Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Gustavo Provensi is active.

Publication


Featured researches published by Gustavo Provensi.


Frontiers in Systems Neuroscience | 2012

Histamine neurons in the tuberomamillary nucleus: a whole center or distinct subpopulations?

Patrizio Blandina; Leonardo Munari; Gustavo Provensi; Maria Beatrice Passani

Histamine axons originate from a single source, the tuberomamillary nucleus (TMN) of the posterior hypothalamus, to innervate almost all central nervous system (CNS) regions. This feature, a compact cell group with widely distributed fibers, resembles that of other amine systems, such as noradrenaline or serotonin, and is consistent with a function for histamine over a host of physiological processes, including the regulation of the sleep-wake cycle, appetite, endocrine homeostasis, body temperature, pain perception, learning, memory, and emotion. An important question is whether these diverse physiological roles are served by different histamine neuronal subpopulation. While the histamine system is generally regarded as one single functional unit that provides histamine throughout the brain, evidence is beginning to accumulate in favor of heterogeneity of histamine neurons. The aim of this review is to summarize experimental evidence demonstrating that histamine neurons are heterogeneous, organized into functionally distinct circuits, impinging on different brain regions, and displaying selective control mechanisms. This could imply independent functions of subsets of histamine neurons according to their respective origin and terminal projections.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Satiety factor oleoylethanolamide recruits the brain histaminergic system to inhibit food intake

Gustavo Provensi; Roberto Coccurello; Hayato Umehara; Leonoardo Munari; Giacomo Giacovazzo; Nicoletta Galeotti; Daniele Nosi; Silvana Gaetani; Adele Romano; Anna Moles; Patrizio Blandina; Maria Beatrice Passani

Significance Several endogenous molecules contribute to the building of a complex network of neural and hormonal signals that align food intake and energy expenditure. Cerebral histamine works as a satiety factor by activating histamine H1 receptor (H1R) in specific hypothalamic nuclei. Indeed, atypical antipsychotics presumably cause obesity by targeting brain H1R. The endogenous lipid messenger oleoylethanolamide (OEA) mediates fat-induced satiety by engaging sensory fibers of the vagus nerve that project centrally. We find that depletion of brain histamine blunts OEA-induced hypophagia in mice. Our study uncovers previously unidentified neural signaling mechanisms involved in the anorectic action of OEA. Our data offer new perspectives for developing more effective and safer pharmacotherapies to treat obesity and ameliorate the profile of centrally acting drugs. Key factors driving eating behavior are hunger and satiety, which are controlled by a complex interplay of central neurotransmitter systems and peripheral stimuli. The lipid-derived messenger oleoylethanolamide (OEA) is released by enterocytes in response to fat intake and indirectly signals satiety to hypothalamic nuclei. Brain histamine is released during the appetitive phase to provide a high level of arousal in anticipation of feeding, and mediates satiety. However, despite the possible functional overlap of satiety signals, it is not known whether histamine participates in OEA-induced hypophagia. Using different experimental settings and diets, we report that the anorexiant effect of OEA is significantly attenuated in mice deficient in the histamine-synthesizing enzyme histidine decarboxylase (HDC-KO) or acutely depleted of histamine via interocerebroventricular infusion of the HDC blocker α-fluoromethylhistidine (α-FMH). α-FMH abolished OEA-induced early occurrence of satiety onset while increasing histamine release in the CNS with an H3 receptor antagonist-increased hypophagia. OEA augmented histamine release in the cortex of fasted mice within a time window compatible to its anorexic effects. OEA also increased c-Fos expression in the oxytocin neurons of the paraventricular nuclei of WT but not HDC-KO mice. The density of c-Fos immunoreactive neurons in other brain regions that receive histaminergic innervation and participate in the expression of feeding behavior was comparable in OEA-treated WT and HDC-KO mice. Our results demonstrate that OEA requires the integrity of the brain histamine system to fully exert its hypophagic effect and that the oxytocin neuron-rich nuclei are the likely hypothalamic area where brain histamine influences the central effects of OEA.


Neuropharmacology | 2013

Selective brain region activation by histamine H3 receptor antagonist/inverse agonist ABT-239 enhances acetylcholine and histamine release and increases c-Fos expression

Leonardo Munari; Gustavo Provensi; Maria Beatrice Passani; Patrizio Blandina

Histamine axons originate solely from the tuberomamillary nucleus (TMN) to innervate almost all brain regions. This feature is consistent with a function for histamine over a host of physiological processes, including regulation of appetite, body temperature, cognitive processes, pain perception and sleep-wake cycle. An important question is whether these diverse physiological roles are served by different histamine neuronal subpopulations. Here we report that systemic administration of the non-imidazole histamine H₃ receptor antagonist 4-(2-{2-[(2R)-2-methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitrile (ABT-239, 3 mg/kg) increased c-Fos expression dose-dependently in rat cortex and nucleus basalis magnocellularis (NBM) but not in the nucleus accumbens (NAcc) nor striatum, and augmented acetylcholine and histamine release from rat prefrontal cortex. To further understand functional histaminergic pathways in the brain, dual-probe microdialysis was used to pharmacologically block H₃ receptors in the TMN. Perfusion of the TMN with ABT-239 (10 μM) increased histamine release from the TMN, NBM, and cortex, but not from the striatum or NAcc. When administered locally, ABT-239 increased histamine release from the NBM, but not from the NAcc. Systemic as well as intra-TMN administration of ABT-239 increased c-Fos expression in the NBM, and cortex, but not in the striatum or NAcc. Thus, as defined by their sensitivity to ABT-239, histaminergic neurons establish distinct pathways according to their terminal projections, and can differentially modulate neurotransmitter release in a brain region-specific manner. This implies independent functions of subsets of histamine neurons according to their terminal projections, with relevant consequences for the development of specific compounds that affect only subsets of histamine neurones, thus increasing target specificity.


British Journal of Pharmacology | 2014

Histamine mediates behavioural and metabolic effects of 3-iodothyroacetic acid, an endogenous end product of thyroid hormone metabolism

Claudia Musilli; Gaetano De Siena; Maria Elena Manni; Andrea Logli; Elisa Landucci; Riccardo Zucchi; Alessandro Saba; Riccardo Donzelli; Maria Beatrice Passani; Gustavo Provensi; Laura Raimondi

3‐Iodothyroacetic acid (TA1) is an end product of thyroid hormone metabolism. So far, it is not known if TA1 is present in mouse brain and if it has any pharmacological effects.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Histamine in the basolateral amygdala promotes inhibitory avoidance learning independently of hippocampus

Fernando Benetti; Cristiane Regina Guerino Furini; Jociane de Carvalho Myskiw; Gustavo Provensi; Maria Beatrice Passani; Elisabetta Baldi; Corrado Bucherelli; Leonardo Munari; Ivan Izquierdo; Patrizio Blandina

Significance Integrity of the brain histaminergic system is necessary for long-term memory (LTM) but not short-term memory of step-down inhibitory avoidance (IA). Histamine depletion in hippocampus or basolateral amygdala (BLA) impairs LTM of that task. Histamine infusion into either structure restores LTM in histamine-depleted rats. The restoring effect in BLA occurs even when hippocampal activity was impaired. Cyclic adenosine monophosphate (cAMP) responsive-element-binding protein phosphorylation correlates anatomically and temporally with histamine-induced memory recall. Thus, histaminergic neurotransmission appears critical to provide the brain with the plasticity necessary for IA through recruitment of alternative circuits. Our findings indicate that the histaminergic system comprises parallel, coordinated pathways that provide compensatory plasticity when one brain structure is compromised. Recent discoveries demonstrated that recruitment of alternative brain circuits permits compensation of memory impairments following damage to brain regions specialized in integrating and/or storing specific memories, including both dorsal hippocampus and basolateral amygdala (BLA). Here, we first report that the integrity of the brain histaminergic system is necessary for long-term, but not for short-term memory of step-down inhibitory avoidance (IA). Second, we found that phosphorylation of cyclic adenosine monophosphate (cAMP) responsive-element-binding protein, a crucial mediator in long-term memory formation, correlated anatomically and temporally with histamine-induced memory retrieval, showing the active involvement of histamine function in CA1 and BLA in different phases of memory consolidation. Third, we found that exogenous application of histamine in either hippocampal CA1 or BLA of brain histamine-depleted rats, hence amnesic, restored long-term memory; however, the time frame of memory rescue was different for the two brain structures, short lived (immediately posttraining) for BLA, long lasting (up to 6 h) for the CA1. Moreover, long-term memory was formed immediately after training restoring of histamine transmission only in the BLA. These findings reveal the essential role of histaminergic neurotransmission to provide the brain with the plasticity necessary to ensure memorization of emotionally salient events, through recruitment of alternative circuits. Hence, our findings indicate that the histaminergic system comprises parallel, coordinated pathways that provide compensatory plasticity when one brain structure is compromised.


Frontiers in Pharmacology | 2015

Central mechanisms mediating the hypophagic effects of oleoylethanolamide and N-acylphosphatidylethanolamines: different lipid signals?

Adele Romano; Bianca Tempesta; Gustavo Provensi; Maria Beatrice Passani; Silvana Gaetani

The spread of “obesity epidemic” and the poor efficacy of many anti-obesity therapies in the long-term highlight the need to develop novel efficacious therapy. This necessity stimulates a large research effort to find novel mechanisms controlling feeding and energy balance. Among these mechanisms a great deal of attention has been attracted by a family of phospholipid-derived signaling molecules that play an important role in the regulation of food-intake. They include N-acylethanolamines (NAEs) and N-acylphosphatidylethanolamines (NAPEs). NAPEs have been considered for a long time simply as phospholipid precursors of the lipid mediator NAEs, but increasing body of evidence suggest a role in many physiological processes including the regulation of feeding behavior. Several observations demonstrated that among NAEs, oleoylethanolamide (OEA) acts as a satiety signal, which is generated in the intestine, upon the ingestion of fat, and signals to the central nervous system. At this level different neuronal pathways, including oxytocinergic, noradrenergic, and histaminergic neurons, seem to mediate its hypophagic action. Similarly to NAEs, NAPE (with particular reference to the N16:0 species) levels were shown to be regulated by the fed state and this finding was initially interpreted as fluctuations of NAE precursors. However, the observation that exogenously administered NAPEs are able to inhibit food intake, not only in normal rats and mice but also in mice lacking the enzyme that converts NAPEs into NAEs, supported the hypothesis of a role of NAPE in the regulation of feeding behavior. Indirect observations suggest that the hypophagic action of NAPEs might involve central mechanisms, although the molecular target remains unknown. The present paper reviews the role that OEA and NAPEs play in the mechanisms that control food intake, further supporting this group of phospholipids as optimal candidate for the development of novel anti-obesity treatments.


Neuropharmacology | 2017

Carbonic anhydrase activation enhances object recognition memory in mice through phosphorylation of the extracellular signal-regulated kinase in the cortex and the hippocampus

Lucas Canto de Souza; Gustavo Provensi; Daniela Vullo; Fabrizio Carta; Andrea Scozzafava; Alessia Costa; Scheila Daiane Schmidt; Maria Beatrice Passani; Claudiu T. Supuran; Patrizio Blandina

ABSTRACT Rats injected with by d‐phenylalanine, a carbonic anhydrase (CA) activator, enhanced spatial learning, whereas rats given acetazolamide, a CA inhibitor, exhibited impairments of fear memory consolidation. However, the related mechanisms are unclear. We investigated if CAs are involved in a non‐spatial recognition memory task assessed using the object recognition test (ORT). Systemic administration of acetazolamide to male CD1 mice caused amnesia in the ORT and reduced CA activity in brain homogenates, while treatment with d‐phenylalanine enhanced memory and increased CA activity. We provided also the first evidence that d‐phenylalanine administration rapidly activated extracellular signal‐regulated kinase (ERK) pathways, a critical step for memory formation, in the cortex and the hippocampus, two brain areas involved in memory processing. Effects elicited by d‐phenylalanine were completely blunted by co‐administration of acetazolamide, but not of 1‐N‐(4‐sulfamoylphenyl‐ethyl)‐2,4,6‐trimethylpyridinium perchlorate (C18), a CA inhibitor that, differently from acetazolamide, does not cross the blood brain barrier. Our results strongly suggest that brain but not peripheral CAs activation potentiates memory as a result of ERK pathway enhanced activation. HIGHLIGHTSAcetazolamide, a carbonic anhydrase (CA) inhibitor, administered to CD1 mice caused amnesia in the object recognition test.C18, a CA inhibitor that does not cross the blood brain barrier, did not produce amnesia in the object recognition test.d‐phenylalanine, a CA activator that crosses the blood brain barrier, enhanced memory and increased CA activity in the brain.d‐phenylalanine administration activated ERK pathway, a critical step for memory formation, in mice hippocampus and cortex.The effects elicited by d‐phenylalanine were blunted by co‐administration of acetazolamide, but not of C18.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Memory retrieval of inhibitory avoidance requires histamine H1 receptor activation in the hippocampus.

Roberta Fabbri; Cristiane Regina Guerino Furini; Maria Beatrice Passani; Gustavo Provensi; Elisabetta Baldi; Corrado Bucherelli; Ivan Izquierdo; Jociane de Carvalho Myskiw; Patrizio Blandina

Significance Several neurotransmitters contribute to memory formation by modulating selectively acquisition, consolidation, and/or retrieval. Integrity of the brain histamine system is necessary for the consolidation of inhibitory avoidance (IA) memory. Here, we report that cerebral histamine depletion also impairs retrieval of IA in rats and blunts retrieval-induced c-Fos activation and cAMP-responsive element binding protein phosphorylation in the CA1 region of the hippocampus. Histamine infusion into the CA1 restores IA retrieval in histamine-depleted rats by targeting brain histamine H1 receptors. Our study uncovers previously unidentified mechanisms involved in memory retrieval and may offer possible targets for eventual pharmacotherapies to treat dysfunctional aversive memories, including phobias, panic attacks, and posttraumatic stress disorders, as well as improve the efficacy of exposure psychotherapies. Retrieval represents a dynamic process that may require neuromodulatory signaling. Here, we report that the integrity of the brain histaminergic system is necessary for retrieval of inhibitory avoidance (IA) memory, because rats depleted of histamine through lateral ventricle injections of α-fluoromethylhistidine (a-FMHis), a suicide inhibitor of histidine decarboxylase, displayed impaired IA memory when tested 2 d after training. a-FMHis was administered 24 h after training, when IA memory trace was already formed. Infusion of histamine in hippocampal CA1 of brain histamine-depleted rats (hence, amnesic) 10 min before the retention test restored IA memory but was ineffective when given in the basolateral amygdala (BLA) or the ventral medial prefrontal cortex (vmPFC). Intra-CA1 injections of selective H1 and H2 receptor agonists showed that histamine exerted its effect by activating the H1 receptor. Noteworthy, the H1 receptor antagonist pyrilamine disrupted IA memory retrieval in rats, thus strongly supporting an active involvement of endogenous histamine; 90 min after the retention test, c-Fos–positive neurons were significantly fewer in the CA1s of a-FMHis–treated rats that displayed amnesia compared with in the control group. We also found reduced levels of phosphorylated cAMP-responsive element binding protein (pCREB) in the CA1s of a-FMHis–treated animals compared with in controls. Increases in pCREB levels are associated with retrieval of associated memories. Targeting the histaminergic system may modify the retrieval of emotional memory; hence, histaminergic ligands might reduce dysfunctional aversive memories and improve the efficacy of exposure psychotherapies.


Neuropharmacology | 2016

Donepezil, an acetylcholine esterase inhibitor, and ABT-239, a histamine H3 receptor antagonist/inverse agonist, require the integrity of brain histamine system to exert biochemical and procognitive effects in the mouse.

Gustavo Provensi; Alessia Costa; Maria Beatrice Passani; Patrizio Blandina

Histaminergic H3 receptors (H3R) antagonists enhance cognition in preclinical models and modulate neurotransmission, in particular acetylcholine (ACh) release in the cortex and hippocampus, two brain areas involved in memory processing. The cognitive deficits seen in aging and Alzheimers disease have been associated with brain cholinergic deficits. Donepezil is one of the acetylcholinesterase (AChE) inhibitor approved for use across the full spectrum of these cognitive disorders. We addressed the question if H3R antagonists and donepezil require an intact histamine neuronal system to exert their procognitive effects. The effect of the H3R antagonist ABT-239 and donepezil were evaluated in the object recognition test (ORT), and on the level of glycogen synthase kinase 3 beta (GSK-3β) phosphorylation in normal and histamine-depleted mice. Systemic administration of ABT-239 or donepezil ameliorated the cognitive performance in the ORT. However, these compounds were ineffective in either genetically (histidine decarboxylase knock-out, HDC-KO) or pharmacologically, by means of intracerebroventricular (i.c.v.) injections of the HDC irreversible inhibitor a-fluoromethylhistidine (a-FMHis), histamine-deficient mice. Western blot analysis revealed that ABT-239 or donepezil systemic treatments increased GSK-3β phosphorylation in cortical and hippocampal homogenates of normal, but not of histamine-depleted mice. Furthermore, administration of the PI3K inhibitor LY294002 that blocks GSK-3β phosphorylation, prevented the procognitive effects of both drugs in normal mice. Our results indicate that both donepezil and ABT-239 require the integrity of the brain histaminergic system to exert their procognitive effects and strongly suggest that impairments of PI3K/AKT/GSK-3β intracellular pathway activation is responsible for the inefficacy of both drugs in histamine-deficient animals.


Journal of Neurochemistry | 2016

Eating disorders: from bench to bedside and back

Silvana Gaetani; Adele Romano; Gustavo Provensi; Valdo Ricca; Thomas A. Lutz; Maria Beatrice Passani

The central nervous system and viscera constitute a functional ensemble, the gut–brain axis, that allows bidirectional information flow that contributes to the control of feeding behavior based not only on the homeostatic, but also on the hedonic aspects of food intake. The prevalence of eating disorders, such as anorexia nervosa, binge eating and obesity, poses an enormous clinical burden, and involves an ever‐growing percentage of the population worldwide. Clinical and preclinical research is constantly adding new information to the field and orienting further studies with the aim of providing a foundation for developing more specific and effective treatment approaches to pathological conditions. A recent symposium at the XVI Congress of the Societá Italiana di Neuroscienze (SINS, 2015) ‘Eating disorders: from bench to bedside and back’ brought together basic scientists and clinicians with the objective of presenting novel perspectives in the neurobiology of eating disorders. Clinical studies presented by V. Ricca illustrated some genetic aspects of the psychopathology of anorexia nervosa. Preclinical studies addressed different issues ranging from the description of animal models that mimic human pathologies such as anorexia nervosa, diet‐induced obesity, and binge eating disorders (T. Lutz), to novel interactions between peripheral signals and central circuits that govern food intake, mood and stress (A. Romano and G. Provensi). The gut–brain axis has received increasing attention in the recent years as preclinical studies are demonstrating that the brain and visceral organs such as the liver and guts, but also the microbiota are constantly engaged in processes of reciprocal communication, with unexpected physiological and pathological implications. Eating is controlled by a plethora of factors; genetic predisposition, early life adverse conditions, peripheral gastrointestinal hormones that act directly or indirectly on the central nervous system, all are receiving attention as they presumably contribute to the development of eating disorders.

Collaboration


Dive into the Gustavo Provensi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Silvana Gaetani

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Raquel Fenner

Universidade Federal do Rio Grande do Sul

View shared research outputs
Top Co-Authors

Avatar

Adele Romano

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge