Network


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

Hotspot


Dive into the research topics where Viviana Filpa is active.

Publication


Featured researches published by Viviana Filpa.


Neuropharmacology | 2016

Role of glutamatergic neurotransmission in the enteric nervous system and brain-gut axis in health and disease

Viviana Filpa; Elisabetta Moro; Marina Protasoni; Francesca Crema; Gianmario Frigo; Cristina Giaroni

Several studies have been carried out in the last 30 years in the attempt to clarify the possible role of glutamate as a neurotransmitter/neuromodulator in the gastrointestinal tract. Such effort has provided immunohistochemical, biomolecular and functional data suggesting that the entire glutamatergic neurotransmitter machinery is present in the complex circuitries of the enteric nervous system (ENS), which participates to the local coordination of gastrointestinal functions. Glutamate is also involved in the regulation of the brain-gut axis, a bi-directional connection pathway between the central nervous system (CNS) and the gut. The neurotransmitter contributes to convey information, via afferent fibers, from the gut to the brain, and to send appropriate signals, via efferent fibers, from the brain to control gut secretion and motility. In analogy with the CNS, an increasing number of studies suggest that dysregulation of the enteric glutamatergic neurotransmitter machinery may lead to gastrointestinal dysfunctions. On the whole, this research field has opened the possibility to find new potential targets for development of drugs for the treatment of gastrointestinal diseases. The present review analyzes the more recent literature on enteric glutamatergic neurotransmission both in physiological and pathological conditions, such as gastroesophageal reflux, gastric acid hypersecretory diseases, inflammatory bowel disease, irritable bowel syndrome and intestinal ischemia/reperfusion injury.


European Journal of Pharmacology | 2015

Interaction between NMDA glutamatergic and nitrergic enteric pathways during in vitro ischemia and reperfusion

Viviana Filpa; Elisa Carpanese; Silvia Marchet; Valeria Prandoni; Elisabetta Moro; Sergio Lecchini; Gianmario Frigo; Cristina Giaroni; Francesca Crema

Nitric oxide (NO) and glutamate, via N-methyl-d-aspartate (NMDA) receptors, participate to changes in neuromuscular responses after ischemic/reperfusion (I/R) injury in the gut. In the present study we investigated the existence of a possible interplay between nitrergic and NMDA receptor pathways in the guinea pig ileum after in vitro I/R injury, resorting to functional and biomolecular approaches. In normal metabolic conditions NMDA concentration-dependently enhanced both glutamate (analyzed by high performance liquid chromatography with fluorimetric detection) and NO (spectrophotometrically quantified as NO2(-) and NO3(-)) spontaneous overflow from isolated ileal segments. Both effects were reduced by the NMDA antagonists, (-)-AP5 (10µM) and 5,7-diCl-kynurenic acid (10µM, 5,7-diCl-KYN). N(ω)-propyl-l-arginine (1µM, NPLA) and 1400W (10µM), respectively, nNOS and iNOS inhibitors, reduced NMDA-stimulated glutamate overflow. After in vitro I/R, glutamate overflow increased, and returned to control values in the presence of NPLA and 1400W. NO2(-) and NO3(-) levels transiently increased during I/R and were reduced by both (-)-AP5 and 5,7-diCl-KYN. In longitudinal muscle myenteric plexus preparations, iNOS mRNA and protein levels increased after in vitro I/R; both parameters were reduced to control values by (-)-AP5 and 5,7-diCl-KYN. Both antagonists were also able to reduce ischemia-induced enhancement of nNOS mRNA levels. Protein levels of GluN1, the ubiquitary subunit of NMDA receptors, increased after I/R and were reduced by both NPLA and 1400W. On the whole, this data suggests the existence of a cross-talk between NMDA receptor and nitrergic pathways in guinea pig ileum myenteric plexus, which may participate to neuronal rearrangements occurring during I/R.


British Journal of Pharmacology | 2017

Antibiotic-induced dysbiosis of the microbiota impairs gut neuromuscular function in juvenile mice

Valentina Caputi; Ilaria Marsilio; Viviana Filpa; Silvia Cerantola; Genny Orso; Michela Bistoletti; Nicola Paccagnella; Sara De Martin; Monica Montopoli; Stefano Dall'Acqua; Francesca Crema; Iole Maria Di Gangi; Francesca Galuppini; Isabella Lante; Sara Bogialli; Massimo Rugge; Patrizia Debetto; Cristina Giaroni; Maria Cecilia Giron

Gut microbiota is essential for the development of the gastrointestinal system, including the enteric nervous system (ENS). Perturbations of gut microbiota in early life have the potential to alter neurodevelopment leading to functional bowel disorders later in life. We examined the hypothesis that gut dysbiosis impairs the structural and functional integrity of the ENS, leading to gut dysmotility in juvenile mice.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2017

Nitric oxide regulates homeoprotein OTX1 and OTX2 expression in the rat myenteric plexus after intestinal ischemia-reperfusion injury

Viviana Filpa; Elisa Carpanese; Silvia Marchet; Cristina Pirrone; Andrea Conti; Alessia Rainero; Elisabetta Moro; Anna Maria Chiaravalli; Ileana Zucchi; Andrea Moriondo; Daniela Negrini; Francesca Crema; Gianmario Frigo; Cristina Giaroni; Giovanni Porta

Neuronal and inducible nitric oxide synthase (nNOS and iNOS) play a protective and damaging role, respectively, on the intestinal neuromuscular function after ischemia-reperfusion (I/R) injury. To uncover the molecular pathways underlying this dichotomy we investigated their possible correlation with the orthodenticle homeobox proteins OTX1 and OTX2 in the rat small intestine myenteric plexus after in vivo I/R. Homeobox genes are fundamental for the regulation of the gut wall homeostasis both during development and in pathological conditions (inflammation, cancer). I/R injury was induced by temporary clamping the superior mesenteric artery under anesthesia, followed by 24 and 48 h of reperfusion. At 48 h after I/R intestinal transit decreased and was further reduced by Nω-propyl-l-arginine hydrochloride (NPLA), a nNOS-selective inhibitor. By contrast this parameter was restored to control values by 1400W, an iNOS-selective inhibitor. In longitudinal muscle myenteric plexus (LMMP) preparations, iNOS, OTX1, and OTX2 mRNA and protein levels increased at 24 and 48 h after I/R. At both time periods, the number of iNOS- and OTX-immunopositive myenteric neurons increased. nNOS mRNA, protein levels, and neurons were unchanged. In LMMPs, OTX1 and OTX2 mRNA and protein upregulation was reduced by 1400W and NPLA, respectively. In myenteric ganglia, OTX1 and OTX2 staining was superimposed with that of iNOS and nNOS, respectively. Thus in myenteric ganglia iNOS- and nNOS-derived NO may promote OTX1 and OTX2 upregulation, respectively. We hypothesize that the neurodamaging and neuroprotective roles of iNOS and nNOS during I/R injury in the gut may involve corresponding activation of molecular pathways downstream of OTX1 and OTX2.NEW & NOTEWORTHY Intestinal ischemia-reperfusion (I/R) injury induces relevant alterations in myenteric neurons leading to dismotility. Nitrergic neurons seem to be selectively involved. In the present study the inference that both neuronal and inducible nitric oxide synthase (nNOS and iNOS) expressing myenteric neurons may undergo important changes sustaining derangements of motor function is reinforced. In addition, we provide data to suggest that NO produced by iNOS and nNOS regulates the expression of the vital transcription factors orthodenticle homeobox protein 1 and 2 during an I/R damage.


PLOS ONE | 2014

Antagonism of Ionotropic Glutamate Receptors Attenuates Chemical Ischemia-Induced Injury in Rat Primary Cultured Myenteric Ganglia

Elisa Carpanese; Paola Moretto; Viviana Filpa; Silvia Marchet; Elisabetta Moro; Francesca Crema; Gianmario Frigo; Cristina Giaroni

Alterations of the enteric glutamatergic transmission may underlay changes in the function of myenteric neurons following intestinal ischemia and reperfusion (I/R) contributing to impairment of gastrointestinal motility occurring in these pathological conditions. The aim of the present study was to evaluate whether glutamate receptors of the NMDA and AMPA/kainate type are involved in myenteric neuron cell damage induced by I/R. Primary cultured rat myenteric ganglia were exposed to sodium azide and glucose deprivation (in vitro chemical ischemia). After 6 days of culture, immunoreactivity for NMDA, AMPA and kainate receptors subunits, GluN1 and GluA1–4, GluK1–3 respectively, was found in myenteric neurons. In myenteric cultured ganglia, in normal metabolic conditions, -AP5, an NMDA antagonist, decreased myenteric neuron number and viability, determined by calcein AM/ethidium homodimer-1 assay, and increased reactive oxygen species (ROS) levels, measured with hydroxyphenyl fluorescein. CNQX, an AMPA/kainate antagonist exerted an opposite action on the same parameters. The total number and viability of myenteric neurons significantly decreased after I/R. In these conditions, the number of neurons staining for GluN1 and GluA1–4 subunits remained unchanged, while, the number of GluK1–3-immunopositive neurons increased. After I/R, -AP5 and CNQX, concentration-dependently increased myenteric neuron number and significantly increased the number of living neurons. Both -AP5 and CNQX (100–500 µM) decreased I/R-induced increase of ROS levels in myenteric ganglia. On the whole, the present data provide evidence that, under normal metabolic conditions, the enteric glutamatergic system exerts a dualistic effect on cultured myenteric ganglia, either by improving or reducing neuron survival via NMDA or AMPA/kainate receptor activation, respectively. However, blockade of both receptor pathways may exert a protective role on myenteric neurons following and I/R damage. The neuroprotective effect may depend, at least in part, on the ability of both receptors to increase intraneuronal ROS production.


