Network


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

Hotspot


Dive into the research topics where Gabriela Bergonzelli is active.

Publication


Featured researches published by Gabriela Bergonzelli.


Neurogastroenterology and Motility | 2011

The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut–brain communication

P. Bercik; Amber J. Park; David A. Sinclair; A. Khoshdel; Jun Lu; Xianxi Huang; Yikang Deng; Patricia Blennerhassett; Margaret Fahnestock; D. Moine; B. Berger; Jan D. Huizinga; Wolfgang A. Kunze; P. McLean; Gabriela Bergonzelli; Stephen M. Collins; Elena F. Verdu

Background  The probiotic Bifidobacterium longum NCC3001 normalizes anxiety‐like behavior and hippocampal brain derived neurotrophic factor (BDNF) in mice with infectious colitis. Using a model of chemical colitis we test whether the anxiolytic effect of B. longum involves vagal integrity, and changes in neural cell function.


Infection and Immunity | 2004

Cell surface-associated elongation factor Tu mediates the attachment of Lactobacillus johnsonii NCC533 (La1) to human intestinal cells and mucins

Dominique Granato; Gabriela Bergonzelli; Raymond David Pridmore; Laure Marvin; Martine Rouvet; Irène Corthesy-Theulaz

ABSTRACT The aim of this work was to identify Lactobacillus johnsonii NCC533 (La1) surface molecules mediating attachment to intestinal epithelial cells and mucins. Incubation of Caco-2 intestinal epithelial cells with an L. johnsonii La1 cell wall extract led to the recognition of elongation factor Tu (EF-Tu) as a novel La1 adhesin-like factor. The presence of EF-Tu at the surface of La1 was confirmed by analysis of purified outer surface protein extract by immunoblotting experiments, by electron microscopy, and by enzyme-linked immunosorbent assays of live bacteria. Furthermore, tandem mass spectrometry analysis proved that EF-TU was expressed at the La1 surface as an intact molecule. Using recombinant La1 EF-Tu protein, we were able to determine that its binding to intestinal cells and to mucins is pH dependent. Competition experiments suggested that EF-Tu has an important role in La1 mucin binding capacity. In addition, immunomodulation studies performed on HT29 cells showed that EF-Tu recombinant protein can induce a proinflammatory response in the presence of soluble CD14. Our in vitro results indicate that EF-Tu, through its binding to the intestinal mucosa, might participate in gut homeostasis.


Gastroenterology | 2010

Chronic Gastrointestinal Inflammation Induces Anxiety-Like Behavior and Alters Central Nervous System Biochemistry in Mice

Premysl Bercik; Elena F. Verdu; Jane A. Foster; Joseph Macri; Murray Potter; Xiaxing Huang; Paul Malinowski; Wendy Jackson; Patricia Blennerhassett; Karen A. Neufeld; Jun Lu; Waliul I. Khan; Irène E. Corthésy–Theulaz; Christine Cherbut; Gabriela Bergonzelli; Stephen M. Collins

BACKGROUND & AIMS Clinical and preclinical studies have associated gastrointestinal inflammation and infection with altered behavior. We investigated whether chronic gut inflammation alters behavior and brain biochemistry and examined underlying mechanisms. METHODS AKR mice were infected with the noninvasive parasite Trichuris muris and given etanercept, budesonide, or specific probiotics. Subdiaphragmatic vagotomy was performed in a subgroup of mice before infection. Gastrointestinal inflammation was assessed by histology and quantification of myeloperoxidase activity. Serum proteins were measured by proteomic analysis, circulating cytokines were measured by fluorescence activated cell sorting array, and serum tryptophan and kynurenine were measured by liquid chromatography. Behavior was assessed using light/dark preference and step-down tests. In situ hybridization was used to assess brain-derived neurotrophic factor (BDNF) expression in the brain. RESULTS T muris caused mild to moderate colonic inflammation and anxiety-like behavior that was associated with decreased hippocampal BDNF messenger RNA (mRNA). Circulating tumor necrosis factor-α and interferon-γ, as well as the kynurenine and kynurenine/tryptophan ratio, were increased. Proteomic analysis showed altered levels of several proteins related to inflammation and neural function. Administration of etanercept, and to a lesser degree of budesonide, normalized behavior, reduced cytokine and kynurenine levels, but did not influence BDNF expression. The probiotic Bifidobacterium longum normalized behavior and BDNF mRNA but did not affect cytokine or kynurenine levels. Anxiety-like behavior was present in infected mice after vagotomy. CONCLUSIONS Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry, which can be normalized by inflammation-dependent and -independent mechanisms, neither of which requires the integrity of the vagus nerve.


Infection and Immunity | 2006

GroEL of Lactobacillus johnsonii La1 (NCC 533) Is Cell Surface Associated: Potential Role in Interactions with the Host and the Gastric Pathogen Helicobacter pylori

Gabriela Bergonzelli; Dominique Granato; Raymond David Pridmore; Laure F. Marvin-Guy; Dominique Donnicola; Irène Corthesy-Theulaz

ABSTRACT Heat shock proteins of the GroEL or Hsp60 class are highly conserved proteins essential to all living organisms. Even though GroEL proteins are classically considered intracellular proteins, they have been found at the surface of several mucosal pathogens and have been implicated in cell attachment and immune modulation. The purpose of the present study was to investigate the GroEL protein of a gram-positive probiotic bacterium, Lactobacillus johnsonii La1 (NCC 533). Its presence at the bacterial surface was demonstrated using a whole-cell enzyme-linked immunosorbent assay and could be detected in bacterial spent culture medium by immunoblotting. To assess binding of La1 GroEL to mucins and intestinal epithelial cells, the La1 GroEL protein was expressed in Escherichia coli. We report here that La1 recombinant GroEL (rGroEL) binds to mucins and epithelial cells and that this binding is pH dependent. Immunomodulation studies showed that La1 rGroEL stimulates interleukin-8 secretion in macrophages and HT29 cells in a CD14-dependent mechanism. This property is common to rGroEL from other gram-positive bacteria but not to the rGroEL of the gastric pathogen Helicobacter pylori. In addition, La1 rGroEL mediates the aggregation of H. pylori but not that of other intestinal pathogens. Our in vitro results suggest that GroEL proteins from La1 and other lactic acid bacteria might play a role in gastrointestinal homeostasis due to their ability to bind to components of the gastrointestinal mucosa and to aggregate H. pylori.


Digestion | 2005

Probiotics as a Treatment Strategy for Gastrointestinal Diseases

Gabriela Bergonzelli; Stephanie Blum; Harald Brüssow; Irene Corthesy-Theulaz

Current interest in probiotics is motivated not only by the clinical data showing efficacy of some probiotic bacteria but also by the increasing antibiotic resistance of pathogenic bacteria (particularly in hospitals) and the rise of consumers’ demand for natural substitutes of drugs. Only few randomized, double-blind placebo-controlled human trials are available, and some involved only small numbers of patients. They are difficult to compare because of differences in probiotic strains employed, doses and formulation. Among probiotic applications, reduction of diarrhea is probably the best-documented effect confirmed by recent meta-analyses. Literature on Helicobacter pylori indicates that probiotics are unable to eradicate the infection but could be useful in decreasing infection levels and as adjuvants of therapy-associated side effects. Studies performed in inflammatory bowel disease suggest that high doses of probiotics and most likely a combination of different lactobacilli and bifidobacteria are more effective in decreasing inflammatory score and maintaining patients in remission than a single probiotic strain. Probiotic studies evaluating amelioration of symptoms in irritable bowel syndrome would require more sustained patient numbers. However, accumulated data is encouraging and suggests that efficacy is strain-dependent. Finally, too few probiotic intervention trials have been reported on colon cancer to allow any firm conclusion.


Journal of Pediatric Gastroenterology and Nutrition | 2006

Nutritional approach to restore impaired intestinal barrier function and growth after neonatal stress in rats.

Clara Lucia Garcia-Rodenas; Gabriela Bergonzelli; Sophie Nutten; Alexandra Schumann; Christine Cherbut; Marco Turini; Kurt Ornstein; Florence Rochat; Irène Corthesy-Theulaz

Objectives: Psychological stress during the neonatal period results in intestinal barrier dysfunction and growth alterations later in life. We aimed to restore impaired barrier function and growth rate by a nutritional intervention. Methods: Male rat pups (n = 84) were assigned to 1 of 2 rearing conditions from postnatal day (PND) 2 to PND14: S, separated 3 h/d from their mothers, or H, 15 min/d handled controls. From PND15 to PND35, rats received a control diet or a similar diet adapted to contain arachidonic and docosahexaenoic acids, galacto- and fructo-oligosaccharides and Lactobacillus paracasei NCC2461. Results: Maternal separation had only a minor impact on the measured gut barrier parameters at PND15, whereas it severely affected them at PND35. At this age, intestinal permeability to macromolecules was higher, mucin content in small intestinal tissues was lower and microbiota composition was altered in S compared with H animals. Feeding the adapted diet normalized the intestinal permeability, although it did not restore intestinal mucin content or microbiota. In addition, the adapted diet improved the growth rate recovery of the S animals after weaning and resulted in increased villus length in small intestine. Conclusion: Our results suggest that an adapted diet containing specific long-chain polyunsaturated fatty acids, prebiotics and probiotics can revert the negative imprinting of neonatal stress on both intestinal barrier function and growth.


Science Translational Medicine | 2017

Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice

De Palma G; Lynch; Jun Lu; Dang Vt; Yikang Deng; Jennifer Jury; Umeh G; Pedro M. Miranda; Pigrau Pastor M; Sacha Sidani; Maria Ines Pinto-Sanchez; Philip; P. McLean; Hagelsieb Mg; Michael G. Surette; Gabriela Bergonzelli; Elena F. Verdu; Britz-McKibbin P; Neufeld Jd; Stephen M. Collins; P. Bercik

Fecal microbiota transplants from patients with irritable bowel syndrome and anxiety alter gut function and behavior in germ-free mice. Connecting the gut-brain axis Irritable bowel syndrome (IBS), the most common gastrointestinal disorder worldwide, is characterized by abdominal pain and altered gut function and often is accompanied by anxiety. An association between intestinal dysbiosis and IBS has been reported, but the functional relevance remains unknown. De Palma and colleagues colonized germ-free mice with fecal microbiota from healthy controls or IBS patients with diarrhea (IBS-D) who did or did not have anxiety. They demonstrated that transplantation of fecal microbiota from patients with IBS-D and anxiety resulted in altered gut function and behavior in mouse recipients, including faster gastrointestinal transit, low-grade inflammation, and anxiety-like behavior. Irritable bowel syndrome (IBS) is a common disorder characterized by altered gut function and often is accompanied by comorbid anxiety. Although changes in the gut microbiota have been documented, their relevance to the clinical expression of IBS is unknown. To evaluate a functional role for commensal gut bacteria in IBS, we colonized germ-free mice with the fecal microbiota from healthy control individuals or IBS patients with diarrhea (IBS-D), with or without anxiety, and monitored gut function and behavior in the transplanted mice. Microbiota profiles in recipient mice clustered according to the microbiota profiles of the human donors. Mice receiving the IBS-D fecal microbiota showed a taxonomically similar microbial composition to that of mice receiving the healthy control fecal microbiota. However, IBS-D mice showed different serum metabolomic profiles. Mice receiving the IBS-D fecal microbiota, but not the healthy control fecal microbiota, exhibited faster gastrointestinal transit, intestinal barrier dysfunction, innate immune activation, and anxiety-like behavior. These results indicate the potential of the gut microbiota to contribute to both intestinal and behavioral manifestations of IBS-D and suggest the potential value of microbiota-directed therapies in IBS patients.


Journal of Proteomics | 2008

Maternal deprivation affects the neuromuscular protein profile of the rat colon in response to an acute stressor later in life.

Luísa V. Lopes; Laure F. Marvin-Guy; Michael Affolter; Ziad Ramadan; Martin Kussmann; Laurent B. Fay; Gabriela Bergonzelli

Early life stress as neonatal maternal deprivation (MD) predisposes rats to alter gut functions in response to acute psychological stressors in adulthood, mimicking features of irritable bowel syndrome (IBS). We applied proteomics to investigate whether MD permanently changes the protein profile of the external colonic neuromuscular layer that may condition the molecular response to an acute stressor later in life. Male rat pups were separated 3 h/day from their mothers during the perinatal period and further submitted to water avoidance (WA) stress during adulthood. Proteins were extracted from the myenteric plexus-longitudinal muscle of control (C), WA and MD+WA rat colon, separated on 2D gels, and identified by mass spectrometry. MD amplified the WA-induced protein changes involved in muscle contractile function, suggesting that stress accumulation along life imbalances the muscle tone towards hypercontractility. Our results also propose a stress dependent regulation of gluconeogenesis. Secretogranin II - the secretoneurin precursor - was induced by MD. The presence of secretoneurin in myenteric ganglia may partially explain the stress-mediated modulation of gastrointestinal motility and/or mucosal inflammation previously described in MD rats. In conclusion, our findings suggest that neonatal stress alters the responses to acute stress in adulthood in intestinal smooth muscle and enteric neurons.


Neurogastroenterology and Motility | 2013

Bifidobacterium longum NCC3001 inhibits AH neuron excitability.

A. Khoshdel; Elena F. Verdu; Wolfgang A. Kunze; P. McLean; Gabriela Bergonzelli; Jan D. Huizinga

Bifidobacterium longum (B. longum) NCC3001 can affect behavior and brain biochemistry, but identification of the cellular targets needs further investigation. Our hypothesis was that the communication with the brain might start with action on enteric sensory neurons.


Neurogastroenterology and Motility | 2017

Luminal contents from the gut of colicky infants induce visceral hypersensitivity in mice

Helene Eutamene; C. L. Garcia-Rodenas; S. Yvon; E. d'Aldebert; F. Foata; B. Berger; J. Sauser; V. Theodorou; Gabriela Bergonzelli; E. Mas

The pathophysiology of infantile colic is poorly understood, though various studies report gut microbiota dysbiosis in colicky infants. We aimed to test the hypothesis that colic‐related dysbiosis is associated with visceral hypersensitivity triggered by an altered luminal milieu.

Collaboration


Dive into the Gabriela Bergonzelli's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge