+ All Categories
Home > Documents > Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH...

Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH...

Date post: 11-Oct-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
72
ROME Università Urbaniana probiotics prebiotics new foods & 6 th September 11-13, 2011
Transcript
Page 1: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

ROMEUniversità Urbaniana

probiotics prebioticsnew foods

&6th

September 1 1 - 1 3 , 20 1 1

Page 2: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The
Page 3: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

probiotics prebioticsnew foods

&6th

L. CapursoG. Delle FaveL. MorelliA. Guarino

c ha i r p e o p l e o f t h e me e t i n g

Page 4: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

2

the meet ing i s organ ised by

OLTRE LA NUTRIZ IONE

under the patronage of

I ta l ian Academy for the study of In test ina l M icrob iota

S IGE - Soc ietà I ta l iana d i Gast roentero log ia

S INUT - Soc ietà I ta l iana d i Nutraceut ica

ESPGHAN - European Soc iety for Paed iat r i c Gast roentero logy,

Hepato logy and Nutr i t ion

S IGENP - Soc ietà I ta l iana d i Gast roentero log ia Epato log ia

e Nutr i z ione Ped iat r i ca

Centro Stud i de l l ’A l imentaz ione

Page 5: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

Sunday, September 11

Aula Magna p. 4

Monday, September 12

Aula Magna p. 6

Aula C p. 8

Aula Newman - Pediatric Day p. 12

Tuesday, September 13

Aula Magna p. 14

Aula C - Oral Communications p. 15

Aula Newman p. 19

Posters p. 20

Abstracts p. 27

Faculty p. 53

Index of Authors p. 57

General Information p. 61

Scientific Information p. 64

Exhibition Area p. 66

I N D E X

3

Page 6: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

10.00 a.m. - 12.45 p.m. ITALIAN SOCIETY OF GASTROENTEROLOGY - GUT MICROBIOTA STUDY GROUPPresident: D. Festi (Italy)Chairpeople: A. Gasbarrini (Italy), G. Capurso (Italy)

The gut barrier: a complex interplay A. Gasbarrini (Italy)

LITERATURE UP TO DATE

Gut mucous barrier E. Scarpellini (Italy)

Neuroenteric system activationR. De Giorgio (Italy)

Gut barrier and pancreatic diseasesG. Capurso (Italy)

Discussion

REPORT OF ONGOING STUDIES

Microbiota, innate system, and gastrointestinal smooth muscles: ongoing studiesC. Severi (Italy)

The pig model to study IBD-associated intestinal inflammation anddysbacteriosis: results from a preliminary studyE. Grilli (Italy)

BREAK

Morphology of segmented filamentous bacteria and their patterns of contactwith the follicle-associated epithelium of the mouse terminal ileum: Implicationsfor the relationship with the immune systemM. Caselli (Italy)

HCV and liver steatosis: viral role and dismetabolic diseasesC. Balsano (Italy)

Gut microbiota, probiotics and liver diseasesC. Loguercio (Italy)

LECTUREThe microbiota in IBSG. Barbara (Italy)

TAKE HOME MESSAGESD. Festi (Italy)

A u l a M a g n a s unday, S e p t emb er 1 1

4

Page 7: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

03.00 - 03.30 p.m. WELCOME ADDRESSL. Capurso (Italy)

03.30 - 05.30 p.m. ROUND TABLESCIENCE AND GUIDELINES:10TH YEARS OF FAO GUIDELINESChairpeople: R. Marabelli (Italy), L. Morelli (Italy)

G. Delle Fave (Italy)

F. Guarner (Spain)

Y. Sanz (Spain)

M. Serafini (Italy)

05.30 - 07.00 p.m LECTURESChairpeople: G. Delle Fave (Italy), A. Guarino (Italy)

Brain-gut axis and intestinal microbiotaS.M. Collins (Canada)

Cellular stress as sensor for luminal factors and the microbiomeD. Haller (Germany)

Functional metagenomics with relevance to host-microbe interactionsJ. Dorè (France)

WELCOME COCKTAIL

sund ay, S ep t em be r 1 1 A u l a M a gn a

5

Page 8: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTHChairman: R. Crittenden (Finland)

Microbiota in Health and DiseaseW.M. de Vos (The Netherlands)

Health Biomarker BacteriaC. Belzer (The Netherlands)

Microbiota and DietA. Salonen (Finland)

Lactobacillus GG - Life After the GenomeR. Kekkonen (Finland)

Round Table Panel: WHAT DO WE NOW KNOW AND HOW CAN WE USE THIS?R. Crittenden (Finland), C. Belzer (The Netherlands), A. Salonen (Finland), R. Kekkonen (Finland), W.M. de Vos (The Netherlands)

10.30 - 11.30 a.m. IMMUNOLOGY AND GUT BACTERIAChairpeople: M. Rescigno (Italy), F. Pallone (Italy)

Probiotics and immunoregulationC. Nicoletti (United Kingdom)

Immune modulation by probiotic bacteria in immune-compromised subjectsA. Castellazzi (Italy)

Aging, immunity and intestinal microbiotaE. Mengheri (Italy)

Fermented dairy product containing Lactobacillus casei CNCM I-1518/DN-114 001reduces the incidence of common infections and modulates the innate immuneresponse in shift workersE. Guillemard (France)

11.30 a.m. - 12.30 p.m. LACTOBACILLUS REUTERI: A SINGLE PROBIOTIC FOR SEVERAL INDICATIONSChairpeople: C. Cricelli (Italy), M. Koch (Italy)

Probiotics, Gutpain and the Nervous SystemJ. Bienenstock (Canada)

Role of Lactobacillus reuteri DSM 17938 in the uncomplicated diverticular disease A. Andriulli (Italy)

Mucosal permeability and immune activation as potential therapeutic targets ofprobiotics in irritable bowel syndromeG. Barbara (Italy)

Role of Lactobacillus reuteri DSM 17938 in the modulation of the bronchial inflammationin patients with cystic fibrosisS. Cucchiara (Italy)

12.30 - 01.30 p.m. NON BACTERIAL PROBIOTICSChairwoman: M. Elli (Italy)

Use of bacillus spores as probiotics for human useE. Ghelardi (Italy)

A u l a M a g n a Mon day, S e p t em be r 1 2

6

Page 9: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

Diet supplementation with Saccahromyces boulardii as a novel strategy to improvethe Metabolic Syndrome in an animal model of obesityI. Castagliuolo (Italy)

New clinically proven yeast probiotic in the area of IBSP. Justen (France)

01.30 - 02.30 p.m. LUNCH

02.30 - 04.00 p.m. NUTRIGENOMICS & GUT HEALTHChairpeople: P. Louis (United Kingdom), G. Perozzi (Italy)

Nutrigenomics as a tool to discover new functions for "old" molecules in chronicdisease preventionF. Virgili (Italy)

Inside the adaptation mechanisms of bifidobacteriam to the gastrointestinal environmentA. Margolles (Spain)

Dietary modulation of the human gut microbiotaP. Louis (United Kingdom)

A metagenomic approach to the “fermented food microbiota”C. Devirgiliis (Italy)

04.00 - 06.00 p.m. INTESTINAL MICROBIOTA AND IBDChairpeople: R. Caprilli (Italy), M.A. Gassull (Spain)

Engineering commensal bacteria and plants for the treatment of intestinal inflammationS. Carding (United Kingdom)

Dysbiosis in IBD and differences in microbiota composition between inflamed andnon inflamed intestineA. Walker (United Kingdom)

Colonization by faecalibacterium prausnitzii and maintenance of clinical remissionin patients with ulcerative colitisF. Guarner (Spain)

Probiotics and IBDM. Rescigno (Italy)

Clinical evidence of probiotic efficacy in IBDS. Danese (Italy)

Interactions between intestinal microbiota and innate immune system in pediatricinflammatory bowel diseaseS. Cucchiara (Italy)

06.00 - 07.00 p.m. LECTURESChairpeople: E. Corazziari (Italy), M. Anti (Italy)

Prebiotics, probiotics and obesity-related disordersH. Tilg (Austria)

Bacterial overgrowth in IBS: the role for probioticsT. Karakan (Turkey)

Mon day, S e pt em be r 1 2 A u l a M a gn a

7

Page 10: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

08.30 - 10.00 a.m. NATURAL PRODUCTS AND GUTChairpeople: D. Matteuzzi (Italy), N. Caporaso (Italy)

The modern analytical determination of complex mixtures of natural productsM. Nicoletti (Italy)

Bioactivation of dietary phytochemicals by intestinal microbiota and bifidobacteriaM. Rossi (Italy)

Polyphenols and human intestinal microflora: implications for healthG. Scapagnini (Italy)

Gut microbiota and metabolic diseasesM. Serino (Italy)

10.00 - 11.30 a.m. ORAL COMMUNICATIONS 1Chairpeople: A. Saggioro (Italy), G. Capurso (Italy)