Scientific Reports | 2017

Changes in hyaluronan deposition in the rat myenteric plexus after experimentally-induced colitis

Viviana Filpa; Michela Bistoletti; Ilaria Caon; Elisabetta Moro; Annalisa Grimaldi; Paola Moretto; Andreina Baj; Maria Cecilia Giron; Evgenia Karousou; Manuela Viola; Francesca Crema; Gianmario Frigo; Alberto Passi; Cristina Giaroni; Davide Vigetti

Myenteric plexus alterations hamper gastrointestinal motor function during intestinal inflammation. Hyaluronan (HA), an extracellular matrix glycosaminoglycan involved in inflammatory responses, may play a role in this process. In the colon of control rats, HA-binding protein (HABP), was detected in myenteric neuron soma, perineuronal space and ganglia surfaces. Prominent hyaluronan synthase 2 (HAS2) staining was found in myenteric neuron cytoplasm, suggesting that myenteric neurons produce HA. In the myenteric plexus of rats with 2, 4-dinitrobenzene sulfonic (DNBS)-induced colitis HABP staining was altered in the perineuronal space, while both HABP staining and HA levels increased in the muscularis propria. HAS2 immunopositive myenteric neurons and HAS2 mRNA and protein levels also increased. Overall, these observations suggest that inflammation alters HA distribution and levels in the gut neuromuscular compartment. Such changes may contribute to alterations in the myenteric plexus.


British Journal of Pharmacology | 2017

Antibiotic-induced microbiota dysbiosis impairs neuromuscular function in juvenile mice

Valentina Caputi; Ilaria Marsilio; Viviana Filpa; Silvia Cerantola; Genny Orso; Michela Bistoletti; Nicola Paccagnella; Sara De Martin; Monica Montopoli; Stefano Dall'Acqua; Francesca Crema; Iole Maria Di Gangi; Francesca Galuppini; Isabella Lante; Sara Bogialli; Massimo Rugge; Patrizia Debetto; Cristina Giaroni; Maria Cecilia Giron

Gut microbiota is essential for the development of the gastrointestinal system, including the enteric nervous system (ENS). Perturbations of gut microbiota in early life have the potential to alter neurodevelopment leading to functional bowel disorders later in life. We examined the hypothesis that gut dysbiosis impairs the structural and functional integrity of the ENS, leading to gut dysmotility in juvenile mice.


British Journal of Pharmacology | 2017

Antibiotic-induced dysbiosis of the microbiota impairs gut neuromuscular function in juvenile mice: Gut dysbiosis impairs neuromuscular function

Valentina Caputi; Ilaria Marsilio; Viviana Filpa; Silvia Cerantola; Genny Orso; Michela Bistoletti; Nicola Paccagnella; Sara De Martin; Monica Montopoli; Stefano Dall'Acqua; Francesca Crema; Iole-Maria Di Gangi; Francesca Galuppini; Isabella Lante; Sara Bogialli; Massimo Rugge; Patrizia Debetto; Cristina Giaroni; Maria Cecilia Giron

Gut microbiota is essential for the development of the gastrointestinal system, including the enteric nervous system (ENS). Perturbations of gut microbiota in early life have the potential to alter neurodevelopment leading to functional bowel disorders later in life. We examined the hypothesis that gut dysbiosis impairs the structural and functional integrity of the ENS, leading to gut dysmotility in juvenile mice.


Gastroenterology | 2017

Impact of Antibiotic-Induced Microbiota Depletion on Small Bowel Excitatory and Inhibitory Neuromuscular Pathways in Adolescent Mice

Valentina Caputi; Ilaria Marsilio; Silvia Cerantola; Viviana Filpa; Isabella Lante; Patrizia Debetto; Genny Orso; Francesca Crema; Rocchina Colucci; Cristina Giaroni; Maria Cecilia Giron


Gastroenterology | 2016

449 Gut Microbiota Depletion Affects Enteric Nervous System Homeostasis in Juvenile Mice

Valentina Caputi; Ilaria Marsilio; Silvia Cerantola; Viviana Filpa; Isabella Lante; Francesca Galuppini; Patrizia Debetto; Massimo Rugge; Genny Orso; Francesca Crema; Cristina Giaroni; Maria Cecilia Giron

Collaboration


Dive into the Viviana Filpa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge