487 research outputs found

    Untargeted metabolomic analysis of Rat neuroblastoma cells as a model system to study the biochemical effects of the acute administration of methamphetamine

    Get PDF
    Methamphetamine is an illicit psychostimulant drug that is linked to a number of diseases of the nervous system. The downstream biochemical effects of its primary mechanisms are not well understood, and the objective of this study was to investigate whether untargeted metabolomic analysis of an in vitro model could generate data relevant to what is already known about this drug. Rat B50 neuroblastoma cells were treated with 1 mM methamphetamine for 48 h, and both intracellular and extracellular metabolites were profiled using gas chromatography–mass spectrometry. Principal component analysis of the data identified 35 metabolites that contributed most to the difference in metabolite profiles. Of these metabolites, the most notable changes were in amino acids, with significant increases observed in glutamate, aspartate and methionine, and decreases in phenylalanine and serine. The data demonstrated that glutamate release and, subsequently, excitotoxicity and oxidative stress were important in the response of the neuronal cell to methamphetamine. Following this, the cells appeared to engage amino acid-based mechanisms to reduce glutamate levels. The potential of untargeted metabolomic analysis has been highlighted, as it has generated biochemically relevant data and identified pathways significantly affected by methamphetamine. This combination of technologies has clear uses as a model for the study of neuronal toxicology

    Repetitive low intensity magnetic field stimulation in a neuronal cell line: a metabolomics study

    Get PDF
    Low intensity repetitive magnetic stimulation of neural tissue modulates neuronal excitability and has promising therapeutic potential in the treatment of neurological disorders. However, the underpinning cellular and biochemical mechanisms remain poorly understood. This study investigates the behavioural effects of low intensity repetitive magnetic stimulation (LI-rMS) at a cellular and biochemical level. We delivered LI-rMS (10 mT) at 1 Hz and 10 Hz to B50 rat neuroblastoma cells in vitro for 10 minutes and measured levels of selected metabolites immediately after stimulation. LI-rMS at both frequencies depleted selected tricarboxylic acid (TCA) cycle metabolites without affecting the main energy supplies. Furthermore, LI-rMS effects were frequency-specific with 1 Hz stimulation having stronger effects than 10 Hz. The observed depletion of metabolites suggested that higher spontaneous activity may have led to an increase in GABA release. Although the absence of organised neural circuits and other cellular contributors (e.g., excitatory neurons and glia) in the B50 cell line limits the degree to which our results can be extrapolated to the human brain, the changes we describe provide novel insights into how LI-rMS modulates neural tissue

    The Roles of Opioid Receptors and Agonists in Health and Disease Conditions

    Get PDF
    The authors graciously acknowledge Queen Margaret University, Edinburgh for the award of the Martlet research Scholarship and the Ahmadu Bello University Zaria-Nigeria for awarding the first author study fellowship to undertake this research studies.Opioid receptors are found in the Central Nervous System (CNS) and are classified as mu (”), kappa (Îș), delta (ÎŽ) and sigma (σ) opioid receptors. Opioid receptors belong to the large family of G Protein Coupled Receptors (GPCRs), and have diverse and important physiological roles. The aim of the present review is to discuss the roles played by opioid receptors, their agonists and antagonists in health and disease conditions. Opioid receptors are not uniformly distributed in the CNS and are found in areas concerned with pain, with the highest concentration in the cerebral cortex, followed by the amygdala, septum, thalamus, hypothalamus, midbrain and spinal cord. Activated delta opioid receptors are coupled to Gi1 while activated mu opioid receptors are coupled to Gi3 in neuroblastoma cells. Mu opioid receptors are activated by mu receptor agonists and are coupled through the Gi1 and GoA. Both mu and kappa opioid receptors are coupled via both Gi and Gz and opioid receptors are important targets for thousands of pharmacological agents. GPCRs typically require activation by agonists for their signalling activity to be initiated but some of the GPCRs may display basal or spontaneous signalling activity in the absence of an agonist. The stimulation of these receptors triggers analgesic effects and affects the function of the nervous system, gastrointestinal tract and other body systems. Hundreds of analogs of opioid peptides have been synthesized in an effort to make the compounds more active, selective, and resistant to biodegradation than the endogenous ligands. All these modifications resulted in obtaining very selective agonists and antagonists with high affinity at mu-, delta-, and kappa-opioid receptors, which are useful in further studies on the pharmacology of opioid receptors in a mammalian organism.sch_dieBailey, C.P. and M. Connor, 2005. Opioids: cellular mechanisms of tolerance and physical dependence. Curr. Opin. Pharmacol., 5(1): 60-68. Berridge, M.J., 2006. Cell Signalling Biology. Portland Press Ltd., Retrieved from: www. cellsignallingbiology.org. Bouaboula, M., C. Poinot Chazel, B. Bourrie, X. Canat, B. Calandra, M. Rinaldi-Carmona, G. Le Fur and P. Casellas, 1995. Activation of mitogen-activated protein kinases by stimulation of the central cannabinoid receptor CB1. Biochem. J., 312: 637-641. Burford, N.T., D. Wang and W. Sade, 2000. G-protein coupling of mu-opioid receptors (OP3): Elevated basal signalling activity. Biochem. J., 348(3): 531-537. Carter, B.D. and F. Medzihradsky, 1993. Go mediates the coupling of the mu opioid receptor to adenylyl cyclase in cloned neural cells and brain. Proc. Natl. Acad. Sci. USA, 90(9): 4062-4066. Chakrabarti, S., P.L. Prather, L. Yu, P.Y. Law and H.H. Loh, 1995. Expression of the :-opioid receptor in CHO cells - ability of :-opioid ligands to promote a-azidoanilido [32P]GTP labelling of multiple Gprotein a subunits. J. Neurochem., 64: 2534-2543. Chaturvedi, K., K.H. Christoffers, K. Singh and R.D. Howells, 2000. Structure and regulation of opioid receptors. Biopolymers, 55(4): 334-346. Corbett, A.D., G. Henderson, A.T. McKnight and S.J. Paterson, 2006. 75 years of opioid research: the exciting but vain quest for the Holy Grail. Br. J. Pharmacol., 147: S153-S162. Corchero, J., J.A. Fuentes and J. Manzanares, 1997. delta 9-Tetrahydrocannabinol increases proopiomelanocortin gene expression in the arcuate nucleus of the rat hypothalamus. Eur. J. Pharmacol., 323(2-3): 193-195. Devi, L.A., 2001. Heterodimerization of G-proteincoupled receptors: Pharmacology, signaling and trafficking. Trend. Pharmacol. Sci., 22: 532-537. Doyle, D., G. Hanks, I. Cherney and K. Calman, 2004. Oxford Textbook of Palliative Medicine. 3rd Edn., Oxford University Press, UK, pp: 367-727. Eap, C.B., T. Buclin and P. Baumann, 2002. Interindividual variability of the clinical pharmacokinetics of methadone: Implications for the treatment of opioid dependence. Clin Pharmacokinet., 41(14): 1153-1193. Eap, C.B., J.J. Deglon and P. Boumann, 1999. Pharmacokinetics and pharmacogenetics of methadone: Clinical relevance. Heroin addiction and related clinical problems. Official J. EUROPAD, 1(1): 19-34. Fattore, L., G. Cossu, M.S. Spano, S. Deiana, P. Fadda, M. Scherma and W. Fratta, 2004. Cannabinoids and reward: Interactions with the opioid system. Crit. Rev Neurobiol., 16(1-2): 147-158. Felder, C.C. and M. Glass, 1998. Cannabinoid receptors and their endogenous agonists. Ann. Rev. Pharmacol. Toxicol., 38: 179-200. Fichna, J., K. Gach, M. Piestrzeniewicz, E. Burgeon, J. Poels, J.V. Broeck and A. Janecka, 2006. Functional characterization of opioid receptor ligands by aequorin luminescence-based calcium assay. J. Pharmacol. Exp. Ther., 317(3): 1150-1154. Freye, E. and J. Levy, 2005. Constitutive opioid receptor activation: A prerequisite mechanism involved in acute opioid withdrawal. Addict Biol., 10(2): 131-137. Fukuda, K., S. Kato, H. Morikawa, T. Shoda and K. Mori, 1996. Functional coupling of the *-, _-, and 6-opioid receptors to mitogen-activated protein kinase and arachidonate release in Chinese hamster ovary cells. J. Neurochem., 67: 1309-1316. Ghozland, S., H.W. Matthes, F. Simonin, D. Filliol, B.L. Kieffer and R. Maldonado, 2002. Motivational effects of cannabinoids are mediated by _-opioid and 6-opioid receptors. J Neurosci., 22(3): 1146-1154. Hasbi, A., S. Allouche, F. Sichel, L. Stanasila, D. Massotte, G. Landemore, J. Polastron and P. Jauzac, 2000. Internalization and recycling of delta-opioid receptor are dependent on a phosphorylation-dephosphorylation mechanism. J. Pharmacol. Exp. Ther., 293(1): 237-247. Kieffer, B.L., 1995. Recent advances in molecular recognition and signal transduction of active peptides: Receptors for opioid peptides. Cell Mol. Neurobiol., 15(6): 615-635. Koch, T. and V. Hollt, 2008. Role of receptor internalization in opioid tolerance and dependence. Pharmacol. Ther., 117(2): 199-206. Br. J. Pharmacol. Toxicol., 2(2): 84-91, 2011 90 Laugwitz, K.L., S. Offermanns, K. Spicher and G. Schultz, 1993. _ and * opioid receptors differentially couple to G protein subtypes in membranes of human neuroblastoma SH-SY5Y cells. Neuron, 10(2): 233-142. Ledent, C., O. Valverde, G. Cossu, F. Petitet, J.F. Aubert, F. Beslot, G.A. Bohme, A. Imperato, T. Pedrazzini, B.P. Roques, G. Vassart, W. Fratta and M. Parmentier, 1999. Unresponsiveness to cannabinoids and reduced addictive effects of opiates in CB1 receptor knockout mice. Science, 283: 401-404. Manzanares, J., J. Corchero, J. Romero, J.J. Fernndez- Ruiz, J.A. Ramos and J.A. Fuentes, 1999. Pharmacological and biochemical interactions between opioids and cannabinoids. Trend. Pharmacol. Sci., 20(7): 287-294. Manzanares, J., S. Ortiz, J.M. Oliva, S. Prez-Rial and T. Palomo, 2005. Interactions between cannabinoid and opioid receptor systems in the mediation of ethanol effects. Alcohol, 40(1): 25-34. Martin, M., C. Ledent, M. Parmentier, R. Maldonado and O. Valverde, 2000. Cocaine, but not morphine, induces conditioned place preference, sensitization to locomotor responses in CB1 knockout mice. Eur. J. Neurosci., 12: 4038-4046. Massotte, D. and B.L. Kieffer, 1998. A molecular basis for opiate action. Essays Biochem., 33: 65-77. Milligan, G., 2004. G protein-coupled receptor dimerization: function and ligand pharmacology. Mol. Pharmacol., 66: 1-7. Milligan, G. and E. Kostenis, 2006. Heterotrimeric Gproteins: a short history. Br. J. Pharmacol., 147(Suppl 1): S46-S55. Navarro, M., J. Chowen, A. Rocio, M. Carrera, I. del Arco, M.A. Villanua, Y. Martin, A.J. Roberts, G.F. Koob and F.R. de Fonseca, 1998. CB1 cannabinoid receptor antagonist-induced opiate withdrawal in morphine-dependent rats. Neuro Report, 9: 3397-3402. Piestrzeniewicz, M.K., J. Michna and A. Janecka, 2006. Opioid receptors and their selective ligands. Postepy Biochem., 52(3): 313-319. Piiper, A. and S. Zeuzem, 2004. Receptor tyrosine kinases are signalling intermediates of G proteincoupled receptors. Curr. Pharm. Des., 10(28): 3539- 3545. Raynor, K., H. Kong, J. Hines, G. Kong, J. Benovic, K. Yasuda, G.I. Bell and T. Reisine, 1994. Molecular mechanisms of agonist-induced desensitization of the cloned mouse kappa opioid receptor. J. Pharmacol. Exp. Ther., 270(3): 1381-1386. Raynor, K., H. Kong, S. Law, J. Heerding, M. Tallent, F. Livingston, J. Hines and T. Reisine 1996. Molecular biology of opioid receptors. NIDA Res. Monogr., 61: 83-103. Reche, I., J.A. Fuentes and M. Ruiz-Gayo, 1996. Potentiation of 9-tetrahydro cannabinol-induced analgesia by morphine in mice: involvement of _- and 6-opioid receptors. Eur. J. Pharmacol., 318: 11-16. Reisine, T. and G.I. Bell, 1993. Molecular biology of opioid receptors. Trend. Neurosci., 16(12): 506-510. Reisine, T. and M.J. Brownstein, 1994. Opioid and cannabinoid receptors. Curr. Opin. Neurobiol., 4(3): 406-412. Rhim, H. and R.J. Miller, 1994. Opioid receptors modulate diverse types of calcium channels in the nucleus tractus solitarius of the rat. J. Neurosci., 14(12): 7608-7615. Rodr_guez de Fonseca, F., P. Rubio, F. Menzaghi, E. Merlo-Pich, J. Rivier, G.F. Koob, and M. Navarro, 1996. Corticotropin-Releasing Factor (CRF) antagonist [D-Phe12, Nle21,38,C alpha MeLeu37]CRF attenuates the acute actions of the highly potent cannabinoid receptor agonist HU-210 on defensive-withdrawal behaviour in rats. J. Pharmacol. Exp. Ther., 276(1): 56-64. Saidak, Z., K. Blake-Palmer, D.L. Hay, J.K. Northup and M. Glass, 2006. Differential activation of G-proteins by mu-opioid receptor agonists. Br. J. Pharmacol., 147(6): 671-680. Smart, R.G. and A.C. Ogborne, 2000. Drug use and drinking among students in 36 countries. Addict. Behav., 25: 455-460. Samways, D.S. and G. Henderson, 2006. Opioid elevation of intracellular free calcium:possible mechanisms and physiological relevance. Cell Signal, 18(2): 151-161. Smith, F.L., D. Cichewicz, Z.L. Martin and S.P. Welch, 1998. The enhancement of morphine antinociception in mice by delta9-tetrahydrocannabinol. Pharmacol. Biochem. Behav., 60(2): 559-566. Smith, P.B., S.P. Welch and B.R. Martin, 1994. Interactions between 9-tetrahydro cannabinol and 6- opioids in mice. J. Pharmacol. Exp. Ther., 268: 1381-1387. Tanda, G., F.E. Pontieri and G. Di Chiara, 1997. Cannabinoid and heroin activation of mesolimbic dopamine transmission by a common mu1 opioid receptor mechanism. Science, 276(5321): 2048-2050. Thorat, S.N. and H.N. Bhargava, 1994. Evidence for a bidirectional cross-tolerance between morphine and 9- tetrahydrocannabinol in mice. Eur. J. Pharmacol., 260: 5-13. Tso, P.H. and Y.H. Wong, 2000. G(z) can mediate the acute actions of mu- and kappa -opioids but is not involved in opioid-induced adenylyl cyclase supersensitization. J. Pharmacol. Exp. Ther., 295(1): 168-176. Br. J. Pharmacol. Toxicol., 2(2): 84-91, 2011 91 Valverde, O., R. Maldonado, E. Valjent, A.M. Zimmer and A. Zimmer, 2000. Cannabinoid withdrawal syndrome is reduced in pre-proenkephalin knock-out mice. J. Neurosci., 20: 9284-9289. Valverde, O., F. Noble, F. Beslot, V. Dauge, M.C. Fournie-Zaluski and B.P. Roques, 2001. 9- tetrahydrocannabinol releases, facilitates the effects of endogenous enkephalins: reduction in morphine withdrawal syndrome without change in rewarding effect. Eur. J. Neurosci., 13: 1816-1824. Vela, G., M. Ruiz-Gayo and J.A. Fuentes, 1995. Anandamide decreases naloxone-precipitated withdrawal signs in mice chronically treated with morphine. Neuro Pharmacol., 34: 665-668. Vigan_, D., T. Rubino and D. Parolaro, 2005. Molecular and cellular basis of cannabinoid and opioid interactions. Pharmacol. Biochem. Behav., 81(2): 360-368. Wang, J., Q. Gao, J. Shen, T.M. Ye and Q. Xia, 2007. Kappa-opioid receptor mediates the cardioprotective effect of ischemic postconditioning. Zhejiang Da Xue Xue Bao Yi Xue Ban, 36(1): 41-47. Williams, J.T., M.J. Christie and O. Manzoni, 2001. Cellular and synaptic adaptations mediating opioid dependence. Physiol. Rev., 81: 299-343. Xiong, L.Z., J. Yang, Q. Wang and Z.H. Lu, 2007. Involvement of delta-and mu-opioid receptors in the delayed cerebral ischemic tolerance induced by repeated electroacupuncture preconditioning in rats. Chin. Med. J. (Engl), 120(5): 394-3992pub2726pub

    The Effect of Hypoxia on G Protein Coupled (CB1) Receptor Gene Expression in Cortical B50 Neurons in Culture

    Get PDF
    The authors acknowledge Queen Margaret University, Edinburgh for the award of the Martlet research Scholarship and the Ahmadu Bello University Zaria-Nigeria for awarding the first author study fellowship to undertake this research studies. The authors would like to thank Promega Corporation for generously providing us with free samples and assay kits and reagents. Our special thanks go to Drs Paul Kelly and Linda Ferrington of the Centre for Neuroscience, University of Edinburgh for their help and guidance in RT-PCR technique. Our thanks goes to Dr Elizabeth Fashola- Stone, Technical Manager European collection of cell cultures (ECACC), for providing specialist and technical advice on the use of B50 cells.Hypoxia adversely affects cells and tissues, and neuronal cells in particular have been shown to be more susceptible to the injurious effects of hypoxia in which they may begin to die when oxygen supply is reduced or completely eliminated. Cannabinoid (CB1) receptor agonists have been shown to elicit several Central Nervous System (CNS) effects, mediated via G protein-coupled receptors. The aim of this study was to examine the effect of hypoxia on G protein coupled receptor (CB1) gene expression in cortical neuronal B50 cell lines in culture. The B50 cells were cultured in normoxia (21% O2; 5% CO2) and hypoxia (5% O2; 5% CO2), and were treated with cannabinoid agonists to determine their effects on hypoxia-induced changes. Three cannabinoid agonists [Win55,212-2 mesylate (Win), arachidonoylethanolamide (AEA) and 2- arachidonylglycerol (2-AG)], were administered to the cells as treatment for 48 hours after 48hours of initial culture for a total of 96hours of culture in hypoxic conditions at concentrations of 10, 50 and 100 nM. The levels of G-protein coupled receptor (CB1) mRNAs were assessed using RT-PCR. The results showed that hypoxia induced morphological changes in B50 cells in hypoxia while the CB1 RT-PCR mRNA levels showed no appreciable changes in normal, hypoxic and treated cells. The results show that B50 neuronal cells are susceptible to damage and injurious effects of hypoxia, as are most brain cells and the cannabinoid agonist treatments showed there were no changes in the level of CB1 receptor gene expression due to hypoxia or agonist treatment in neuronal B50 cells in culture.sch_dieAguado, T., A. Carracedo, B. Julien, G. Velasco, G. Milman, R. Mechoulam, L. Alvarez, M. Guzman and I. Galve-Roperh, 2007. Cannabinoids induce glioma stem-like cell differentiation and inhibit gliomagenesis. J. Biol. Chem., 282(9): 6854-6862. Begg, M., P. Pacher, S. Batkai, D. Osei-Hyiaman, L. Offertaler, F.M. Mo, J. Liu and G. Kunos 2005. Evidence for novel cannabinoid receptors. Pharmacol Ther., 106(2):133-145. Berghuis, P., M.B. Dobszay, R.M. Ibanez, P. Ernfors and T. Harkany, 2004. Turning the heterogeneous into homogeneous: studies on selectively isolated GABAergic interneuron subsets. Int. J. Dev. Neurosci., 22(7): 533-543 Berghuis, P., M.B. Dobszay, X. Wang, S. Spano, F. Ledda, K.M. Sousa, G. Schulte, P. Ernfors, K. Mackie, G. Paratcha, Y.L. Hurd and T. Harkany, 2005. Endo-cannabinoids regulate interneuron migration and morphogenesis by transactivating the TrkB receptor. Proc Natl Acad Sci USA, 102(52): 19115-19120. Biegon, A., 2004. Cannabinoids as neuroprotective agents in traumatic brain injury. Curr. Pharm. Des., 10(18): 2177-2183. Burdyga, G., S. Lal, A. Varro, R. Dimaline, D.G. Thompson and G.J. Dockray, 2004. Expression of cannabinoid CB1 receptors by vagal afferent neurons is inhibited by cholecystokinin. J. Neurosci., 24(11): 2708-2715. Bustin, S.A., 2002. Quantification of mRNA using realtime reverse transcription PCR (RT-PCR): trends and problems. J. Mol. Endocrinol., 29(1): 23-39. Carter, G.T. and P. Weydt, 2002. Cannabis: old medicine with new promise for neurological disorders. Curr. Opin. Invest. Drugs, 3(3): 437-440. Br. J. Pharm. Toxicol., 2(1): 27-36, 2011 35 Chang, W.C., J.D. Capite, C. Nelson and A.B. Parekh, 2007. All-or-None activation of CRAC channels by agonist elicits graded responses in populations of mast cells. J. Immunol., 179: 5255-5263. Chen, J., C.T. Lee, S. Errico, X. Deng, J.L. Cadet and W.J. Freed, 2005. Protective effects of Delta(9)- tetrahydrocannabinol against N-methyl-d-aspartateinduced AF5 cell death. Brain Res. Mol. Brain Res., 134(2): 215-225. Cohen, C.D., K. Frach, D. Schlndorff and M. Kretzler, 2002. Quantitative gene expression analysis in renal biopsies: a novel protocol for a high-throughput multicenter application. Kidney Int., 61(1): 133-140. Davies, S.N., R.G. Pertwee and G. Riedel, 2002. Functions of cannabinoid receptors in the hippocampus. Neuropharmacology, 42(8): 993-1007. Devane, W.A., L. Hanus, A. Breuer, R.G. Pertwee, L.A. Stevenson, G. Griffin, D. Gibson, A. Mandelbaum, A. Etinger and R. Mechoulam, 1992. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science, 258(5090): 1946-1949. Downer, E.J., M.P. Fogarty and V.A. Campbell, 2003. Tetrahydrocannabinol-induced neurotoxicity depends on CB1 receptor-mediated c-Jun N-terminal kinase activation in cultured cortical neurons. Br. J. Pharmacol., 140: 547-557. Drysdale, A.J. and B. Platt, 2003. Cannabinoids: mechanisms and therapeutic applications in the CNS. Curr. Med. Chem., 10(24): 2719-2732. Esposito, G., A. Ligresti, A.A. Izzo, T. Bisogno, M. Ruvo, M. Di Rosa, V. Di Marzo and T. Iuvone, 2002. The endocannabinoid system protects rat glioma cells against HIV-1 Tat protein-induced cytotoxicity. Mechanism and regulation. J. Biol. Chem., 27; 277 (52): 50348-50354. Galve-Roperh, I., T. Aguado, J. Palazuelos and M. Guzmn, 2007. The endocannabinoid system and neurogenesis in health and disease. Neuroscientist, 13(2): 109-114. Gardner, F., 2006. Marijuana Might Really Make you Cool. In: Cockburn, A. and J. Clair (Eds.), Counter Pounch. Retrieved from:http://www.counterpunch. org/ gardner09092005.html. Glass, M. and C.C. Felder, 1997. Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors augments cAMP accumulation in striatal neurons: Evidence for a Gs linkage to the CB1 receptor. J. Neurosci., 17(14): 5327-5333. Gnanapavan, S., B. Kola, S.A. Bustin, D.G. Morris, P. McGee, P. Fairclough, S. Bhattacharya, R. Carpenter, A.B. Grossman and M. Korbonits, 2002. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J. Clin. Endocrinol. Metab, 87(6): 2988-2991. Grundy, R.I., 2002. The therapeutic potential of the cannabinoids in neuroprotection. Expert Opin. Inv. Drug., 11(10): 1365-1374. Hansen, H.H., I. Azcoitia, S. Pons, J. Romero, L.M. Garcia-Segura, J.A. Ramos, H.S. Hansen, and J. Fernandez-Ruiz, 2002. Blockade of cannabinoid CB(1) receptor function protects against in vivo disseminating brain damage following NMDAinduced excitotoxicity. J. Neurochem., 82(1): 154-158. Harkany, T., M. Guzmn, I. Galve-Roperh, P. Berghuis, L.A. Devi and K. Mackie, 2007. The emerging functions of endocannabinoid signaling during CNS development. Trends Pharmacol. Sci., 28(2): 83-92. Hillard, C.J., S. Manna, M.J. Greenberg, R. DiCamelli, R.A. Ross, L.A. Stevenson, V. Murphy, R.G. Pertwee and W.B. Campbell, 1997. Synthesis and characterization of potent and selective agonists of the neuronal cannabinoid receptor (CB1). J. Pharmacol. Exp. Ther., 289(3): 1427-1433. Howlett, A.C., 2004. Efficacy in CB1 receptor-mediated signal transduction. Br. J. Pharmacol., 142(8): 1209-1218. Van-Ham, I. and Y. Oron, 2005. Go G-proteins mediate rapid heterologous desensitization of G-protein coupled receptors in Xenopus oocytes. J. Cell Physiol., 204(2): 455-462. Jordan, J.D., J.C. He, N.J. Eungdamrong, I. Gomes, W. Ali, T. Nguyen, T.G. Bivona, M.R. Philips, L.A. Devi and R. Iyengar, 2005. Cannabinoid receptor-induced neurite outgrowth is mediated by Rap1 activation through Go/i-triggered proteasomal degradation of Rap1GAP II. J. Biol. Chem., 280: 11413-11421. Kearn, C.S., M.J. Greenberg, R. DiCamelli, K. Kurzawa and C.J. Hillard, 1999. Relationships between ligand affinities for the cerebellar cannabinoid receptor CB1 and the induction of GDP/GTP exchange. J. Neurochem., 72(6): 2379-2387. Lalonde, M.R., C.A. Jollimore, K. Stevens, S. Barnes and ME. Kelly, 2006. Cannabinoid receptor-mediated inhibition of calcium signaling in rat retinal ganglion cells. Mol. Vis., 12: 1160-1166. Liu, J., P. Feldman and T.D. Chung, 2002. Real-time monitoring in vitro transcription using molecular beacons. Anal. Biochem., 300(1): 40-45. Mackie, K., 2006. Cannabinoid Receptors as therapeutic targets. Ann. Rev. Pharm. Toxi., 46: 101-122. Matsuda, L.A., S.J. Lolait, M.J. Brownstein, A.C. Young and T.I. Bonner, 1990. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature, 346(6284): 561-564. Malan, T.P., M.M. Ibrahim, H. Deng, Q. Liu, H.P. Mata, T. Vanderah, F. Porreca and A Makriyannis, 2001. CB2 cannabinoid receptor-mediated peripheral antinociception. Pain, 93(3): 239-245. Br. J. Pharm. Toxicol., 2(1): 27-36, 2011 36 Mechoulam, R., D. Panikashvili and E. Shohami, 2002. Cannabinoids and brain injury: Therapeutic implications. Trends Mol. Med., 8(2): 58-61. Munro, S., K.L. Thomas and M. Abu-Shaar, 1993. Molecular characterization of a peripheral receptor for cannabinoids. Nature, 365(6441): 61-65. Nie, J. and D.L. Lewis, 2001. Structural domains of the CB1 cannabinoid receptor that contribute to constitutive activity and G-protein sequestration. J. Neurosci., 21(22): 8758-8764. Nolan, T., R.E. Hands and S.A. Bustin, 2006. Quantification of mRNA using real-time RT-PCR. Nat. Protoc., 1(3): 1559-1582. Orlando, C., P. Pinzani and M. Pazzagli, 1998. Developments in quantitative PCR. Clin. Chem. Lab. Med., 36(5): 255-269. Park, F., D.L. Mattson, L.A. Roberts and A.W. Cowley, 1997. Evidence for the presence of smooth muscle alpha-actin within pericytes of the renal medulla. Am. J. Physiol., 273(5 Pt 2): R1742-R1748. Prasad, M., I.M. Fearon, M. Zhang, M. Laing, C. Vollmer and C.A. Nurse, 2001. Expression of P2X2 and P2X3 receptor subunits in rat carotid body afferent neurones: Role in chemosensory signalling. J. Physiol., 537(Pt 3): 667-677. Pryce, G., Z. Ahmed, D.J. Hankey, S.J. Jackson, J.L. Croxford, J.M. Pocock, C. Ledent, A. Petzold, A.J. Thompson, G. Giovannoni, M.L. Cuzner and D. Baker, 2003. Cannabinoids inhibit Neuro degeneration in models of multiple sclerosis. Brain, 126(Pt 10): 2191-2202 Sarne, Y. and R. Mechoulam, 2005. Cannabinoids: between neuroprotection and neurotoxicity. Curr. Drug Targets CNS Neurol Disord, 4(6): 677-684. Showalter, S.A., N.A. Baker, C. Tang and K.B. Hall, 2005. Iron responsive element RNA flexibility described by NMR and isotropic reorientational eigenmode dynamics. J. Biomol. NMR, 32(3): 179-193. Slessareva, J.E., H. Ma, K.M. Depree, L.A. Flood, H. Bae, T.M. Cabrera-Vera, H.E. Hamm and S.G. Graber, 2003. Closely related G-protein-coupled receptors use multiple and distinct domains on Gprotein alpha-subunits for selective coupling. J. Biol. Chem., 278(50): 50530-50536. Soderstrom, K. and F. Johnson, 2000. CB1 cannabinoid receptor expression in brain regions associated with zebra finch song control. Brain Res., 857(1-2): 151-157. Steiner, H., T.I. Bonner, A.M. Zimmer, S.T. Kitai and A. Zimmer, 1999. Altered gene expression in striatal projection neurons in CB1cannabinoid receptor knockout mice. Proc. Natl. Acad. Sci. USA., 1(96): 5786-5790. Sugiura, T., S. Kondo, A. Sukagawa, S. Nakane, A. Shinoda, K. Itoh, A. Yamashita and K. Waku, 1995. 2-Arachidonoylgylcerol: A possible endogenous cannabinoid receptor ligand in brain. Biochl Bio. Res. Comns., 215(1): 89-97. Vsquez, C. and D.L. Lewis, 1999. The CB1 cannabinoid receptor can sequester G-proteins, making them unavailable to couple to other receptors. J. Neurosci., 19(21): 9271-9280. Wall, S.J. and D.R. Edwards, 2002. Quantitative reverse transcription-polymerase chain reaction (RT-PCR): a comparison of primer-dropping, competitive, and real-time RT-PCRs. Anal. Biochem., 300(2): 269-273. Wang, J., Q. Gao, J. Shen, T.M. Ye and Q. Xia, 2007. Kappa-opioid receptor mediates the cardioprotective effect of ischemic postconditioning. Zhejiang Da Xue Xue Bao Yi Xue Ban, 36(1): 41-47. Yao, L., K. McFarland, P. Fan, Z. Jiang, T. Ueda and I. Diamond, 2006. Adenosine A2a blockade prevents synergy between :-opiate and cannabinoid CB1 receptors and eliminates heroin-seeking behaviour in addicted rats. Proc. Natl. Acad. Sci. USA, 103(20): 7877-7882. Yuen, T., W. Zhang, B.J. Ebersole and S.C. Sealfon, 2002. Monitoring G-protein-coupled receptor signaling with DNA microarrays and real-time polymerase chain reaction. Methods Enzymol., 345: 556-569. Zhang, J.H., T. Lo, G. Mychaskiw and A. Colohan, 2005. Mechanisms of hyperbaric oxygen and neuroprotection in stroke. Pathophysiology, 12(1): 63-77. Zhang, J., H. Qian, P. Zhao, S.S. Hong and Y. Xia, 2006. Rapid hypoxia preconditioning protects cortical neurons from glutamate toxicity through delta-opioid receptor. Stroke, 37(4): 1094-1099. Zhuang, S.Y., A. Boon, S.A. McLeod, S. Hayashizaki, C. Padgett, R.E. Hampson and S.A. Deadwyler, 2001. Protection from NMDA toxicity by cannabinoids involves ryanodine-sensitive calcium. Soc. Neurosci. Abstract, 27: 668.11.2pub2724pub

    A wide search for obscured active galactic nuclei using XMM-Newton and WISE

    Get PDF
    Heavily obscured and Compton-thick active galactic nuclei (AGNs) are missing even in the deepest X-ray surveys, and indirect methods are required to detect them. Here we use a combination of the XMM–Newton serendipitous X-ray survey with the optical Sloan Digital Sky Survey (SDSS), and the infrared WISE all-sky survey in order to check the efficiency of the low X-ray-to-infrared luminosity selection method in finding heavily obscured AGNs. We select the sources which are detected in the hard X-ray band (2–8 keV), and also have a redshift determination (photometric or spectroscopic) in the SDSS catalogue. We match this sample with the WISE catalogue, and fit the spectral energy distributions of the 2844 sources which have three, or more, photometric data points in the infrared. We then select the heavily obscured AGN candidates by comparing their 12 Όm luminosity to the observed 2–10 keV X-ray luminosity and the intrinsic relation between the X-ray and the mid-infrared luminosities. With this approach, we find 20 candidate heavily obscured AGNs and we then examine their X-ray and optical spectra. Of the 20 initial candidates, we find nine (64 per cent; out of the 14, for which X-ray spectra could be fitted) based on the X-ray spectra, and seven (78 per cent; out of the nine detected spectroscopically in the SDSS) based on the [O III] line fluxes. Combining all criteria, we determine the final number of heavily obscured AGNs to be 12–19, and the number of Compton-thick AGNs to be 2–5, showing that the method is reliable in finding obscured AGNs, but not Compton thick. However, those numbers are smaller than what would be expected from X-ray background population synthesis models, which demonstrates how the optical–infrared selection and the scatter of the Lx-LMIR relation limit the efficiency of the method. Finally, we test popular obscured AGN selection methods based on mid-infrared colours, and find that the probability of an AGN to be selected by its mid-infrared colours increases with the X-ray luminosity. The (observed) X-ray luminosities of heavily obscured AGNs are relatively low (L2−10keV<1044ergs−1), even though most of them are located in the ‘quasi stellar object (QSO) locus’. However, a selection scheme based on a relatively low X-ray luminosity and mid-infrared colours characteristic of QSOs would not select ∌25 per cent of the heavily obscured AGNs of our sample

    Toxicological screening and DNA sequencing detects contamination and adulteration in regulated herbal medicines and supplements for diet, weight loss and cardiovascular health

    Get PDF
    Use of herbal medicines and supplements by consumers to prevent or treat disease, particularly chronic conditions continues to grow, leading to increased awareness of the minimal regulation standards in many countries. Fraudulent, adulterated and contaminated herbal and traditional medicines and dietary supplements are a risk to consumer health, with adverse effects and events including overdose, drug-herb interactions and hospitalisation. The scope of the risk has been difficult to determine, prompting calls for new approaches, such as the combination of DNA metabarcoding and mass spectrometry used in this study. Here we show that nearly 50% of products tested had contamination issues, in terms of DNA, chemical composition or both. Two samples were clear cases of pharmaceutical adulteration, including a combination of paracetamol and chlorpheniramine in one product and trace amounts of buclizine, a drug no longer in use in Australia, in another. Other issues include the undeclared presence of stimulants such as caffeine, synephrine or ephedrine. DNA data highlighted potential allergy concerns (nuts, wheat), presence of potential toxins (Neem oil) and animal ingredients (reindeer, frog, shrew), and possible substitution of bird cartilage in place of shark. Only 21% of the tested products were able to have at least one ingredient corroborated by DNA sequencing. This study demonstrates that, despite current monitoring approaches, contaminated and adulterated products are still reaching the consumer. We suggest that a better solution is stronger pre-market evaluation, using techniques such as that outlined in this study

    The Roles of Guanine Nucleotide Binding Proteins in Health and Disease

    Get PDF
    G-proteins are important mediators of cellular and tissue functions and are characterised by a recognition site for Guanine Triphosphate (GTP), Guanine Diphosphate (GDP) and possess intrinsic GTPase activity. They play important roles in signal transduction responsible for cytoskeletal remodelling, cellular differentiation and vesicular transport. They are made up of three types namely, the small G-proteins, the sensors and the heterotrimeric G-proteins. The G-protein heterotrimers consist of G-alpha (G), G-beta (G)andG−gamma(G()subunits.EachheterotrimericG−proteinhavedifferentsubunitsandthecombinationofthesesubunitsdefinethespecificroleofeachG−protein.TheactivationofGsubunitsregulatestheactivityofeffectorenzymesandionchannelswhileG) and G-gamma (G() subunits. Each heterotrimeric G-protein have different subunits and the combination of these subunits define the specific role of each G-protein. The activation of G subunits regulates the activity of effector enzymes and ion channels while G( subunits function in the regulation of mitogen-activated protein kinase (MAP-kinase) pathway. The G-protein-mediated signal transduction is important in the regulation of a cells morphological and physiological response to external stimuli. MAPKs are involved in the phosphorylation of transcription factors that stimulate gene transcription. Gs stimulates adenylate cyclase, thereby increasing cyclic adenosine monophosphate (cAMP) leading to the phosphorylation and subsequent activation of Ca_+ channels. G proteins are involved in disease pathology through several mechanisms which interfere with the G protein activity. Other disease pathologies associated with abnormal mutations in G proteins can interfere with signal transduction pathways which may involve signal transmission that is either excessive, by augmentation of G protein function, or insufficient, via inactivation of G proteins.sch_dieBenians, A., M. Nobles, S. Hosny and A. Tinker, 2005. Regulators of G-protein signalling form a quaternary complex with the agonist, receptor, and G-protein. A novel explanation for the acceleration of signalling activation kinetics. J. Biol. Chem., 280(14): 13383-13394. Berman, D.M. and A.G. Gilman, 1998. Mammalian RGS proteins: barbarians at the gate. J Biol. Chem., 273(3): 1269-1272. Berridge, M.J., 2006. Cell Signalling Biology. Portland Press Ltd. Retrieved from: www.cellsignalling biology.org. Berridge, M.J., M.D. Bootman and H.L. Roderick, 2003. Calcium signalling: Dynamics, homeostasis and remodelling. Nat. Rev. Mol. Cell Biol., 4(7): 517-529. Blackmer, T., E.C. Larsen, M. Takahashi, T.F.J. Martin, S. Alford and H.E. Hamm, 2001. G protein ( subunit-mediated presynaptic inhibition: Regulation of exocytotic fusion downstream of Ca2+ entry. Science, 292(5515): 293-297. Blaukat, A., A. Barac, M.J. Cross, S. Offermanns and I. Dikic, 2000. G protein-coupled receptor-mediated mitogen-activated protein kinase activation through cooperation of Galpha(q) and Galpha(i) signals. Mol Cell Biol., 20(18): 6837-6848 Burgoyne, R.D., 2007. Neuronal calcium sensor proteins: generating diversity in neuronal Ca2+ signalling. Nat. Rev. Neurosci., 8: 182-193. Cabrera-Vera, T.M., J. Vanhauwe, T.O. Thomas, M. Medkova, A. Preininger, M.R. Mazzoni and H.E. Hamm, 2003. Insights into G protein structure, function, and regulation. Endocr Rev., 24(6): 765-781. Clapham, D.E., 1996. Intracellular signalling: More jobs for G beta gamma. Curr. Biol., 6(7): 814-816. Danner, S. and M.J. Lohse, 1996. Phosducin is a ubiquitous G-protein regulator. Proc. Natl. Acad. Sci. U.S.A., 93(19): 10145-10150. Dhanasekaran, N. and M.V. Prasad, 1998. G protein subunits and cell proliferation. Biol Signals Recept., 7(2): 109-117. Dignard, D., D. Andr and M. Whiteway, 2008. Heterotrimeric G protein subunit function in Candida albicans: both the {} and {} subunits of the pheromone response G protein are required for mating. Eukaryot Cell, 7(9): 1591-1599. Dohlman, H.G. and J. Thorner, 1997. RGS proteins and signaling by heterotrimeric G proteins. J Biol Chem., 272(7): 3871-3874. Dolphin, A.C., 1990. G protein modulation of calcium currents in neurons. Ann. Rev. Physiol., 52: 243-255. Dolphin, A.C., 1996. Facilitation of Ca2+ current in excitable cells. Trends Neurosci., 19(1): 35-43. Durchnkov, D., J. Novotn_ and P. Svoboda, 2008. The time-course of agonist-induced solubilization of trimeric G(q)/G(11) proteins resolved by twodimensional electrophoresis. Physiol Res., 57(2): 195-203. Farfel, Z., H.R. Bourne and T. Iiri, 1999. The expanding spectrum of G protein diseases.N. Engl. J. Med., 340(13): 1012-1020. Flavahan, N.A. and P.M. Vanhoutte, 1990. G-proteins and endothelial responses. Blood Vessels, 27(2-5): 218-229. Fromm, C., O.A. Coso, S. Montaner, N. Xu and J.S. Gutkind, 1997. The small GTP-binding protein Rho links G protein-coupled receptors and Galpha12 to the serum response element and to cellular transformation. Proc. Natl. Acad. Sci. USA, 94(19): 10098-10103. Hamm, H.E. and A. Gilchrist, 1996. Heterotrimeric G proteins. Curr. Opin. Cell Biol., 8(2): 189-196. Hepler, J.R. and A.G. Gilman, 1992. G proteins. Trend. Biochem. Sci., 17(10): 383-387. Howe, L.R. and C.J. Marshall, 1993. Lysophosphatidic acid stimulates mitogen-activated protein kinase activation via a G-protein-coupled pathway requiring p21ras and p74raf-1. J. Biol. Chem., 268(28): 20717-20720. Jiang, M., M.S. Gold, G. Boulay, K. Spicher, M. Peyton, P. Brabet, Y. Srinivasan, U. Rudolph, G. Ellison and L. Birnbaumer, 1998. Multiple neurological abnormalities in mice deficient in the G protein Go. Proc. Natl. Acad. Sci. USA, 95(6): 3269-3274. Kaziro, Y., H. Itoh, T. Kozasa, M. Nakafuku and T. Satoh, 1991. Structure and function of signaltransducing GTP-binding proteins. Annu. Rev. Biochem., 60: 349-400. Kitanaka, N., J. Kitanaka, F.S. Hall, T. Tatsuta, Y. Morita, M. Takemura, X.B. Wang and G.R. Uhl, 2008. Alterations in the levels of heterotrimeric G protein subunits induced by psychostimulants, opiates, barbiturates, and ethanol: Implications for drug dependence, tolerance, and withdrawal. Synapse, 62(9): 689-699. Kolch, W., G. Heidecker, G. Kochs, R. Hummel, H. Vahidi, H. Mischak, G. Finkenzeller, D. Marme and U.R. Rapp, 1993. Protein kinase C alpha activates RAF-1 by direct phosphorylation. Nature, 364(6434): 249-252. Levitzki, A., 1990. GTP-GDP exchange proteins. Science, 248(4957): 794. Lorenz, S., R. Frenzel, R. Paschke, G.E. Breitwieser and S.U. Miedlich, 2007. Functional desensitization of the extracellular calcium-sensing receptor is regulated via distinct mechanisms: Role of G proteincoupled receptor kinases, protein kinase C and {$}- arrestins. Endocrinology, 148(5): 2398-2404. Lowes, V.L., N.Y. Ip and Y.H. Wong, 2002. Integration of signals from receptor tyrosine kinases and g protein-coupled receptors. Neurosignals, 11(1): 5-19. Luttrell, L.M., Y. Daaka, G.J. Della Rocca and R.J. Lefkowitz, 1997. G protein-coupled receptors mediate two functionally distinct pathways of tyrosine phosphorylation in rat 1a fibroblasts. Shc phosphorylation and receptor endocytosis correlate with activation of ERK kinases. J. Biol. Chem., 272(50): 31648-31656. Milligan, G., D.A. Groarke, A. McLean, R. Ward, C.W. Fong, A. Cavalli and T. Drmota, 1999. Diversity in the signalling and regulation of Gprotein- coupled receptors. Biochem. Soc. Trans., 27(2): 149-154. Milligan, G., I. Mullaney and F.R. McKenzie, 1990. Specificity of interactions of receptors and effectors with GTP-binding proteins in native membranes. Biochem. Soc Symp., 56: 21-34. Morris, A.J. and C.C. Malbon, 2000. Physiological regulation of G protein-linked signalling. Physiol. Rev., 79(4): 1373-1430. Mullaney, I., 1999. Signal transduction: A practical approach. Milligan G., 5: 73-90. Muller, S. and M.J. Lohse, 1995. The role of G-protein beta gamma subunits in signal transduction. Biochem. Soc. Trans., 23(1): 141-148. Murray, A.J. and D.A. Shewan, 2008. Epac mediates cyclic AMP-dependent axon growth, guidance and regeneration. Mol. Cell Neurosci., 38(4): 578-588. Neer, E.J., 1995. Heterotrimeric G proteins: organizers of transmembrane signals. Cell, 80(2): 249-257. Novotny, J. and P. Svoboda, 1998. The long (Gs()-L) and short (Gs()-S) variants of the stimulatory guanine nucleotide-binding protein. Do they behave in an identical way? J. Mol. Endocrinol., 20(2): 163-173. Nunn, C., H. Mao, P. Chidiac and P.R. Albert, 2006. RGS17/RGSZ2 and the RZ/A family of regulators of G-protein signaling. Semin Cell Dev. Biol., 17(3): 390-399. Ohkubo, S. and N. Nakahata, 2007. Role of lipid rafts in trimeric G protein-mediated signal transduction. Yakugaku Zasshi, 127(1): 27-40. Oldham, W.M. and H.E. Hamm, 2006. Structural basis of function in heterotrimeric G proteins. Q. Rev. Biophys., 39(2): 117-166. Schneider, T., P. Igelmund and J. Hescheler, 1997. G protein interaction with K+ and Ca2+ channels. Trend. Pharmacol. Sci., 18(1): 8-11. Schrder, S. and M.J. Lohse, 1996. Inhibition of Gprotein betagamma-subunit functions by phosducinlike protein. Proc. Natl. Acad. Sci. USA, 93(5): 2100- 2104. Siegel, G.J., B.W. Agranoff, R.W. Albers, S.K. Fisher and M.D. Uhler, 1999. Basic Neurochemistry; Molecular, Cellular and Medical Aspects. 6th Edn., Lippincott Williams and Wilkins, Philadelphia, pp: 1023-1120. Siegel, G.J., R.W. Albers, S.T. Brady and D.L. Price, 2006. Basic Neurochemistry: Molecular, Cellular and Medical Aspects. 7th Edn., Elsevier Academic Press, San Diego, pp: 339-346. Slessareva, J.E., H. Ma, K.M. Depree, L.A. Flood, H. Bae, T.M. Cabrera-Vera, H.E. Hamm and S.G. Graber 2003. Closely related G-protein-coupled receptors use multiple and distinct domains on Gprotein alpha-subunits for selective coupling. J. Biol. Chem., 278(50): 50530-50536. Sprang, S.R., 1997. G proteins, effectors and GAPs: structure and mechanism.Curr. Opin. Struct. Biol., 7(6): 849-856. Straiker, A.J., C.R. Borden and J.M. Sullivan, 2002. GProtein subunit isoforms couple differentially to receptors that mediate presynaptic inhibition at rat hippocampal synapses. J. Neurosci., 22(7): 2460-2468. Wang, L., 1999. Multi-associative neural networks and their applications to learning and retrieving complex spatio-temporal sequences. IEEE Trans. Syst. Man. Cybern B Cybern, 29(1): 73-82. Walter, L. and N. Stella, 2004. Cannabinoids and neuroinflammation. Br. J. Pharmcl., 141(5): 775-785. Walter, L., A. Franklin, A. Witting, C. Wade, Y. Xie, G. Kunos, K. Mackie and N. Stella, 2003. Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J. Neurosci., 23(4): 1398-1405. Wettschureck, N. and S. Offermanns, 2005. Mammalian G proteins and their cell type specific functions. Physiol. Rev., 85(4): 1159-1204. Wickman, K.D. and D.E. Clapham, 1995. G-protein regulation of ion channels. Curr. Opin. Neurobiol., 5(3): 278-285. Xie, G.X. and PP. Palmer 2007. How regulators of G protein signaling achieve selective regulation. J. Mol. Biol., 366(2): 349-365. Zhong, M., M. Yang and BM. Sanborn, 2003. Extracellular signal-regulated kinase 1/2 activation by myometrial oxytocin receptor involves Galpha(q)Gbetagamma and epidermal growth factor receptor tyrosine kinase activation. Endocrinology, 144(7): 2947-2956.2pub2712pub

    The evolving AGN duty cycle in galaxies since z ∌ 3 as encoded in the X-ray luminosity function

    Get PDF
    We present a new modeling of the X-ray luminosity function (XLF) of active galactic nuclei (AGNs) out to z ~ 3, dissecting the contributions of main-sequence (MS) and starburst (SB) galaxies. For each galaxy population, we convolved the observed galaxy stellar mass (M sstarf) function with a grid of M sstarf-independent Eddington ratio (λ EDD) distributions, normalized via empirical black hole accretion rate (BHAR) to star formation rate (SFR) relations. Our simple approach yields an excellent agreement with the observed XLF since z ~ 3. We find that the redshift evolution of the observed XLF can only be reproduced through an intrinsic flattening of the λ EDD distribution and with a positive shift of the break λ*, consistent with an antihierarchical behavior. The AGN accretion history is predominantly made by massive (1010 44.36 + 1.28 × (1 + z). We infer that the probability of finding highly accreting (λ EDD > 10%) AGNs significantly increases with redshift, from 0.4% (3.0%) at z = 0.5%–6.5% (15.3%) at z = 3 for MS (SB) galaxies, implying a longer AGN duty cycle in the early universe. Our results strongly favor a M sstarf-dependent ratio between BHAR and SFR, as BHAR/SFR ∝ M⋆0.73[+0.22,−0.29]{M}_{\star }^{0.73[+0.22,-0.29]}, supporting a nonlinear BH buildup relative to the host. Finally, this framework opens potential questions on super-Eddington BH accretion and different λ EDD prescriptions for understanding the cosmic BH mass assembly
    • 

    corecore