cyclization pathway resulting in cADPR.


Asunto(s)
ADP-Ribosil Ciclasa/genética , Proteínas del Helminto/genética , Mutación Missense , NAD/metabolismo , Schistosoma mansoni/genética , ADP-Ribosil Ciclasa/metabolismo , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Calcio/metabolismo , Catálisis , ADP-Ribosa Cíclica/genética , ADP-Ribosa Cíclica/metabolismo , Proteínas del Helminto/metabolismo , Humanos , Schistosoma mansoni/enzimología
16.
J Cell Biol ; 163(4): 837-45, 2003 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-14623867

RESUMEN

The role of cyclic ADP-ribose in the amplification of subcellular and global Ca2+ signaling upon stimulation of P2Y purinergic receptors was studied in 3T3 fibroblasts. Either (1) 3T3 fibroblasts (CD38- cells), (2) 3T3 fibroblasts preloaded by incubation with extracellular cyclic ADP-ribose (cADPR), (3) 3T3 fibroblasts microinjected with ryanodine, or (4) 3T3 fibroblasts transfected to express the ADP-ribosyl cyclase CD38 (CD38+ cells) were used. Both preincubation with cADPR and CD38 expression resulted in comparable intracellular amounts of cyclic ADP-ribose (42.3 +/- 5.2 and 50.5 +/- 8.0 pmol/mg protein). P2Y receptor stimulation of CD38- cells yielded a small increase of intracellular Ca2+ concentration and a much higher Ca2+ signal in CD38-transfected cells, in cADPR-preloaded cells, or in cells microinjected with ryanodine. Confocal Ca2+ imaging revealed that stimulation of ryanodine receptors by cADPR or ryanodine amplified localized pacemaker Ca2+ signals with properties resembling Ca2+ quarks and triggered the propagation of such localized signals from the plasma membrane toward the internal environment, thereby initiating a global Ca2+ wave.


Asunto(s)
Señalización del Calcio/fisiología , Membrana Celular/metabolismo , ADP-Ribosa Cíclica/metabolismo , Fibroblastos/metabolismo , Receptores Purinérgicos P2/metabolismo , ADP-Ribosil Ciclasa/deficiencia , ADP-Ribosil Ciclasa/genética , ADP-Ribosil Ciclasa 1 , Animales , Antígenos CD/genética , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Membrana Celular/efectos de los fármacos , ADP-Ribosa Cíclica/genética , ADP-Ribosa Cíclica/farmacología , Fibroblastos/efectos de los fármacos , Glicoproteínas de Membrana , Ratones , Células 3T3 NIH , Receptores Purinérgicos P2/efectos de los fármacos , Receptores Purinérgicos P2Y1 , Rianodina/farmacología , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA