当前位置: 首页 > 期刊 > 《糖尿病学杂志》 > 2006年第12期 > 正文
编号:11410938
Protein Inhibitor of Neuronal Nitric Oxide Synthase (PIN) Is a New Reg
http://www.100md.com 《糖尿病学杂志》
     1 Centre National de la Recherche Scientifique, Unite Mixte de Recherche (CNRS UMR) 5160, Center for Pharmacology and Health Biotechnology, Montpellier, France

    2 Department of Biochemistry, University Hospital, Montpellier, France

    3 INNODIA SAS, Montpellier, France

    4 CNRS UMR 5203, Institut de Genomique Fonctionnelle, Montpellier, France

    5 Centre Regional d’Imagerie Cellulaire, Institut Universitaire de Recherche Clinique, Montpellier, France

    eNOS, endothelial nitric oxide synthase; FITC, fluorescein isothiocyanate; iNOS, cytokine-inducible nitric oxide synthase; L-NAME, N-nitro-L-arginine methyl ester; nNOS, neuronal nitric oxide synthase; NOS, nitric oxide synthase; PIN, protein inhibitor of nNOS; SNP, sodium nitroprusside

    ABSTRACT

    We previously showed that pancreatic -cells express neuronal nitric oxide synthase (nNOS) that controls insulin secretion through two catalytic activities: nitric oxide (NO) production and cytochrome c reductase activity. We now provide evidence that the endogenous protein inhibitor of nNOS (PIN) is expressed in rat pancreatic islets and INS-1 cells. Double-immunofluorescence studies showed a colocalization of PIN with both nNOS and myosin Va in insulin-secreting -cells. Electron microscopy studies confirmed that PIN is mainly associated with insulin secretory granules and colocated with nNOS in the latter. In addition, PIN overexpression in INS-1 cells enhanced glucose-induced insulin secretion, which is only partly reversed by addition of an NO donor, sodium nitroprusside (SNP), and unaffected by the inhibitor of cytochrome c reductase activity, miconazole. In contrast, the pharmacological inhibitor of nNOS, N-nitro-L-arginine methyl ester, amplified glucose-induced insulin secretion, an effect insensitive to SNP but completely normalized by the addition of miconazole. Thus, PIN insulinotropic effect could be related to its colocalization with the actin-based molecular motor myosin Va and as such be implicated in the physiological regulation of glucose-induced insulin secretion at the level of the exocytotic machinery.

    The second messenger nitric oxide (NO) is synthesized from L-arginine by a family of enzymes known as NO synthases (NOSs). Three NOS isoforms have been identified, including the constitutive neuronal NO synthase (nNOS), endothelial NOS (eNOS), and the cytokine-inducible NOS (iNOS) (1). Constitutive NOSs produce small amounts of NO, implicated in physiological functions such as synaptic transmission (2) and vascular tone (3), whereas iNOS produces high amounts of NO, acting as a cytotoxic mediator in immune response (4). In addition to these beneficial roles, NO exerts deleterious effects as a result of its overproduction, leading to neuronal death during brain ischemia and certain neurodegenerative pathologies (5) and to autoimmune diseases (6). For these reasons, a tight regulation of NOS catalytic activity is essential for its physiological function. Modulation of iNOS expression by cytokines is believed to be the most important component of iNOS regulation, but posttranscriptional mechanisms also operate (7,8). As for the constitutive isoforms, catalytic activity is closely modulated by different mechanisms. The enzyme’s activity is first controlled by the reversible binding of Ca2+-calmodulin (9), which allows the transfer of NADPH-derived electrons from the reductase to the oxygenase domain of the enzyme (10). It is also positively or negatively regulated through phosphorylation by various protein kinases such as cAMP-dependent protein kinase and protein kinase C (11,12). In addition to the role of intracellular Ca2+ levels and protein kinases, constitutive NOSs are also regulated via protein-protein interactions. Thus, eNOS-associated protein-1 (ENAP-1) (13), eNOS interacting protein (NOSIP) (14), and caveolins (15) are known to modulate eNOS activity. Concerning nNOS, Jaffrey and Snyder (16) identified in rat brain a 10-kDa protein, protein inhibitor of nNOS (PIN), which interacts with the NH2-terminal part of the enzyme. Binding of PIN to nNOS prevents homodimerization of the enzyme resulting in an inhibition of NO production (16). PIN is an ubiquitous protein expressed in rats and humans (16,17), to an extent depending on the tissue studied. Thus, PIN is largely present in testis; moderately in different brain areas, kidney, and ventilary muscles; and faintly in other tissues (18–20). Furthermore, PIN has also been identified as the light chain of cytoplasmic and flagellar dyneins and myosin V, two cytoskeletal components involved in the traffic of intracellular organelles and flagellar movements (21,22). In addition, PIN is also able to interact with other partners as diverse as Bim (a proapoptotic factor of the Bcl-2 family) (23), IB (the cytoplasmic inhibitor of nuclear factor-B) (24), DAP1 (an N-methyl-D-aspartate scaffolding protein) (25), NRF-1 (nuclear respiratory factor-1) (26), Swallow (an mRNA localization protein) (27), and gephyrin (a -aminobutyric acid [GABA] or glycine scaffolding protein) (28), suggesting an interplay of PIN in a number of biological functions.

    In previous studies, we showed that pancreatic -cells express nNOS that displays three mutations when compared with cerebellar nNOS (29). Pancreatic -cell nNOS is for a great part localized in insulin secretory granules but also in mitochondria and nucleus. The enzyme is implicated in the control of insulin secretion and, as previously shown for brain NOS, is able to exert two catalytic activities: NO production and a cytochrome c reductase activity (29,30). Furthermore, we could show that a normal balance between the two catalytic activities is essential for -cell function, but the mechanisms that could regulate nNOS catalytic activities in pancreatic -cells remain to be investigated. In an attempt to get insight into -cell nNOS regulation, we hypothesized that PIN could be one of the factors implicated in nNOS regulation. This prompted us, first, to investigate if PIN is expressed in rat pancreatic -cells, second, to study its subcellular localization, and finally to determine, by using the insulin-secreting cell line INS-1, whether PIN could play a role in the regulation of glucose-induced insulin secretion.

    RESEARCH DESIGN AND METHODS

    Cell culture and isolation of islets.

    The insulin-secreting cell line INS-1 (a generous gift from Professor C.B. Wollheim) was cultured as previously described by Asfari et al. (31). Islets were isolated from adult male Wistar rats (Iffa Credo, Lyon, France) using collagenase digestion and separation on a Ficoll density gradient (32). For electron microscopy, islets were then incubated for 1 h at 37°C in Krebs-Ringer bicarbonate buffer (108 mmol/l NaCl, 1.19 mmol/l KH2PO4, 4.74 mmol/l KCl, 2.54 mmol/l CaCl2, 1.19 mmol/l MgSO4.7H2O, and 18 mmol/l NaHCO3), pH 7.4, supplemented with 1 g/l BSA and 2.8 mmol/l glucose. For immunofluorescence studies, isolated cells from islets were obtained after dissociation with 0.025% trypsin/0.27 mmol/l EDTA and then cultured in complete RPMI-1640 containing 5 mmol/l glucose for 5 days.

    RNA isolation and RT-PCR.

    Total RNA from rat-isolated islets and INS-1 cells was extracted with the Trizol reagent (Invitrogen, Carlsbad, CA). First-strand cDNA was obtained from 10 μg total RNA using 3 μg random hexanucleotide primers (Invitrogen), 1 μg oligo(dT) (Invitrogen), and Superscript II RNAse H– Reverse Transcriptase (Invitrogen). PCR was then performed with specific primers for PIN (5'-TTGAGCGGCGCCAGCACCTTCCCT-3' and 5'-CGAGGTGTTCCCTTAGCAAGGCTG-3') and for 2-micro-globulin (5'-ATCTTTCTGGTGCTTGTCTC-3' and 5'-AGTGTGAGCCAGGATGTAG-3') for 40 cycles (denaturation 94°C, 30 s; annealing 60°C, 1 min; elongation 72°C, 1 min). PCR products were finally separated on 1.5% agarose gel and visualized by ethidium bromide staining.

    Western blotting.

    Isolated islets, INS-1 cells, testis, and brain were homogenized in 20 mmol/l Tris lysis buffer, pH 7.4, containing 150 mmol/l NaCl, 1% Triton X-100, 0.5% NP-40, and a cocktail of protease inhibitors (Roche Applied Science, Mannheim, Germany). Insoluble materials were removed by centrifugation. The proteins (75 μg) were fractionated on a 14% tricine polyacrylamide gel and transferred to a nitrocellulose membrane. After blocking with 5% dried skim milk, filters were then incubated overnight with a monoclonal anti-PIN antibody (diluted 1:500; Transduction Laboratories, Lexington, KY). After three washings, the membrane was finally incubated with a horseradish peroxidase–conjugated anti-mouse antibody (diluted 1:3,000; Sigma-Aldrich, Steinheim, Germany). Immunoreactivity was detected using an enhanced chemiluminescence reaction (Amersham Biosciences, Little Chalfont, U.K.). To analyze the presence of PIN dimers, we performed, before the Western blotting, a cross-linking reaction on an INS-1 extract with difluoro-2,4-dinitrobenzene (5 mmol/l) according to the conditions described by Benashski et al. (21).

    Immunofluorescence.

    INS-1 and islet cells were seeded on a poly-L-lysine (Sigma Aldrich)–coated Lab-Tek Chamber Slide System. After fixation with 2% paraformaldehyde for 20 min, the cells were permeabilized 5 min in 0.1% Triton X-100 and saturated in 2% BSA. A combination of the following antibodies was then applied to the cells overnight: a monoclonal anti-PIN (diluted 1:25; Transduction Laboratories), a goat anti-PIN (diluted 1:25; Santa Cruz Biotechnology, Santa Cruz, CA), a rabbit anti-nNOS (diluted 1:100; Euro-Diagnostica, Malm, Sweden), a rabbit anti–myosin Va (diluted 1:100; Sigma Aldrich), a monoclonal anti-dynein (diluted 1:50; Chemicon, Chandlers Ford, U.K.), and a guinea pig anti-insulin (diluted 1:300; MP Biomedicals, Irvine, CA) antibody. After several washings, the cells were incubated for 1 h with a fluorescein isothiocyanate (FITC)-conjugated anti–guinea pig, an FITC-conjugated anti-mouse, an FITC-conjugated anti-goat, a Texas Red–conjugated anti-mouse, a Texas Red–conjugated anti-goat, or a Texas Red–conjugated anti-rabbit antibody (diluted 1:100; Vector Laboratories, Burlingame, CA). After three additional washings, the cells were finally mounted in Citifluor (Citifluor, London, U.K.) and observed with the BioRad MRC 1024 confocal microscope using the facilities of the Centre Regional d’Imagerie Cellulaire (Montpellier, France). The negative control was performed by incubating the cells with only the secondary antibody.

    Electron microscopy.

    Freshly isolated islets were fixed in a solution of 100 mmol/l phosphate buffer, pH 7, containing 2.5% paraformaldehyde and 0.1% glutaraldehyde for 1 h at room temperature. After several washings in 50 mmol/l NH4Cl PBS, the islets were postfixed in 1% osmium tetroxide for 2 min. The islets were then dehydrated in an ascending series of ethanol and routinely embedded in LR White (Electron Microscopy Sciences, Fort Washington, PA). Ultrathin sections of 60 nm were cut using a Reichert ultramicrotome (Ultracut S, Vienna, Austria) and deposited on gold grids. Sections were first saturated with 10% fetal calf serum and then incubated overnight with either the monoclonal anti-PIN (diluted 1:25), the goat anti-PIN (diluted 1:25), the rabbit anti-nNOS (diluted 1:100), or the guinea pig anti-insulin (diluted 1:500) antibody. After several washings, anti-mouse, anti-goat, anti-rabbit, and anti–guinea pig antibodies labeled respectively with 10, 15, 10, and 5 nm gold particles (diluted 1:25; British Biocell, Cardiff, U.K.) were applied to the sections for 1 h at room temperature. Sections were rinsed in deionized water, stained with 2% uranyl acetate for 20 min, and then observed with a transmission electron microscope (Hitachi H-7100; Hitachi, Düsseldorf, Germany). The specificity of the immune reaction was tested by incubating the sections with only the secondary antibody.

    Transfection and incubation experiments.

    INS-1 cells were seeded on poly-L-lysine–coated 24-well plates at a density of 4 x 105 cells per well for cells nontransfected and at 3.5 x 105 cells per well for transfected cells. After 3 days of culture, cells were transfected with a eukaryote expression vector (pCR3.1; Invitrogen) containing the cDNA of rat PIN using Lipofectamine Plus Reagent (Invitrogen), according to the manufacturer’s protocol. Control cells were obtained by transfection with the empty vector under the same conditions. Forty-eight hours after transfection, overexpression of PIN was estimated by RT-PCR, as described above. At this time, the cells transfected, or not, were washed and preincubated for 1 h at 37°C in Krebs-Ringer bicarbonate buffer containing 2 g/l BSA in the absence of glucose. After removal of the medium, the cells were incubated for another hour at 37°C in the same buffer in the presence of 5 mmol/l glucose with or without N-nitro-L-arginine methyl ester (L-NAME; 5 mmol/l), miconazole nitrate salt (10 μmol/l), and sodium nitroprusside dihydrate (3 nmol/l) (Sigma Aldrich). At the end of the incubation period, the medium was collected and insulin measured by the radioimmunological method of Herbert et al. (33) with rat insulin (Linco Research, St. Charles, MO) as the standard, allowing a sensitivity of 0.06 ng/ml.

    NOS assay.

    nNOS catalytic activity has been estimated in INS-1 extracts by measuring the production of citrulline from arginine in the presence of the enzyme cofactors, i.e., BH4, FAD (flavin adenine dinucleotide), FMN (flavin mononucleotide), NADPH, Ca2+, and calmodulin. After transfection and incubation as described above, INS-1 cells were harvested and lysed in homogenization buffer containing 25 mmol/l Tris-HCl, 1 mmol/l EDTA, and 1 mmol/l EGTA, according to the manufacturer’s recommendation (Nitric Oxide Synthase Assay Kit; Calbiochem, San Diego, CA). NOS activity was measured after a 1-h reaction at 37°C in the presence of 3 μmol/l BH4, 1 μmol/l FAD, 1 μmol/l FMN, 25 μmol/l NADPH, 15 μmol/l CaCl2, and 10 μCi L-[2,3,4,5-3H]arginine (MP Biomedicals), and separation of labeled citrulline from arginine was by affinity chromatography.

    Statistical analysis.

    Insulin secretion values are plotted on the graphs as means ± SE. Each value represents the mean of n experiments performed in quadruplicate. The data were analyzed by Student’s t test.

    RESULTS

    PIN is expressed in rat pancreatic islets and INS-1 cells.

    To investigate whether PIN, previously identified in rat brain (16), is, as nNOS, also present in rat pancreatic -cells (29), we used the RT-PCR approach with primers based on the sequence of rat brain PIN (accession no. U66461). As shown in Fig. 1A, a single band at the predicted size was obtained in islets of Langerhans and in INS-1 cells (representative of three experiments). Sequencing of this fragment revealed complete analogy with the sequence of rat brain PIN (data not shown). This result is consistent with the fact that PIN has been shown to be a highly conserved protein; a 90% sequence homology is observed among Chlamydomonas, Caenorhabditis elegans, Drosophila, and humans.

    Expression of the PIN protein was confirmed in islet and INS-1 cell extracts by Western blotting with a monoclonal anti-PIN antibody (Fig. 1B, representative of three experiments). We identified a 10-kDa protein, displaying a molecular mass identical to that observed in brain and testis. Nevertheless, the brain PIN band appeared slightly higher than in testis and islets, suggesting that different posttranslational modifications of the protein occur in these tissues. The level of PIN expression in the islets and INS-1 cells is lower than in testis and brain. These experiments provide evidence that pancreatic islets and INS-1 cells produce the PIN protein.

    PIN is localized in insulin-secreting -cells in association with nNOS and myosin Va.

    To confirm the presence of PIN in pancreatic -cells, we performed double-immunofluorescence studies on isolated cells obtained from rat islets and on INS-1 cells using a monoclonal (Fig. 2B, C, H, and I) and a polyclonal (Fig. 2E, F, K, and L) anti-PIN antibody in combination with an anti-insulin antibody (Fig. 2A, C, D, F, G, I, J, and L). These experiments clearly show that PIN is expressed in islet -cells (Fig. 2A–F) and in INS-1 cells (Fig. 2G–L). PIN is also present in glucagon-secreting -cells and to a lesser extent in somatostatin-secreting -cells (data not shown). PIN staining was found to be cytoplasmic and displayed a punctuated aspect, identical for the two antibodies used. Furthermore, PIN was strongly colocalized with insulin, as shown by the appearance of a yellow color after merging the two fluorescences (Fig. 2C, F, I, and L). This strong colocalization suggests that PIN is associated with insulin secretory granules, as previously demonstrated for nNOS (29).

    As PIN is able to interact with the NH2-terminal part of nNOS (19), we also studied the colocation of the two proteins in islet -cells (Fig. 3A–F) and in INS-1 cells (Fig. 3G–L). PIN staining was strongly associated with nNOS (Fig. 3B, C, E, F, H, I, K, and L) when using the monoclonal (Fig. 3A, C, G, and I) or the polyclonal (Fig. 3D, F, J, and L) antibodies, whereas nNOS was not exclusively colocalized with PIN. These results suggest that PIN is associated with nNOS at the level of the secretory granules.

    As PIN is the light chain of dynein and myosin V (21,22), we then studied the colocation of the inhibitor with these two motors. Our results confirm the expression of both motors in islet -cells (Fig. 4A–C and G–I) and in INS-1 cells (Fig. 4D–F and J–L). Myosin Va staining is punctuated and partially colocalized with insulin (Fig. 4A–F), as previously shown by others (34,35). Interestingly, PIN displayed a strong colocalization with myosin Va (Fig. 4G–L). Concerning dynein, the labeling occurred mainly close to the membrane of the -cell and to a lesser extent in the cytoplasm where a weak colocalization with PIN could be observed (data not shown).

    To determine the subcellular localization of PIN, we performed electron microscopy on sections of pancreatic islets using the same set of anti-PIN antibodies (Fig. 5). A similar location was observed with the monoclonal (Fig. 5A and B) and the polyclonal (Fig. 5C–H) antibodies. In -cells, PIN was mainly found to be associated with mature insulin secretory granules at the level of the dense core (large arrow head), the halo (thin arrow), and the membrane of the granules (large arrow) (Fig. 5A–C). Weak staining also appeared in less mature granules but only in association with the dense core (Fig. 5A, D, and E; empty arrow). A small part of PIN was also present in the cytoplasm (Fig. 5A; small arrow head), as well as in the mitochondria (data not shown). Interestingly, dimers of PIN could also be detected in the dense core (Fig. 5D; large arrow head), the halo (Fig. 5F; thin arrow), and the membrane (Fig. 5E; large arrow) of insulin secretory granules, suggesting that dimerization of PIN occurs in -cells as previously shown in the brain (21). The occurrence of PIN dimers was confirmed by cross-linking experiments followed by Western blotting in INS-1 cells (data not shown). Double staining of PIN and nNOS confirmed colocalization of both proteins at the level of insulin granules (Fig. 5G and H).

    PIN overexpression induced an enhanced glucose-induced insulin secretion in INS-1 cells.

    To assess the physiological function of PIN on insulin secretion, we performed overexpression experiments in INS-1 cells. After 48 h of transfection, we observed a clear increase in PIN mRNA by RT-PCR (Fig. 6A, representative of six experiments). In the presence of 5 mmol/l glucose, insulin secretion was increased by 33% in cells overexpressing PIN compared with cells transfected with the control vector (P < 0.001, n = 6) (Fig. 6B).

    To understand the mechanism by which PIN overexpression enhanced insulin secretion, we investigated if, and to what extent, the two catalytic activities of nNOS could interfere in PIN insulinotropic effect. A possible decrease in NO production was tested using sodium nitroprusside (SNP), an exogenous NO donor, and an increased cytochrome c reductase activity was challenged with a specific inhibitor, miconazole (29). Data from PIN-overexpressing cells were also compared with those obtained in nontransfected cells in the presence of L-NAME, a nonmetabolizable analog of arginine, an inhibitor of nNOS activity. We first determined the dose of SNP able to decrease glucose-induced insulin secretion in INS-1 cells. The NO donor, at 3 and 30 nmol/l, decreased insulin secretion by 21% (P < 0.01 vs. glucose alone, n = 3) and 14% (P < 0.05 vs. glucose alone, n = 3), respectively, an inhibition no longer observed for higher SNP concentrations (data not shown). In PIN-overexpressing cells, SNP (3 nmol/l) reduced insulin secretion to the same extent as in control cells but failed to bring insulin secretion back to normal values (P < 0.01, n = 3) (Fig. 6B). Besides, the 65% increase in insulin release induced by L-NAME (P < 0.001 vs. glucose alone, n = 3), at a dose of 5 mmol/l, which was previously shown not to produce nonspecific depolarizing effects (36), was not affected by SNP at increasing concentrations from 3 nmol/l (Fig. 6C, n = 3) to 300 μmol/l (data not shown). Concerning cytochrome c reductase activity, miconazole, at a dose of 10 μmol/l, which does not affect basal glucose response (29), only slightly and nonsignificantly reduced the effect of PIN overexpression (Fig. 6B, n = 6). This contrasts with the 95% reduction of the L-NAME secretory effect (P < 0.001, n = 3) (Fig. 6C), in accordance with previous data obtained in the isolated perfused rat pancreas (29). The secretory effect induced by blockade of nNOS with L-NAME is thus related to an increased cytochrome c reductase activity of the enzyme, which, in contrast, accounts for only a minor part of the positive insulinotropic effect of PIN, suggesting that the two nNOS inhibitors act through different mechanisms.

    Therefore, we also studied the combined effects of PIN overexpression and L-NAME treatment in the presence of 5 mmol/l glucose. In control cells, L-NAME again stimulated insulin secretion (P < 0.001 vs. control cells with glucose alone, n = 6), and the inhibitor produced, in PIN-overexpressing cells, a 48% increase (P < 0.001 vs. PIN-overexpressing cells with glucose alone) (Fig. 7A). Thus, L-NAME remains able to amplify insulin release in cells overexpressing PIN (+20% vs. control cells with L-NAME, P < 0.05), bringing additional evidence that PIN and L-NAME exert their positive effects on insulin secretion through different mechanisms. To ascertain whether the two inhibitors target nNOS, we measured the effects of PIN overexpression and L-NAME either alone or when combined on nNOS catalytic activity in the presence of exogenously labeled arginine. PIN overexpression only slightly reduced (–11%; P = NS, n = 4) nNOS activity, whereas a 46% inhibition was observed with L-NAME in control cells (P < 0.01) versus 75% in PIN-transfected cells (P < 0.01 vs. control cells with glucose alone) (Fig. 7B).

    DISCUSSION

    Our data clearly show that PIN is expressed in rat pancreatic -cells, as previously shown for nNOS (29), and positively modulates insulin secretion. Coincidence of nNOS and PIN also occurs in other tissues and has been reported in brain (18), kidney (20), and ventilatory muscles (19). nNOS is always coexpressed with its endogenous inhibitor, but PIN, which is moderately expressed in all rat and human tissues (16,17), does not necessarily occur with nNOS. The relative level of expression of the two proteins differs depending on both tissues and species. In brain, nNOS is mainly expressed in the cerebellum, whereas PIN is essentially found in the cortex, medulla, and hippocampus (18). Likewise, nNOS expression varies among species, which is not the case for PIN, and is found to be relatively constant (19). Therefore, it might be assumed that the magnitude of nNOS catalytic activity in a tissue is the result of the relative level of expression of both proteins.

    In addition to coincidence of both proteins in pancreatic -cells, immunofluorescence studies showed that PIN was strongly colocalized with nNOS, whereas the enzyme was not exclusively associated with the inhibitor. This discordance probably results from the fact that a part of nNOS is present in -cell mitochondria, where PIN is only faintly expressed. In other tissues, PIN often colocalizes with nNOS in the intracellular compartments, as shown in kidney, where PIN is present in the apical membranes of the inner-medullary collecting duct, a site where nNOS is strongly expressed (20). Colocalization of PIN and nNOS in pancreatic -cells makes it possible for them to interact at a site where a great part of nNOS is localized, the insulin secretory granules. This assumption is strongly supported by electron microscopy studies, which showed that PIN is mainly associated with insulin secretory granules and that PIN-nNOS association is detected at the levels of the vesicles membrane and the dense core. Interestingly, we also found that PIN colocalized with myosin Va, previously shown to be expressed in pancreatic -cells (34,35). Thus, it is tempting to assume that PIN is associated on one hand with myosin Va and on the other hand with nNOS at the level of insulin secretory granules.

    The physiological role played by PIN is not yet known. One function is the modulation of nNOS catalytic activity to prevent deleterious overproduction of NO. Indeed, it has been reported that a cyclooxygenase-induced increase of PIN expression is, through an inhibition of the production of superoxide and NO by nNOS, able to prevent nerve growth factor–deprived PC12 cells from apoptosis (37). Concerning pancreatic -cells, PIN overexpression resulted in a clear increase in insulin secretion, bearing evidence that this highly conserved protein plays a role in the stimulus-secretion coupling of the -cell. The mechanism involved is, however, less tightly coupled to nNOS catalytic activity. Indeed, the NO donor SNP reduced insulin secretion in control cells but only poorly overcame the effect of PIN overexpression. Furthermore, in vitro, a strong increase in PIN mRNA did not significantly affect nNOS catalytic activity, which contrasts with the initial identification of PIN as a specific inhibitor of nNOS catalytic activity (16). This could not be confirmed by others (38), but it must be emphasized that, at present, all studies have been performed using recombinant proteins and cells transfected with exogenous nNOS. Therefore, our finding of a slight nonsignificant reduction of nNOS activity after overexpression of endogenous PIN in -cells expressing nNOS warrants further investigations to assess if PIN does regulate nNOS activity. Taken together, our data suggest that the secretory effect of PIN may occur at a more distal level. Interestingly, PIN, as a subunit of dynein and myosin V, is involved in the trafficking of organelles and the association of intracellular proteins with the cytoskeleton. Thus, PIN has been shown to be essential for the retrograde particle movement of intraflagellar transport in Chlamydomonas reinhardtii (39). In addition, binding of PIN to different partners has been shown to be involved in cytosolic retention of the cytoplasmic inhibitor of nuclear factor-B (24) and prevention of nuclear translocation of p53 induced by DNA damage (40). In pancreatic -cells, PIN could thus be involved in the migration of insulin secretory granules as suggested by the PIN-nNOS colocalization at the level of the granules and the strong colocalization of PIN with myosin Va. Indeed, myosin Va has been shown to be implicated in the transport of dense-core secretory vesicles (34) and to control insulin granule recruitment during late-phase secretion (35). Dynein, which locates essentially at the periphery of the -cell, only partly colocalizes with PIN in the cytoplasmic part of the cells. This could be related to the finding that dynein is involved in insulin granule recovery after rapid kiss-and-run exocytosis in -cells (41) and in the subcellular distribution of mitochondria in HeLa cells (42).

    Additional evidence for the role of PIN in intracellular trafficking events also finds support in data obtained using L-NAME and the ability of the latter to reinforce the effect of PIN overexpression. In previous studies, we showed that the nonmetabolizable analog of arginine stimulates glucose-induced insulin secretion in great part through an increase in cytochrome c reductase activity. We now confirm this in INS-1 cells, where miconazole, an inhibitor of nNOS-induced cytochrome c reductase activity, is able to suppress the L-NAME secretory effect. This is not the case in PIN-overexpressing cells, which further excludes a metabolic interplay in the PIN effect. However, that L-NAME reinforced PIN effects is likely the result of an increased reduction of cytochrome c, a well-known electron transporter for mitochondrial production of ATP that may act as fuel for PIN-associated ATP-driven molecular motors. Such an assumption is also supported by the inability of the natural analog of L-NAME, L-arginine, to strengthen the PIN effect (data not shown), as the positively charged amino acid is known to depolarize pancreatic -cells via biophysical events not related to ATP production (43).

    In summary, taken together, the data we obtained by immunofluorescence and electron microscopic studies, as well as in functional experiments, clearly point to an effect of PIN in the intracellular trafficking of insulin secretory granules. As a matter of fact, PIN and nNOS clearly interact in the insulin secretory granules, and such events also occur at the vesicle membrane. Since PIN colocalizes with myosin Va, it is tempting to assume that PIN and nNOS, as two specific interacting partners, interfere with other multiprotein complexes of the exocytotic machinery.

    ACKNOWLEDGMENTS

    We are indebted to S.L. Salhi for presubmission editorial assistance. We also thank N. Lautredou, C. Cazevieille, and P. Paulet from the Centre Regional d’Imagerie Cellulaire for assistance with the confocal and electron microscopic studies.

    FOOTNOTES

    The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    REFERENCES

    Moncada S, Palmer RM, Higgs EA: Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev 43:109–142, 1991

    Bult H, Boeckxstaens GE, Pelckmans PA, Jordaens FH, Van Maercke YM, Herman AG: Nitric oxide as an inhibitory non-adrenergic non-cholinergic neurotransmitter. Nature 345:346–347, 1990

    Palmer RM, Rees DD, Ashton DS, Moncada S: L-arginine is the physiological precursor for the formation of nitric oxide in endothelium-dependent relaxation. Biochem Biophys Res Commun 153:1251–1256, 1988

    Hibbs JB, Taintor RR, Vavrin Z, Rachlin EM: Nitric oxide: a cytotoxic activated macrophage effector molecule. Biochem Biophys Res Commun 157:87–94, 1988

    Duncan AJ, Heales SJ: Nitric oxide and neurological disorders. Mol Aspects Med 26:67–96, 2005

    Kolb H, Kolb-Bachofen V: Nitric oxide in autoimmune disease: cytotoxic or regulatory mediator Immunol Today 19:556–561, 1998

    Felley-Bosco E, Bender FC, Courjault-Gautier F, Bron C, Quest AF: Caveolin-1 down-regulates inducible nitric oxide synthase via the proteasome pathway in human colon carcinoma cells. Proc Natl Acad Sci U S A 97:14334–14339, 2000

    Kleinert H, Pautz A, Linker K, Schwarz PM: Regulation of the expression of inducible nitric oxide synthase. Eur J Pharmacol 500:255–266, 2004

    Bredt DS, Snyder SH: Isolation of nitric oxide synthetase, a calmodulin-requiring enzyme. Proc Natl Acad Sci U S A 87:682–685, 1990

    Abu-Soud HM, Yoho LL, Stuehr DJ: Calmodulin controls neuronal nitric-oxide synthase by a dual mechanism: activation of intra- and interdomain electron transfer. J Biol Chem 269:32047–32050, 1994

    Bredt DS, Ferris CD, Snyder SH: Nitric oxide synthase regulatory sites: phosphorylation by cyclic AMP-dependent protein kinase, protein kinase C, and calcium/calmodulin protein kinase: identification of flavin and calmodulin binding sites. J Biol Chem 267:10976–10981, 1992

    Michell BJ, Chen ZP, Tiganis T, Stapleton D, Katsis F, Power DA, Sim AT, Kemp BE: Coordinated control of endothelial nitric-oxide synthase phosphorylation by protein kinase C and the cAMP-dependent protein kinase. J Biol Chem 276:17625–17628, 2001

    Venema VJ, Marrero MB, Venema RC: Bradykinin-stimulated protein tyrosine phosphorylation promotes endothelial nitric oxide synthase translocation to the cytoskeleton. Biochem Biophys Res Commun 226:703–710, 1996

    Dedio J, Konig P, Wohlfart P, Schroeder C, Kummer W, Muller-Esterl W: NOSIP, a novel modulator of endothelial nitric oxide synthase activity. FASEB J 15:79–89, 2001

    Feron O, Belhassen L, Kobzik L, Smith TW, Kelly RA, Michel T: Endothelial nitric oxide synthase targeting to caveolae: specific interactions with caveolin isoforms in cardiac myocytes and endothelial cells. J Biol Chem 271:22810–22814, 1996

    Jaffrey SR, Snyder SH: PIN: an associated protein inhibitor of neuronal nitric oxide synthase. Science 274:774–777, 1996

    Dick T, Ray K, Salz HK, Chia W: Cytoplasmic dynein (ddlc1) mutations cause morphogenetic defects and apoptotic cell death in Drosophila melanogaster. Mol Cell Biol 16:1966–1977, 1996

    Greenwood MT, Guo Y, Kumar U, Beausejours S, Hussain SN: Distribution of protein inhibitor of neuronal nitric oxide synthase in rat brain. Biochem Biophys Res Commun 238:617–621, 1997

    Guo Y, Greenwood MT, Petrof BJ, Hussain SN: Expression and regulation of protein inhibitor of neuronal nitric oxide synthase in ventilatory muscles. Am J Respir Cell Mol Biol 20:319–326, 1999

    Roczniak A, Levine DZ, Burns KD: Localization of protein inhibitor of neuronal nitric oxide synthase in rat kidney. Am J Physiol Renal Physiol 278:F702–F707, 2000

    Benashski SE, Harrison A, Patel-King RS, King SM: Dimerization of the highly conserved light chain shared by dynein and myosin V. J Biol Chem 272:20929–20935, 1997

    King SM, Barbarese E, Dillman JF, Patel-King RS, Carson JH, Pfister KK: Brain cytoplasmic and flagellar outer arm dyneins share a highly conserved Mr 8,000 light chain. J Biol Chem 271:19358–19366, 1996

    Puthalakath H, Huang DC, O’Reilly LA, King SM, Strasser A: The proapoptotic activity of the Bcl-2 family member Bim is regulated by interaction with the dynein motor complex. Mol Cell 3:287–296, 1999

    Crepieux P, Kwon H, Leclerc N, Spencer W, Richard S, Lin R, Hiscott J: I kappaB alpha physically interacts with a cytoskeleton-associated protein through its signal response domain. Mol Cell Biol 17:7375–7385, 1997

    Haraguchi K, Satoh K, Yanai H, Hamada F, Kawabuchi M, Akiyama T: The hDLG-associated protein DAP interacts with dynein light chain and neuronal nitric oxide synthase. Genes Cells 5:905–911, 2000

    Herzig RP, Andersson U, Scarpulla RC: Dynein light chain interacts with NRF-1 and EWG, structurally and functionally related transcription factors from humans and drosophila. J Cell Sci 113:4263–4273, 2000

    Schnorrer F, Bohmann K, Nusslein-Volhard C: The molecular motor dynein is involved in targeting swallow and bicoid RNA to the anterior pole of Drosophila oocytes. Nat Cell Biol 2:185–190, 2000

    Fuhrmann JC, Kins S, Rostaing P, El Far O, Kirsch J, Sheng M, Triller A, Betz H, Kneussel M: Gephyrin interacts with Dynein light chains 1 and 2, components of motor protein complexes. J Neurosci 22:5393–5402, 2002

    Lajoix AD, Reggio H, Chardes T, Peraldi-Roux S, Tribillac F, Roye M, Dietz S, Broca C, Manteghetti M, Ribes G, Wollheim CB, Gross R: A neuronal isoform of nitric oxide synthase expressed in pancreatic -cells controls insulin secretion. Diabetes 50:1311–1323, 2001

    Lajoix AD, Pugniere M, Roquet F, Mani JC, Dietz S, Linck N, Faurie F, Ribes G, Petit P, Gross R: Changes in the dimeric state of neuronal nitric oxide synthase affect the kinetics of secretagogue-induced insulin response. Diabetes 53:1467–1474, 2004

    Asfari M, Janjic D, Meda P, Li G, Halban PA, Wollheim CB: Establishment of 2-mercaptoethanol-dependent differentiated insulin-secreting cell lines. Endocrinology 130:167–178, 1992

    Shibata A, Ludvigsen CW Jr, Naber SP, McDaniel ML, Lacy PE: Standardization of a digestion-filtration method for isolation of pancreatic islets. Diabetes 25:667–672, 1976

    Herbert V, Lau KS, Gottlieb CW, Bleicher SJ: Coated charcoal immunoassay of insulin. J Clin Endocrinol Metab 25:1375–1384, 1965

    Varadi A, Tsuboi T, Rutter GA: Myosin Va transports dense core secretory vesicles in pancreatic MIN6 beta-cells. Mol Biol Cell 16:2670–2680, 2005

    Ivarsson R, Jing X, Waselle L, Regazzi R, Renstrom E: Myosin 5a controls insulin granule recruitment during late-phase secretion. Traffic 6:1027–1035, 2005

    Beffy P, Lajoix AD, Masiello P, Dietz S, Peraldi-Roux S, Chardes T, Ribes G, Gross R: A constitutive nitric oxide synthase modulates insulin secretion in the INS-1 cell line. Mol Cell Endocrinol 183:41–48, 2001

    Chang YW, Jakobi R, McGinty A, Foschi M, Dunn MJ, Sorokin A: Cyclooxygenase 2 promotes cell survival by stimulation of dynein light chain expression and inhibition of neuronal nitric oxide synthase activity. Mol Cell Biol 20:8571–8579, 2000

    Rodriguez-Crespo I, Straub W, Gavilanes F, Ortiz de Montellano PR: Binding of dynein light chain (PIN) to neuronal nitric oxide synthase in the absence of inhibition. Arch Biochem Biophys 359:297–304, 1998

    Pazour GJ, Wilkerson CG, Witman GB: A dynein light chain is essential for the retrograde particle movement of intraflagellar transport (IFT). J Cell Biol 141:979–992, 1998

    Lo KW, Kan HM, Chan LN, Xu WG, Wang KP, Wu Z, Sheng M, Zhang M: The 8-kDa dynein light chain binds to p53-binding protein 1 and mediates DNA damage-induced p53 nuclear accumulation. J Biol Chem 280:8172–8179, 2005

    Varadi A, Tsuboi T, Johnson-Cadwell LI, Allan VJ, Rutter GA: Kinesin I and cytoplasmic dynein orchestrate glucose-stimulated insulin-containing vesicle movements in clonal MIN6 beta-cells. Biochem Biophys Res Commun 311:272–282, 2003

    Varadi A, Johnson-Cadwell LI, Cirulli V, Yoon Y, Allan VJ, Rutter GA: Cytoplasmic dynein regulates the subcellular distribution of mitochondria by controlling the recruitment of the fission factor dynamin-related protein-1. J Cell Sci 117:4389–4400, 2004

    Smith PA, Sakura H, Coles B, Gummerson N, Proks P, Ashcroft FM: Electrogenic arginine transport mediates stimulus-secretion coupling in mouse pancreatic beta-cells. J Physiol 499:625–635, 1997(Anne-Dominique Lajoix, Stephanie Badiou,)