OC 1.1 - PATHOGEN AND PROBIOTIC BACTERIA DIFFERENTIALLY STIMULATENITRIC OXIDE PRODUCTION AND S100B PROTEIN EXPRESSION IN HUMANENTEROGLIAL CELLSTurco Fabio*[1], Sarnelli Giovanni[1], Cirillo Carla[2], Mango Annamaria[1], Nasti Anna[1],D'Alessandro Alessandra[1], Farina Virginia[1], Cuomo Rosario[1]

[1]Università Federico II - Napoli, Italy - [2]K.U. Leuven - Leuven, Belgium

OC 1.2 - A PROBIOTIC COMBINATION TO REDUCE ANTIBIOTIC ASSOCIATEDDIARRHOEA AND OTHER SIDE-EFFECTS OF ANTIBIOTIC-USE: A DOSE-RESPONSE STUDYOuwehand Arthur*[1]

[1]Danisco Sweeteners - Kantvik, Finland

OC 1.3 - ANTI-INFLAMMATORY EFFECTS OF LACTOBACILLUS RHAMNOSUS(LGG) COUNTERACT LIPOPOLYSACCHARIDE (LPS)-INDUCED PERSISTENTALTERATIONS OF HUMAN COLONIC SMOOTH MUSCLEAmmoscato Francesca*[1], Matarrese Paola[2], Scirocco Annunziata[1], Petitta Chiara[1],Ascione Barbara[2], Di Natale Giuseppe[3], Marignani Massimo[4], Malorni Walter[2], SeveriCarola[1]

[1]Gastroenterology Unit A, Dip.Medicina Interna e Specialità Mediche, Università Sapienza - Roma,Italy [2]Department of Drug Research and Evaluation, Istituto Superiore di Sanità - Roma, Italy -[3]Department of Surgery, F. Durante, University ‘‘Sapienza’’ - Roma, Italy - [4]UOC Gastroenterologia,Ospedale S. Andrea - Roma, Italy

OC 1.4 - BEHAVIOUR OF ENTEROHEMORRHAGIC ESCHERICHIA COLI O157: H7IN HUMAN SIMULATED DIGESTIVE CONDITIONS AND ANTAGONISTICPROPERTIES OF A SACCHAROMYCES CEREVISIAE PROBIOTIC YEAST STRAIN Etienne-Mesmin Lucie*[1,2], Livrelli Valérie[2], Chassaing Benoit[2], Privat Maud[2], DenisSylvain[1], Alric Monique[1], Darfeuille-Michaud Arlette[2], Blanquet-Diot Stéphanie[1]]

[1]ERT 18, Equipe de Recherche Technologique «Conception, Ingénierie et Développement del’Aliment et du Médicament », Université d'Auvergne, Clermont-Ferrand, F-63000, France [2]E 2526 USC INRA 2018, Evolution des bactéries pathogènes et susceptibilité génétique de l’hôte,Université d'Auvergne, Clermont-Ferrand, F-63000, France

A u l a C Mon day, S e p t em be r 1 2

8

Page 11: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

Mon day, S e pt em be r 1 2 A u l a C

9

OC 1.5 - BIFIDOBACTERIUM LACTIS BL-04™ REDUCES SYMPTOMS OF COMMONCOLD IN HEALTHY PHYSICALLY ACTIVE INDIVIDUALS: A RANDOMISEDCONTROLLED TRIALLahtinen Sampo*[1], West Nic[2], Pyne David[2], Horn Peggy[2], Cripps Allan[3], HopkinsWill[5], Brun Mary[6], Warren Hilary[6], Wu Fan[6], Fricker Peter[2]

[1]Danisco Health & Nutrition - Kantvik, Finland - [2]Australian Institute of Sport - Canberra, Australia- [3]Griffith University - Gold Coast, Australia - [5]Auckland University - Auckland, New Zealand - [6]Canberra Hospital - Canberra, Australia

OC 1.6 - HEAT INACTIVATED PROBIOTIC STRAINS SPECIFICALLY STIMULATENFK - MAP KINASE PATHWAYS, DIFFERENT MIRNAS AND MATURATION IN CACOII ENDOTHELIAL CELLS AND IN DENDRITIC CELLSLadan Giahi[1], Eva Aumueller[1], Manuela Nestlberger[1], Ibrahim Elmadfa[1], AlexanderHaslberger*[1]

[1]Univ Vienna, Dep. Nutritional Sciences - Vienna, Austria

OC 1.7 - SUPPLEMENTATION OF YOGURT BY COMMERCIALLY AVAILABLEBACILLUS CLAUSII ENDOSPORESPal Karoly*[1], Szarvas Jozsef [ 4 ] , Hilyakne Kadlott Maria[1], Szen Orsolya[5], Naar Zoltan[1], Kiss Attila[4]

[1]Eszterhazy Karoly College, Dept. of Food Chemistry and Biochemistry - Eger, Hungary - [4]EszterhazyKaroly College, Dept. of Microbiology and Food Technology - Eger, Hungary - [5]Eszterhazy Karoly College,EGERFOOD Regional Knowledge Centre - Eger, Hungary

OC 1.8 - GALACTOOLIGOSACCHARIDES PRODUCTION FROM WHEY USINGENZYME ISOLATED FROM STREPTOCOCCUS THERMOPHILUS AND ASSESSMENTOF THEIR PREBIOTIC POTENTIALSangwan Vikas*[1], Tomar Sudhir Kumar[1], Ali Babar[1], Singh R.R.B.[1]

[1]National Dairy Research Institute - Karnal, India

OC 1.9 - DOSE-RESPONSE EFFECT OF BIFIDOBACTERIUM LACTIS HN019 ONWHOLE GUT TRANSIT TIME AND FUNCTIONAL GASTROINTESTINAL SYMPTOMSIN ADULTSOuwehand Arthur*[1], Waller Philip[2], Gopal Pramod[3], Leyer Greg[4], Reifer Cheryl[5], Stewart Morgan[5], Miller Larry[5]

[1]Danisco Sweeteners - Kantvik, Finland - [2]Accurate Clinical Research - Huston, USA - [3]Fonterra - Palmerston North, New Zealand - [4]Danisco USA - Madison, USA - [5]SPRIM - San Francisco, USA

OC 1.10 - MORPHOLOGY OF SEGMENTED FILAMENTOUS BACTERIA AND THEIRPATTERNS OF CONTACT WITH THE FOLLICLE-ASSOCIATED EPITHELIUM OF THEMOUSE TERMINAL ILEUM: IMPLICATIONS FOR THE RELATIONSHIP WITH THEIMMUNE SYSTEMCaselli M.[1], Cassol F.[1], Boldrini P.[1], Vaira D.[1], Calò G.[1]

[1]School of Gastroenterology - Department of Experimental and Clinical Medicine; University ofFerrara, Ferrara, Italy

OC 1.11 - THE PIG MODEL TO STUDY IBD-ASSOCIATED INTESTINAL INFLAMMATIONAND DYSBACTERIOSIS: RESULTS FROM A PRELIMINARY STUDYE. Grilli[1], B. Tugnoli[1], A. Zannoni[1], M. L. Bacci[1], M. Forni[1], A. Piva[1]

[1]DSMVET, Faculty of Veterinary Medicine, University of Bologna, Italy

Page 12: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

A u l a c Mon day, S e p t em be r 1 2

10

11.30 a.m - 12.30 p.m. NEW FOODS AND LIVER DISEASE: EVIDENCE IN ANIMAL MODELS Chairpeople: A.F. Attili (Italy), F. Morisco (Italy)

Coffee components and NAFLDP. Vitaglione (Italy)

Garlic and liver what news?G. D'Argenio (Italy)

Nanoparticles and food component delivery G. Peluso (Italy)

12.30 - 01.00 p.m. LECTUREFlavonoids probable made of actionM. Serafini (Italy)

01.00 - 01.30 p.m. LECTUREProbiotics and prebiotics: unconventional useM. Miraglia del Giudice (Italy)

01.30 - 02.30 p.m. LUNCH

02.30 - 04.00 p.m. NEW FOODS SESSION Chairman: V. Fogliano (Italy)

Prebiotic and cryoprotective properties of soluble fiber from hazelnut skin: effecton growth and viability of L. plantarumM. Arlorio (Italy)

Biological effects of yerba matéD. Bastos (Brazil)

Whole grain and cardiovascular healthV. Fogliano (Italy)

Effects of dietary starch on the gut microbiotaK.P. Scott (United Kingdom)

Gut in local and systemic anticancer responseL. Vannucci (Czech Republic)

Apple polyphenols and colon cancerL. Ricciardiello (Italy)

Page 13: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

Mon day, S e pt em be r 1 2 A u l a C

1 1

04.00 - 05.30 p.m. HEALTH AGEING PERSPECTIVEChairpeople: M. Calvani (Italy), F. Marotta (Italy)

Gene-Nutraceutical interplay in a healthy-aging perspective: present tips & futureavenuesF. Marotta (Italy)

Aging and menopausal transition: effects of Klamath algae extracts on well-beingand oxidative statusA.D. Genazzani (Italy)

Proteic malnutrition, aminoacids and elderlyV. Marigliano (Italy)

Exploring the role of “estrogen factor” in ageing men and its modulation throughnutraceuticsA. Polimeni (Italy)

Microbiota and healthM. Calvani (Italy)

Page 14: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

PEDIATRIC DAYWith the endorsement of ESPGHAN

Chairman: A. Guarino (Italy)

08.30 - 10.00 a.m. DETERMINANTS OF MICROFLORA IN CHILDREN AND THEIR FUNCTIONAL MODIFICATION Chairpeople: G. Buonocore (Italy), J. Vanderhoof (USA)

Molecular approaches to intestinal microfloraA. Swidsinski (Germany)

Nutrimetabolomics and its applicationsV. Fanos (Italy)

Microbiota composition and nutrition effectsP. Lionetti (Italy)

Early immunomodulation by intestinal microfloraL. de Ridder (The Netherlands)

10.00 - 10.30 a.m. LECTUREIntestinal microbiome and the metabolic consequences in neonatesJ.B.H. van Goudoever (United Kingdom)

10.30 - 11.00 a.m. COFFEE BREAK

Chairpeople: F. Indrio (Italy), F. Mosca (Italy), R. Shamir (Israel)

11.00 - 11.30 a.m. LECTUREIs early intestinal colonization a determinant of obesity?E. Isolauri (Finland)

11.30 a.m. - 12.00 p.m. LECTUREThe gut barrier: new acquisitions and therapeutical approachesA. Gasbarrini (Italy)

12.00 - 01.30 p.m. EARLY FUNCTIONAL NUTRITION

Early nutrition for the prevention of atopyC. Dupont (France)

Intestinal motility and microflora: a novel target for intestinal functional disorders(probiotics in IBS)S. Guandalini (USA)

Functional nutrition as adjunctive treatment in Cystic FibrosisE. Bruzzese (Italy)

Bifidogenics effects by prebiotics: are they clinically important?I. Hojsak (Croatia)

A u l a N ewman Mon day, S e p t em be r 1 2

12

Page 15: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

01.30 - 02.30 p.m. LUNCH

02.30 - 04.00 p.m. SAFETY AND QUALITY OF INFANT FORMULA AND ITS REGULATIONChairpeople: M. Giovannini (Italy), H. Szajewska (Poland)

IntroductionP. Aggett (United Kingdom)

True and potential dangers from nutrient-deficient formulae. Lessons from athiamine deficient formulaR. Shamir (Israel)

The role of national and international health authorities in the control of infantformulaV. Di Giorgi Gerevini (Italy)

Functional foods or functional nutrients for infants and children?C. Agostoni (Italy)

04.00 - 04.30 p.m. COFFEE BREAK

Chairpeople: M. Giovannini (Italy), J.B.H. van Goudoever (United Kingdom)

04.30 - 05.00 p.m. LECTURESupplementation of infant formula with probiotics/prebiotics: lessons learned withregard to documentation of outcomesH. Szajewska (Poland)

05.00 - 06.30 p.m. FUNCTIONAL FOODS FOR PREVENTION IN THE GENERAL POPULATION AND IN RISKGROUPS

Clinical effects of PUFA in infant formula E. Verduci (Italy)

Time course of the bifidogenic effect and its clinical implications F. Salvini (Italy)

Co-administration of antibiotics and probiotic: what is the purposeY. Vandenplas (Belgium)

Functional nutrition in preterm infants F. Indrio (Italy)

Mon day, S e pt em be r 1 2 A u l a N ewman

13

Page 16: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

08.30 - 11.30 a.m. PROBIOTICS, DIETARY PHENOLICS AND HEALTHChairpeople: A. Andriulli (Italy), M. Crespi (Italy)

Microbiota studies from European MetaHIT projectF. Guarner (Spain)

Diet, prebiotics and intestinal microbiotaK. Tuohy (United Kingdom)

Prebiotics and calcium absorptionM.L. Brandi (Italy)

Probiotic benefits for athletesM. Gleeson (United Kingdom)

The aging gut microbiota: a new perspectiveP. Brigidi (Italy)

Antiageing strategies: the role of pre-probiotics and polyphenols from red wineE. Jirillo (Italy)

Dietary phenolics in human health and disease: from molecular mechanisms to preventive and therapeutic opportunitiesD. Del Rio (Italy)

Predictive biomarkers for response to functional foodsP. Patrignani (Italy)

The microbiota and IBS; from dysbiosis to SIBO, from prebiotics and probiotics to antibiotics E. Quigley (Ireland)

11.30 a.m. - 01.00 p.m. CELIAC DISEASE SESSIONChairpeople: G.R. Corazza (Italy), M. Del Piano (Italy)

Enzyme strategies to detoxify glutenM. Rossi (Italy)

Gut microbiota analysis of italian children at-risk for celiac disease C. Catassi (Italy)

Weaning and prevention of food intoleranceM. Silano (Italy)

Gut microbes and gliadin interactions in celiac disease pathogenesisY. Sanz (Spain)

A u l a m a g n a T u e s day, S e pt em be r 1 3

14

Page 17: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

08.30 - 09.30 a.m. FUNCTIONAL MEDICINE AND PRE-PROBIOTICSChairman: A. Saggioro (Italy)

An introduction to functional medicineF. Ongaro (Italy)

Leaky gut and chronic inflammationA. Saggioro (Italy)

Prebiotics and microbiota health effectsM. Roberfroid (Belgium)

Gut microbiota links gut barrier and metabolic endotoxemia to obesity and diabetesP.D. Cani (Belgium)

09.30 - 10.30 a.m. LECTURESChairman: G. Fatati (Italy)

Synthesis "in vitro" of antioxidant compounds by enzymatic complexes isolatedfrom germinated wheatG.L. Gianfranceschi (Italy)

Probiotics and cytochromes expressionE. Bezirtzoglou (Greece)

10.30 a.m. - 01.00 p.m. ORAL COMMUNICATIONS 2Chairpeople: M. Marignani (Italy), D. Festi (Italy)

OC 2.1 - COLONIC MUSCLE CONTRACTILE ACTIVITY FOLLOWING EXPOSURETO LACTOBACILLUS RHAMNOSUS GGGuarino Michele[1], Cocca Silvia*[1], Altomare Annamaria[1], Ammoscato Francesca[2],Alloni Rossana[1], Severi Carola[2], Cicala Michele[1]

[1]Campus Bio-Medico - Roma, Italy - [2]Università degli Studi "La Sapienza" - Roma, Italy

OC 2.2 - DETECTION, IDENTIFICATION AND QUANTIFICATION OF DIFFERENTPROBIOTIC STRAINS IN FOOD BY USING A MULTIPLEX QPCRHerbel Stefan*[1], Gunther Sebastian[3], Wieler Lothar H.[2]

[1]Stefan Roland Herbel - Berlin, Germany - [2]Lothar H. Wieler - Berlin, Germany - [3], SebastianGunther - Berlin, Germany

OC 2.3 - EVALUTATION OF PROBIOTIC BACTERIAL ADHESION TO NORMAL ANDDIS PLASTIC COLONIC MUCOSA BY AN EX-VIVO ORGAN CULTUREEXPERIMENTAL MODELPagnini Cristiano*[1], Corleto Vito[1], Di Giulio Emilio[1], Delle Fave Gianfranco[1]

[1]Universita' di Roma "La Sapienza" - Roma, Italy

Tu es day, S e p t em be r 1 3 A u l a C

15

Page 18: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

OC 2.4 - LACTOBACILLUS CASEI RHAMNOSUS STRAIN GG INHIBITS THEOXIDATIVE STRESS INDUCED BY ROTAVIRUS IN HUMAN ENTEROCYTESBuccigrossi Vittoria*[1], Laudiero Gabriella[1], Sofia Morena[1], Oliva Valentina[1], VerroneMaria Antonietta[1], Wudy Anna[1], Guarino Alfredo[1]

[1]Dept. of Pediatrics University of Naples "Federico II°" - Naples, Italy

OC 2.5 - MOLECULAR BIOLOGICAL METHODS FOR RAPID DETERMINATIONOF LACTIC ACID BACTERIA STRAINS ISOLATED FROM FOOD ANDENVIRONMENTAL SAMPLESSzen Orsolya*[3], Pal Karoly[4], Naar Zoltan[4], Kiss Attila[5]

[3]Egerfood National Knowledge Centre - Eszterhazy Karoly College - Eger, Hungary -[4]Eszterhazy Karoly College - Dept. of Microbiology and Food Technology - Eger, Hungary -[5]Eszterhazy Karoly College - Dept. of Food Chemistry and Biochemistry - Eger, Hungary

OC 2.6 - PROBIOTIC STRUCTURE FUNCTION ANALYSIS REVEALS PRTP-ENCODED LACTOCEPIN TO MEDIATE ANTI-INFLAMMATORY EFFECTS VIASELECTIVE DEGRADATION OF IP-10Szen Orsolya*[3], Pal Karoly[4], Naar Hörmannsperger Gabriele*[1], von Schillde Marie-Anne[1], Weiher Monika[1], Alpert Carl-Alfred[2], Hahne Hannes[3], Bäuerl Christine[4],Perez Gaspar[4], Haller Dirk[1]

[1]Biofunctionality, Technical University of Munich - Freising-Weihenstephan, Germany -[2]Gastrointestinal microbiology, German Institute for Human Nutrition - Potsdam-Rehbrücke,Germany - [3]Chair for Bioanalytics, Technical University of Munich - Freising-Weihenstephan,Germany - [4]Departamento de Biotecnología, Instituto de Agroquímica y Tecnología deAlimentos - Valencia, Spain

OC 2.7 - PROTECTION AGAINST SEPSIS BY PROBIOTIC THERAPY ISCORRELATED WITH STIMULATION OF A NOT PREVIOUSLY DESCRIBEDBACTERIAL PHYLOTYPEGerritsen Jacoline*[1], Timmerman Harro M.[2], Fuentes Susana[1], van Minnen L. Paul[2],Panneman Henk[3], Konstantinov Sergey R.[1], Rombouts Frans M.[1], Gooszen HeinG.[2], Akkermans Louis M. A.[2], Smidt Hauke[1], Rijkers Ger T.[2]

[1]Wageningen University - Wageningen, Netherlands - [2]University Medical Center - Utrecht,Netherlands - [3]Dr. van Haeringen Laboratorium B.V. - Wageningen, Netherlands

OC 2.8 - EFFECT OF A NOVEL TRANS-GALACTOOLIGOSACCHARIDE MIXTURE(B-GOS) ON METABOLIC SYNDROME RISK FACTORS IN OVERWEIGHTADULTSVulevic Jelena*[1], Juric Aleksandra[2], Tzortzis George[2], Gibson Glenn[3]

[1]University of Reading/Clasado Ltd. - Reading, United Kingdom - [2]Clasado Ltd. - Reading,United Kingdom - [3]University of Reading - Reading, United Kingdom

OC 2.9 - EFFECTS OF ANTIBIOTIC THERAPY ON THE GASTROINTESTINALMICROBIOTA AND THE INTERVENTION WITH L.CASEIAngelika Pirker[1], Berit Hippe[1], Christoph Kamhuber[2], Felix Stockenhuber[2], Alexander Haslberger*[1]

[1]Univ. Vienna, Dep for Nutritional Sciences - Vienna, Austria - [2]Krankenhaus Oberpullendorf, -Vienna, Austria

A u l a c T u e s day, S e pt em be r 1 3

16

Page 19: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

T u e sday, S e p t embe r 1 3 A u l a c

17

OC 2.10 - GLYCOSAMINOGLYCANS OF HUMAN MILK DURING THE FIRST MONTHOF LACTATION: FURTHER POTENTIAL PREBIOTICS FOR THE BREASTFED INFANTCoppa Giovanni Valentino*[1], Gabrielli Orazio[1], Zampini Lucia[1], Galeazzi Tiziana[1],Padella Lucia[1], Bertino, Enrico[2], Maccari Francesca[3], Volpi Nicola[3]

[1]Università Politecnica Marche - Ancona, Italy - [2]Università di Torino - Torino, Italy -[3]Università Modena e Reggio Emilia - Modena, Italy

OC 2.11 - L. PLANTARUM TENSIA COMPRISING PROBIOTIC CHEESE WITHHYPOTENSIVE EFFECTHutt Pirje*[1], Songisepp Epp[2], Rätsep Merle[2], Shkut Elena[2], Zilmer Mihkel [3], EhrlichKersti[3], Mikelsaar Marika [1]

[1]University of Tartu, Dept. of Microbiology - Tartu, Estonia - [2]Bio-Competence Centre ofHealthy Dairy Products LLC - Tartu, Estonia - [3]University of Tartu, Dept. of Biochemistry -Tartu, Estonia

OC 2.12 - STUDY OF PROBIOTIC WHEY BASED ORAL REHYDRATINGSOLUTION (BIO-ORS) AGAINST SHIGELLA DYSENTERIAE INFECTION IN MICEGoyal Nupur*[1]

[1]Amity Institute of Biotechnology,Amity University - Noida, India

OC 2.13 - THERAPEUTIC EFFECTS OF OAT AND MILK BASED PROBIOTICFERMENTED PRODUCT AGAINST TYPE 2 DIABETESSangwan Seema*[1], Karasi Anbu K[1], Nanda Dhiraj K[2], Poply Sarang[1], SinghRameshwar[1]

[1]National Dairy Research Institute - Karnal, India - [2]National Bureau of Animal GeneticResources - Karnal, India

OC 2.14 - TANAGEL REDUCE COLITIS SEVERITY IN DEXTRAN SODIUMSULPHATE (DSS) MODEL OF MURINE ACUTE COLITISLopetuso Loris Riccardo*[1], Scaldaferri Franco[1], Cufino Valerio[2], Petito Valentina[2],Gerardi Viviana[1], Pizzoferrato Marco[1], Pecere Silvia[1], Laterza Lucrezia[1], StiglianoEgidio[2], Arena Vincenzo[2], Sgambato Alessandro[2], Gasbarrini Antonio[1]

[1]Internal Medicine, CATHOLIC UNIVERSITY OF ROME - Roma, Italy - [2]Pathology, CATHOLICUNIVERSITY OF ROME - Roma, Italy

OC 2.15 - MODULATION OF THE FAECAL MICROBIOTA PROFILE AND IMMUNEMARKERS BY A NOVEL TRANS-GALACOOLIGOSACCHARIDE MIXTURE (B-GOS)IN OVERWEIGHT ADULTSVulevic Jelena*[1], Juric Aleksandra[3], Tzortzis George[2], Gibson Glenn[4]

[1]University of Reading/Clasado Ltd. - Reading, United Kingdom - [2]Clasado Ltd. - Reading,United Kingdom - [3]Clasado Ltd. - Reading, United Kingdom - [4]University of Reading - Reading,United Kingdom

OC 2.16 - OBESITY-INDUCED CHANGES IN GUT MICROBIOTA ARE GENERATEDBY MUCOSAL DEFENSINS AND MODULATED BY LACTOBACILLUS CRISPATUSM247 DIET SUPPLEMENTATIONCavallo Donatella[1], Elli Marina[2], Morelli Lorenzo[3], Moratelli Ketty[1], CastagliuoloIgnazio[1], Martines Diego[1], Brun Paola*[1]

[1]Università di Padova - Padova, Italy - [2]AAT - Piacenza, taly - [3]Cattolic University -Piacenza, Italy

Page 20: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

A u l a c T u e s day, S e pt em be r 1 3

18

OC 2.17 - ADMINISTRATION OF BERBERINE IMPROVES HEPATIC NECRO-INFLAMMATION IN MURINE STEATOHEPATITIS, BUT IS ASSOCIATED WITHINCREASED MORTALITYElisa Vivoli[1], Angela Provenzano[1], Stefania Madiai[1], Erica Novo[1], Maurizio Parola[1],Fabio Marra [1]

[1]University of Florence - Florence, Italy

OC 2.18 - FACTORS INFLUENCING CLINICAL EFFICACY OF VAGINAL PROBIOTICSHeczko Piotr*[1], Strus Magdalena[1], Wiecek Grazyna[1], Kupka Anna[1], Kryczyk Jadwiga[1]

[1]Jagiellonian University Medical College - Krakow, Poland

OC 2.19 - PROBIOTIC VSL#3 MAY BE EFFECTIVE TO CHANGE THE PROFILEOF CYTOCHINES AND IMMUNOGLOBULINS IN BREAST MILK ?Baldassarre Mariella*[1], Fanelli Margherita[2], Tafaro Angela[3], Laforgia Nicola[1]

[1]Ospedale Policlinico-Neonatology and NICU, University of Bari - Bari, Italy - [2]Dept of InternalMedicin and Public Health, Section of Diagnostic Imaging, Medical Statistics, University of Bari- Bari, Italy - [3]IRCCS Ospedale "S.De Bellis" - Castellana Grotte, Italy

OC 2.20 - EFFECT OF DIETARY SUPPLEMENTATION OF A LACTOBACILLUSPLANTARUM STRAIN IN AN ARTIFICIALLY INDUCED NECROTIZINGENTEROCOLITIS MODELCastro Erica*[1], Jofre Jaime[1], Vera Rodrigo[1], Monsalvez Elizabeth[1], Pardo Karen[1],Aguayo Maria[1], Soza Francisco[1], Stillfried Nicolas[1], Medina Rossi[1], Labra Alan[1],Montecinos Hernan[1]

[1]Universidad De Concepciòn - Concepciòn, Chile

Page 21: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

T u e sd ay, S ep t em be r 1 3

19

A u l a N EWMAN

08.30 - 10.30 a.m. DERMATOLOGY SESSION Chairman: M. Picardo (Italy)

Nutrition and skinM. Picardo (Italy)

Probiotics and atopic dermatitisV. Fabiano (Italy)

Probiotics and seborrheic dermatitisC. Vincenzi (Italy)

Dietary antioxidants and photoprotectionE. Camera (Italy)

Antioxidants and skin diseasesM. Picardo (Italy)

Nutraceuticals for hair and nailsB.M. Piraccini (Italy)

Effects of Lactobacillus salivarius Ls01 (DSM 22775) treatment on atopic dermatitis in adults: randomized placebo-controlled study L. Drago (Italy)

10.30 - 11.15 a.m. LECTURESChairman: G. Scapagnini (Italy)

The use of carnitine in patients with elevated levels of Lp(a)C. Sirtori (Italy)

The role of meta-analysis in the evaluation of probiotics: arguments for and againstH. Szajewska (Poland)

11.15a.m.- 12.30 p.m. PROBIOTICS IN FOODSChairpeople: P. Aureli (Italy), G.L. Gianfranceschi (Italy)

Role of the probiotic strain Lactobacillus paracasei LMGP22043 carried by artichokes in influencing faecal bacteria and biochemical parameters in human subjectsP. Lavermicocca (Italy)

Use of Lactobacillus paracasei enriched artichokes in the treatment of functional constipationG. Riezzo (Italy)

Effects of a diet with inulin-enriched pasta on intestinal permeability in healthy young volunteersF. Russo (Italy)

Antiproliferative effects of LGG and L. paracasei on HGC-27 and DLD-1 human gastrointestinal cell linesA. Orlando (Italy)

Lactotripeptides from L. helveticum and blood pressure modulationA.F.G. Cicero (Italy)

Dysbiosis, probiotics and IBSR. Francavilla (Italy)

Page 22: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

pos t e r s

20

P1 INTERFENCES OF A SYMBIOTIC FORMULATION ON A GASTRIC AND INTESTINAL PERMEABILITYIN RATS WITH EXPERIMENTALLY INDUCED CHRONIC LIVER DAMAGECariello Rita[1], Tuccillo Concetta[1], Mazzone Giovanna[2], Ribecco Maria Teresa[2], Federico Alessandro[1], Iadevaia Maddalena[1], De Magistris Laura[1], D'Argenio Giuseppe*[2], Grossi Enzo[3], Caporaso Nicola[2],Loguercio Carmela[1]

[1]Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale, SUN - Napoli, Italy - [2]Dipartimento di MedicinaClinica e Sperimentale, Univ. Federico II - Napoli, Italy - [3]Bracco Spa, Italy

P2 EVALUATION AND GENETIC VALIDATION OF MEDIA SELECTIVE FOR BIFIDOBACTERIUMRebecchi Annalisa*[1], Pisacane Vincenza[1], Callegari Maria L.[1], Morelli Lorenzo[1]

[1]Centro Ricerche Biotecnologiche- Università Cattolica del Sacro Cuore - Cremona, Italy

P3 IN VITRO PROBIOTIC EVALUATION USING A MICROBIAL ENGINEERING APPROACH WITH THE3S-ECSIM, A 3-STAGES ENVIRONMENTAL CONTROL SYSTEM FOR INTESTINAL MICROBIOTADavid Féria-Gervasio[1], William Tottey*[1], Pascal Vandekerckove[2], Monique Alric[1], Jean-François Brugère[1]

[1]ERT-CIDAM - Clermont-Ferrand, France - [2]Lesaffre International Sarl - Marcq En Baroeul, France

P4 INVESTIGATION OF ANTIBACTERIAL ACTIVITY OF LACTOBACILLUS SPECIESSzen Orsolya*[1], Pal Karoly[2], Hilyakne Kadlott Maria[2], Naar Zoltan[2], Kiss Attila[3]

[1]Egerfood National Knowledge Centre - Eszterhazy Karoly College - Eger, Hungary - [2]Eszterhazy Karoly College -Dept. of Microbiology and Food Technology - Eger, Hungary - [3]Eszterhazy Karoly College - Dept. of Food Chemistryand Biochemistry - Eger, Hungary

P5 LACTOBACILLUS PLANTARUM TENSIA AND LACOBACILLUS PLANTARUM INDUCESANTILISTERIAL ACTIVITY IN EXPERIMENTAL CHEESERätsep Merle*[1], Smidt Imbi [2], Songisepp Epp [1]

[1]Bio-Competence Centre of Healthy Dairy Products LLC - Tartu, Estonia - [2]University of Tartu - Tartu, Estonia

P6 LACTOBACILLUS REUTERI IMPROVES THE ERADICATION RATE OF HELICOBACTER PYLORIEfrati Cesare*[1], Nicolini Giorgia[1], Cannaviello Claudio[1]

[1]Ospedale israelitico - Roma, Italy

P7 LACTOBACILLUS RHAMNOSUS LR06 DSM 21981, LACTOBACILLUS PENTOSUS LPS01 DSM21980, LACTOBACILLUS PLANTARUM LP01 LMG P-21021 AND LACTOBACILLUS DELBRUECKIISUBSP. DELBRUECKII LDD01 DSM 22106 IN VITRO STRONGLY INHIBIT DIFFERENTESCHERICHIA COLI SEROTYPES, INCLUDED E. COLI O157:H7M. Del Piano *[1], G.P. Strozzi[2], F. Deidda[3], S. Allesina[3], M. Barba[3], L. Soattini[4], F. Sforza[4], G. Mogna[2]

[1]Gastroenterology Independent Operating Unit, Maggiore della Carità Hospital - Novara, Italy - [2]Probiotical SpA -Novara, Italy - [3]Biolab Research Srl - Novara, Italy - [4]Casa di Cura I Cedri - Novara, Italy

P8 PROBIOTICS FOR PREVENTION OF NECROTIZING ENTEROCOLITIS IN PRETERM INFANTS Al Faleh Khaled*[1], Anabrees Jasim[1], Bassler D[2], Al-Kharfi T[1]

[1]King Saud University - Riyadh, Saudi Arabia - [2]U - Germany

P9 QUANTIFICATION OF LACTIC ACID AND ENTERIC BACTERIA BY MEANS OF QPCRPal Karoly*[1], Szen Orsolya[2], Naar Zoltan[1], Kiss Attila[3]

[1]Eszterhazy Karoly College, Dept. of Microbiology and Food Technology - Eger, Hungary - [2]Eszterhazy Karoly College,EGERFOOD Regional Knowledge Centre - Eger, Hungary - [3]Eszterhazy Karoly College, Dept. of Food Chemistry and Biochemistry - Eger, Hungary

Page 23: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

pos t e r s

21

P10 THE USE OF LACTOBACILLUS GG IN CHILDREN WITH FUNCTIONAL ABDOMINAL PAIN: ADOUBLE-BLIND RANDOMIZED CONTROL TRIALSabbi Tamara*[1], Palumbo Massimo[1]

[1]Belcolle Hospital Viterbo - Pediatric Unit, Italy

P11 IN VITRO EFFECT OF FOUR NOVEL FLOURS FERMENTATION ON GUT MICROBIOTA PARAMETERSChitarrari Roberto*[1], Carnevali Paola[2], Costabile Adele[1]

[1]Food Microbial Sciences, School of Food and Nutritional Sciences, University of Reading - Reading, United Kingdom- [2]Barilla G. e R. Fratelli - Parma, Italy

P12 SURVIVAL OF LACTOBACILLUS RHAMNOSUS LR06 DSM 21981, LACTOBACILLUS PENTOSUSLPS01 DSM 21980, LACTOBACILLUS PLANTARUM LP01 LMG P-21021 AND LACTOBACILLUSDELBRUECKII SUBSP. DELBRUECKII LDD01 DSM 22106 IN DIFFERENT PARTS OF THEDIGESTIVE TRACT OF PATIENTS CHRONICALLY TREATED WITH PPIM. Del Piano *[1], M. Ballarè[1], M. Pagliarulo[1], A. Anderloni[1], M. Balzarini[1], M. Orsello[1], S. Carmagnola[1],R. Tari[1], F. Deidda[3], S. Allesina[3], M. Barba[3], G.P. Strozzi[2], G. Mogna[2], L. Mogna[2], F. Sforza[4]

[1]Gastroenterology Independent Operating Unit, Maggiore della Carità Hospital, Novara - Italy - [2]Probiotical SpA,Novara, Italy - [3]Biolab Research Srl - Novara, Italy - [4]Casa di Cura I Cedri - Novara, Italy

P13 STRUCTURAL CHARACTERIZATION OF CHONDROITIN SULFATE FROM ITALIAN CHEESEPARMIGIANO REGGIANOCoppa Giovanni[1], Maccari Francesca[2], Zampini Lucia[1], Santoro Lucia[1], Galeazzi Tiziana[1], GabrielliOrazio[1], Volpi Nicola*[2]

[1]Polytechnic University of the Marche, Ospedali Riuniti, Presidio Salesi - Ancona, Italy - [2]University of Modena andReggio Emilia - Modena, Italy

P14 FLAVONOIDES AND GASTROINTESTINAL TRACT: FROM THE BENCH TO CLINICAL PERSPECTIVESMarotta Francesco*[1], Tomella Claudio[1], Polimeni Ascanio[1], Joyal Steven[2]

[1]ReGenera Res Group - Milano, Italy - [2]Life Extension Foundation - Ft. Lauderdale, USA

P15 SUPPLEMENTATION WITH LACTOBACILLUS HELVETICUS AND BIDIFOBACTERIUM LONGUMINDUCED IMMUNOLOGICAL CHANGES IN MODERATE MALNOURISHED ELDERLY SUBJECTSFinamore Alberto*[1], Roselli Marianna[1], Brasili Elisa[1], Donini Lorenzo M [2], Neri Barbara [3], CarnevaliPaola [4], Mengheri Elena[1]

[1]National Research Institute on Food and Nutrition (INRAN) - Roma, Italy - [2]Sapienza University - Rome, Italy -[3]Villa delle Querce Rehabilitation Institute - Nemi, Italy - [4]Barilla G. e R. Fratelli - Parma, Italy

P16 CAN LACTOBACILLUS RHAMNOSUS LR06 DSM 21981, LACTOBACILLUS PENTOSUS LPS01DSM 21980, LACTOBACILLUS PLANTARUM LP01 LMG P-21021 AND LACTOBACILLUSDELBRUECKII SUBSP. DELBRUECKII LDD01 DSM 22106 RESTORE THE “GASTRIC BARRIEREFFECT” IN PATIENTS CHRONICALLY TREATED WITH PPI?M. Del Piano*[1], M. Ballarè[1], M. Pagliarulo[1], A. Anderloni[1], M. Balzarini[1], M. Orsello[1], S. Carmagnola[1],R. Tari[1], F. Deidda[3], S. Allesina[3], M. Barba[3], G.P.Strozzi[2], G. Mogna[2], L. Mogna[2], F. Sforza[4]

[1]Gastroenterology Independent Operating Unit, Maggiore della Carità Hospital, Novara, Italy - [2]Probiotical SpA,Novara, Italy - [3]Biolab Research Srl, Novara, Italy - [4]Casa di Cura I Cedri, Novara, Italy

P17 CHARACTERISATION OF THE CYANOBACTERIAL TOXIN REMOVAL PROCESS IN THE PRESENCEOF PROBIOTIC BACTERIA Nybom Sonja*[1], Dziga Dariusz[2], Salminen Seppo[3], Meriluoto Jussi[1]

[1]Åbo Akademi University/Department of Biosciences - Turku, Finland - [2]Jagiellonian University - Krakow, Poland -[3]University of Turku - Turku, Finland

Page 24: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

P18 PROBIOTIC LACTOBACILLUS RHAMNOSUS LGA UPREGULATES ß-DEFENSIN AND EXPRESSIONIN CULTURED CHICKEN SMALL INTESTINAL EPITHELIAL CELLSGuanhong Li*, Siguo Liu, Zhimin Hong, Yongjie Jia, Jinming You, Minreng Qu Minreng[1]

[1]College of Animal Science and Technology, Jiangxi Agricultural University - Nanchang, China - [2]Harbin VeterinaryResearch Institute of Chinese Academy of Agricultural Sciences - Harbin, China

P19 SELENIUM AND ZINC INTERNALIZED BY LACTOBACILLUS BUCHNERI LB26 DSM 16341 ANDBIFIDOBACTERIUM LACTIS BB1 DSM 17850: DEVELOPMENT OF A NEW BIOLOGICAL METHODTO EVALUATE THE BIOAVAILABILITY OF THE TWO MINERALSM. Pane*[2], M. D'Andrea[3], S. Nicola[3], G.P. Strozzi[2], G. Mogna[2], M. Del Piano [1]

[1]Gastroenterology Independent Operating Unit, Maggiore della Carità Hospital - Novara, Italy - [2]Probiotical SpA -Novara, Italy - [3]Biolab Research Srl - Novara, Italy

P20 SAFETY OF A PROBIOTIC CHEESE COMPRISING L. PLANTARUM TENSIA ACCORDING VARIETYOF HEALTH INDICES IN DIFFERENT AGE GROUPSSongisepp Epp*[1], Hu tt Pirje [2], Rätsep Merle [1], Shkut Elena [1], Zilmer Mihkel [2], Kõljalg Siiri[2], TruusaluKai[2], Smidt Imbi[2], Kolk Helgi[2], Zagura Maksim [3], Mikelsaar Marika [2]

[1]Bio-Competence Centre of Healthy Dairy Products LLC - Tartu, Estonia - [2]University of Tartu - Tartu, Estonia -[3]Tartu University Clinics - Tartu, Estonia

P21 EFFECT OF LAB ON CYTOKINE SECRETION BY THP-1 CELLS STIMULATED BY LPSHacin Biljana*[2], Citar Manuela[2], Tompa Gorazd[1], Rogelj Irena[1]

[1]University of Ljubljana - Ljubljana, Slovenia - [2]Medis, d.o.o., Ljubljana, Slovenia

P22 SLOWL RELEASE EFFERVESCENT TABLETS WITH L. FERMENTUM LF10 DSM 19187 AND L.ACIDOPHILUS LA02 DSM 21717 INHIBIT CANDIDA AND PUTRESCENT FLORAG. Mogna[1], F. Deidda[2], S. Allesina[2], M. Pane[2], M. Barba[2], M. D’Andrea[2], P. Lorenzini[2], S. Nicola[2],E. Raiteri[2], G.P. Strozzi[1], L. Mogna[2], F. Vicariotto[3]

[1]Probiotical SpA - Novara, Italy - [2]Biolab Research Srl - Novara, Italy [3] - Department of Obstetrics and Gynecology,San Pio X Hospital - Milan, Italy

P23 SYMPTOM RESOLUTION AND IMMUNE MATURATION IN INFANTS WITH ATOPIC DERMATITISRECEIVING HYDROLYZED FORMULA WITH LACTOBACILLUS GG (LGG)Nermes Merja[1], Salminen Seppo[1], Isolauri Erika*[1]

[1]University of Turku - Turku, Finland

P24 THE GLOBAL PHENOTYPIC ANALYSIS OF PUTATIVE ANTIALLERGIC POTENTIAL OF THREELACTOBACILLUS STRAINSAleksandrzak-Piekarczyk Tamara*[1], Koryszewska-Baginska Anna[1], Bardowski Jacek[1]

[1]Institute of Biochemistry and Biophysics Polish Academy of Sciences - Warsaw, Poland

P25 IS CRANBERRY USEFUL FOR PREVENTION RECURRENT URINARY TRACT INFECTIONS INCHILDREN? Dessì Angelica*[1], Fanos Vassilios[1]

[1]Department of Paediatrics, Neonatal Intensive Care Unit, Neonatal Pathology, Puericultura Institute and NeonatalSection, University of Cagliari - Cagliari, Italy

pos t e r s

22

Page 25: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

P26 LACTOBACILLUS SPP. STRAINS OF CULTURE COLLECTION OF TARTU UNIVERSITY, ESTONIAŠtšepetova Jelena*[1], Rööp Tiiu[1], Mändar Reet[1], Sepp Epp[1], Mikelsaar Marika[1]

[1]University of Tartu - Tartu, Estonia

P27 ASSESSMENT OF SAFETY AND TOLERANCE OF A FERMENTED DAIRY FOOD CONTAININGNOVEL, POTENTIALLY PROBIOTIC STRAINS Chambaud Isabelle*[1], Jeansen Stéphanie[1], Elfakir Anissa[1], Banning Federike[2], QueudotJean- Christophe[3], Bouchez Elodie[3], Bourlioux Pierre[4], Marteau Philippe[5], Schrezenmeir Juerguen[6]

[1]Danone Research - Palaiseau, France - [2]Harrison Clinical Research Deutschland GmbH - Munich, Germany - [3]CITSafety and Health Research Laboratories - Evreux, France - [4]Faculty of Parmacy - Paris-Sud University - Paris, France- [5]University Denis Diderot, Paris 7 & AP-HP, Lariboisière ospital - Paris, France - [6]Gutenberg-University Mainz -Kiel, Germany

P28 PROBIOTIC TREATMENT INDUCED AGE DEPENDENT METABOLIC CHANGESBrasili Elisa[1], Tomassini Alberta [2], Finamore Alberto*[1], Roselli Marianna [1], Mengheri Elena[1], CapuaniGiorgio[2], Sciubba Fabio[2], Miccheli Alfredo[2]

[1]National Research Institute on Food and Nutrition, INRAN - Roma, Italy - [2]Sapienza University - Roma, Italy

P29 COMPARATIVE STUDY ON THE CELL SURFACE PROPERTIES AND STRUCTURE OFEXOPOLYSACCHARIDES PRODUCED BY LACTOBACILLUS CASEI AND LACTOBACILLUSPARACASEI STRAINSGórska-Fraczek Sabina*[1], Gamian Andrzej[1], Kozakova Hana[2], Schwarzer Martin[2]

[1]Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, - Wroclaw, Poland - [2] Departmentof Immunology and Gnotobiology, Institute of Microbiology of the Academy of Sciences of the Czech Republic, v. v.i - Novy Hradek, Czech Republic

P30 PREBIOTIC AND ANTIMICROBIAL EFFECT OF KEFIRANVardjan Tinkara*[1], Canžek Majhenic Andreja[2], Rogelj Irena[2]

[1]Kele & Kele, d.o.o. - Logatec, Slovenia - [2]University of Ljubljana, Biotechnical Faculty - Domžale, Slovenia

pos t er s

23

Page 26: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The
Page 27: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

ROME

probiotics prebioticsnew foods

&7 th

September 20 13

Page 28: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The
Page 29: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

ABSTRACTS

Page 30: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

OC1.1PATHOGEN AND PROBIOTIC BACTERIA DIFFERENTIALLYSTIMULATE NITRIC OXIDE PRODUCTION AND S100BPROTEIN EXPRESSION IN HUMAN ENTEROGLIAL CELLSTurco Fabio*[2], Sarnelli Giovanni[2], Cirillo Carla[3], MangoAnnamaria[2], Nasti Anna[2], D'Alessandro Alessandra[2],Farina Virginia[2], Cuomo Rosario[2]

[2]Università Federico II - Napoli, Italy - [3]K.U. Leuven - Leuven, Belgium

Background and aim: Enteric glial cells (EGC) are involved inintestinal homeostasis and may contribute to regulate host-bacteria interaction. Astrocytes, the equivalent of enteroglialcells (EGC) in Central Nervous System respond to bacteriareleasing nitric oxide (NO), whether this occur in bacterial-EGCinteraction and whether glial derived S100B protein is involvedin this response is not known. We aimed to investigate theeffects of pathogens and probiotics on NO release from EGC.Material and methods: Human EGC were obtained accordingto a method previously described by our group. Briefly,myenteric plexus preparations were isolated from ileum ofpatients undergoing surgery and enzimatically dissociated.Ganglia were plated and cell cultures were grown tosubconfluence. After 21 days, EGC were purified by incubationwith the anti-Thy-1.1 ab-coated magnetic beads and separatedusing a Dynal Magnet®. EGC were incubated for 24 hours withthe probiotic Lactobacillus Paracasei F19 (LP F19) and thepathogen Enteroinvasive Escherichia Coli (EIEC). 2 differentbacteria/cells ratios were used (0.1/1 and 10/1, respectively).Nitrite assay and Western Blot analysis were respectively usedto evaluate NO release and S100B expression in stimulatedcells compared to unstimulated cells that served as controls.Data are expressed as mean±SD of 3 independentexperiments. Results: Glial derived S100B protein expressionwas significantly higher in response to EIEC than to LP F19(+2.9±0.2 and +0.9±0.3 fold increase vs control; p<0.05). EIECinduced a significantly higher NO release than LP F19 both ata 0.1/1 (17.7±0.7 vs 4.0±0.1 nmol x 10^6 cells; p<0.001) andat 10/1 ratio (20.7±2.1 vs 9.0±0.1 nmol x 10^6 cells; p<0.001).Compared to control conditions (3.7±0.1 nmol x 10^6 cells),EIEC and high concentration of LP F19 induced a significantincrease of NO release (all p<0.001). Conclusions: We showthat EGC are able to release nitric oxide when challenged withbacteria and that this is dependent on the different expressionof S100B protein. As bacterial induced NO production wasdifferent between pathogens and probiotics, we suggest thathuman EGCs likely participate to host-bacteria interaction viaa different NO release and that probiotics may exertimmunostimulatory and/or immunomodulatory effectsinteracting with EGCs.

OC1.2A PROBIOTIC COMBINATION TO REDUCE ANTIBIOTICASSOCIATED DIARRHOEA AND OTHER SIDE-EFFECTS OFANTIBIOTIC-USE; A DOSE-RESPONSE STUDYOuwehand Arthur*[1]

[1]Danisco Sweeteners - Kantvik, Finland

By their nature, antibiotics kill bacteria and although narrowspectrum antibiotics are available even these will inevitablyaffect bacterial groups other then the pathogen they areintended to kill. Antibiotic consumption has therefore been

shown to negatively influence the composition andfunctionality of our intestinal microbiota. The most prominentsign of this is antibiotic associated diarrhoea, but it can alsomanifest itself in more benign side effects as bloating, gas,cramps, etc. Probiotics have in several studies shown toameliorate the risk for antibiotic associated diarrhoea (AAD);in particular Saccharomyces cerevisiae (boulardii), but alsovarious strains of lactobacillus probiotics. We have recentlyshown that a combination of lactobacilli and bifidobacteriawas able to maintain the stability of the whole microbiota ofvolunteers consuming antibiotics. We therefore aimed toinvestigate the effect of this probiotic combination on patientsseeking clinical care and being prescribed antibiotics. Werecruited 503 patients whom were randomised over threegroups receiving either a placebo (micro crystalline cellulose)or a combination of L. paracasei Lpc-37, L. acidophilusNCFM, B. lactis Bi-07 and B. lactis Bl-04 at a dose of 2.5 x109 CFU/day (Low) or 1010 CFU/day (High). Incidence andduration of diarrhoea was assessed as well as the incidenceof Clostridium difficile associated diarrhoea Subjects were50 years of age (±11 years). Older subjects were more likelyto suffer from AAD then younger subjects; also subjects witha longer exposure to antibiotics were more likely to sufferfrom AAD. Women tended to have a higher incidence of AAD,but this did not reach statistical significance. Over allincidence of AAD was 19% while overall incidence of C.difficile associated diarrhoea (CDAD) was 2.8%. There wasa significant difference in the incidence of AAD between thetreatment groups; 25%, 20% and 13%, for placebo, Low andHigh dose, respectively. There was also a significantdifference in CDAD between the treatment groups; 4.8%,1.8% and 1.8%, for placebo, Low and High dose,respectively. Also the duration of diarrhoea was significantlydifferent between groups; 5.4, 2.6 and 3.5 days (placebo,Low and High dose, respectively) as well as bloating, feverand abdominal pain. This is one of the few dose-responsestudies with probiotics; showing a dose dependent reductionin side effects of antibiotic use.

OC1.3ANTI-INFLAMMATORY EFFECTS OF LACTOBACILLUSRHAMNOSUS (LGG) COUNTERACT LIPOPOLYSACCHARIDE(LPS)-INDUCED PERSISTENT ALTERATIONS OF HUMANCOLONIC SMOOTH MUSCLEAmmoscato Francesca*[1], Matarrese Paola[2], SciroccoAnnunziata[1], Petitta Chiara[1], Ascione Barbara[2], Di NataleGiuseppe[3], Marignani Massimo[4], Malorni Walter[2], SeveriCarola[1]

[1]Gastroenterology Unit A, Dip.Medicina Interna e Specialità Mediche,Università Sapienza - Roma, Italy - [2]Department of Drug Research andEvaluation, Istituto Superiore di Sanità - Roma, Italy - [3]Department ofSurgery, ‘‘F. Durante’’, University ‘‘Sapienza’’ - Roma, Italy - [4]UOCGastroenterologia, Ospedale S.Andrea - Roma, Italy

Toll-like receptor (TLR) 2, heterodimerized with TLR1 and 6,recognizes Gram-positive bacteria and exerts anti-inflammatory effects in myocardium by inhibition of NFkBactivation through the PI3K/Akt/GSK3ß pathway. We havepreviously described that human colonic smooth musclecells (SMC) express functional TLR2 and TLR4 receptors andthat the activation of TLR4 by LPS persistently alters

ORA L C OMMUN I C AT I O N S 1

28

Page 31: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

biological and morphological SMC features whereas that ofTLR2 by LGG reduces LPS-induced effects. AIMS: to analyzecellular mechanisms involved in LGG protective role againstLPS-induced motor alterations. METHODS: The direct effectof 120x106CFU/ml LGG (ATCC 53103 strain) was tested ona highly pure human SMC culture exposed to the TLR4agonist LPS (1µg/ml) for 24h. LGG effects were evaluated onLPS-induced NFkB activation, cytokines production andmuscular morphofunctional properties. NFkB activation wasexamined by ELISA determination of the phosphorylation ofp65 NFkB subunits Ser468 and Ser536. Cytokines productionwas determined by ELISA for the pro-inflammatory IL6 andthe anti-inflammatory IL10. Data are expressed as mean±SD,p<0.05 considered significant. RESULTS: LPS induced apersistent significant (p<0.05) increase in phosphorylationof NFkB subunits (Ser468 89.9%±12.6; Ser536117.0%±15.9). In the presence of LGG, LPS-induced NFkBactivation was inhibited by up to 74.8±12.1% for Ser468 and90.3±3.0% for Ser536. Furthermore LPS induced asignificant 8-fold increase (p<0.05) in secretion of the pro-inflammatory IL6 that resulted inhibited by up of 85.6%±19.6in the presence of LGG. In parallel, LPS induced a 91%±10decrease (p<0.05) in the secretion of the anti-inflammatorycytokines IL10 whose levels return to control levels in thepresence of LGG. These LGG anti-inflammatory effectscounteract LPS-induced morpho-functional cellularalterations. In fact the LPS-induced 21.4%±1.7 cellshortening and 44.4%±7.8 decrease in acetylcholine-inducedcontraction were significantly reduced in the presence ofLGG, the cell shortening by 2.96±0.19 times and the decreasein contractile response by 1.63±0.20 times indicating aprotective role of LGG against motor alterations induced bypathogenic infective bursts. Conclusion: LGG has a directanti-inflammatory effect on colonic muscle and might be anappropriate candidate for probiotic intervention in bacterial-related intestinal motor disorders.

OC1.4BEHAVIOUR OF ENTEROHEMORRHAGIC ESCHERICHIA COLIO157:H7 IN HUMAN SIMULATED DIGESTIVE CONDITIONSAND ANTAGONISTIC PROPERTIES OF A SACCHAROMYCESCEREVISIAE PROBIOTIC YEAST STRAINEtienne-Mesmin Lucie*[1], Livrelli Valérie[2], ChassaingBenoit[2], Privat Maud[2], Denis Sylvain[1], Alric Monique[1],Darfeuille-Michaud Arlette[2], Blanquet-Diot Stéphanie[1]ERT 18, Equipe de Recherche Technologique «Conception, Ingénierieet Développement de l’Aliment et du Médicament», Universitéd'Auvergne, Clermont-Ferrand, F-63000 - France - [2]E 2526 USC INRA2018, Evolution des bactéries pathogènes et susceptibilité génétique del’hôte, Université d'Auvergne, Clermont-Ferrand, F-63000, France

Introduction/objectives: Enterohemorrhagic Escherichia coli(EHEC) O157:H7 is an important food-borne pathogen thatcauses diarrhea, hemorrhagic colitis and life-threateningcomplications such as hemolytic-uremic syndrome (HUS).Both the survival of EHEC O157:H7 and the translocation ofbacteria and Shiga-toxins (Stx) in the human digestiveenvironment are key factors in bacterial pathogenesis but themechanisms involved remain unclear owing to lack ofrelevant models. As no specific treatment is available, and asantibiotic therapy has worsened clinical outcomes, alternative

strategies using probiotics have been considered.Complementary in vitro approaches have been used to betterunderstand the behavior of EHEC in the human digestiveenvironment and investigate the antagonist properties ofSaccharomyces cerevisiae CNCM I-3856 probiotic strain.AIMS & methods: First, in vitro digestions of a standard mealcontaining ground beef inoculated with EHEC O157:H7 or EHECwith S. cerevisiae were performed in a dynamic gastro-intestinaltract model (TIM). Second, we analyzed the ability of EHECO157:H7 (i) to interact in vivo with murine Peyer’s Patches (PPs)in ileal loop assay and ex vivo in Ussing chambers and (ii) totranslocate in vitro using an M cell model. RESULTS: Bacterialmortality was observed in the stomach and duodenum of theTIM, showing the sensitivity of EHEC O157:H7 to gastricacidity and digestive secretions. By contrast, growthresumption was shown in the distal parts of the smallintestine. The co-administration of S. cerevisiae CNCM I-3856 led to a significant decrease in bacterial growth.Moreover, EHEC O157:H7 was able to target in vivo murinePPs, to translocate ex vivo through murine ileal mucosa withPPs and across an in vitro M cell model. EHEC were alsofound to survive and produce Stx within macrophages,leading to host cell apoptosis and Stx release. Once again, S.cerevisiae showed antagonistic properties against EHEC bysignificantly decreasing the number of translocated bacteriaacross M cell. Conclusion This study brought importantinformation on EHEC behaviour in simulated human digestiveenvironment. In addition, we showed that S. cerevisiae couldbe used both to reduce the amount of bacteria reaching thelarge intestine and the uptake of EHEC by M cells. Thisprobiotic yeast emerges as a relevant agent in the fightagainst EHEC infections.

OC1.5BIFIDOBACTERIUM LACTIS BL-04™ REDUCES SYMPTOMSOF COMMON COLD IN HEALTHY PHYSICALLY ACTIVEINDIVIDUALS: A RANDOMISED CONTROLLED TRIALArthur Ouwehand[1], West Nic[2], Pyne David[2], Horn Peggy[2],Cripps Allan[3], Hopkins Will[5], Brun Mary[6], WarrenHilary[6], Wu Fan[6], Fricker Peter[2]

[1]Danisco Health & Nutrition - Kantvik, Finland - [2]Australian Instituteof Sport - Canberra, Australia - [3]Griffith University - Gold Coast,Australia - [5]Auckland University - Auckland, New Zealand - [6]CanberraHospital - Canberra, Australia

Healthy, physically active individuals represent a key targetgroup for probiotics. The aim of this study was to determinethe clinical and immunological effects of Bifidobacterium lactisBl-04™ supplementation in such individuals. A total of 117males and 109 females (age 36 ± 10 y; mean ± SD) ingestedeither B. lactis Bl-04 (dosage 2x109 live cells per day; Bl-04)or placebo in powder form that was dissolved in a beveragedaily in a double-blind placebo-controlled design over 150 dthat incorporated autumn, winter and spring. Subjectsrecorded symptoms of illness (symptom type, duration andseverity), medication usage and daily physical activity patternson a Web-based self-reported questionnaire. A reduction of10% in illness symptoms was determined as the thresholdvalue for a substantial difference between treatments. URTIepisodes lasting for 5 or 7 days or more were considered tobe caused by infections. The incidence of URTI lasting 5 days

ORA L COMMU N I C AT I O N S 1

29

Page 32: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

or 7 days was reduced by 35% (CI 99% 0.33-1.29) and by46% (0.21-1.30), respectively, following Bl-04 intake.Incidence of lower respiratory tract infections lasting 5 daysor more was reduced by 45% (0.24-1.27). In line with theclinical effects, the medication use during infections lasting5 days or more was reduced by 45% (0.30-0.99). Illness totalload (severity x duration) was 27% (0.45-1.18) lower insubjects taking B. lactis Bl-04 than those on the placebo. Nosubstantial effects of supplementation were evident ongastrointestinal illness between the probiotic and placebogroups. The data from this study indicate that B. lactis BI-04has beneficial clinical effects in healthy physically activeindividuals. Other researchers report that healthy individualswant an on average 25% to 57% reduction in common coldillness severity to justify costs of treatments. Therefore, B.lactis Bl-04 supplementation in this study achieved asufficiently important difference in the reduction of upperrespiratory tract illness to meet this criterion. Furthermore,the findings of this study indicate that prophylactic use ofthis supplement could substantially reduce the economiccost of the common cold.

OC1.6HEAT INACTIVATED PROBIOTIC STRAINS SPECIFICALLYSTIMULATE NFK-, MAP KINASE PATHWAYS, DIFFERENTMIRNAS AND MATURATION IN CACO II ENDOTHELIAL CELLSAND IN DENDRITIC CELLS Ladan Giahi[1], Eva Aumueller[1], Manuela Nestlberger[1],Ibrahim Elmadfa[1], Alexander Haslberger*[1]

[1]Univ Vienna, Dep. Nutritional Sciences - Vienna, Austria

Introduction: Probiotic strains have been shown to activatecell responses involving toll like receptors (TLRs) and NF-kbpathways. Objectives and methods: We investigated effectsof heat inactivated Lactobacillus, Bifido- and Streptococcusstrains on expression of inflammatory mediators, maturationmarkers, NF-kb and miRNAs in the endothelial CACO II cellline and in dendritic cells derived from whole blood.Expression of immune- and inflammatory mediators, kbs andmiRNAs were analysed by qRT-PCR. MiRU6 was used as acontrol. Maturation markers of dendritic cells weredetermined with FACS-analysis. Results: In untreated orIL1ß- pretreated CACO2 cells the expression of IL6 and TNFaas well as NF?Bp65 and Ikb as well as p38 was increaseddifferently after treatment with heat inactivated cells ofLactobacillus acidophilus, Streptococcus thermophilus andBifidobacterium lactis. KBs showed an increased expressionin the first 60 min and decreased thereafter. LPS andprobiotic strains to different degrees decreased miRNAlevels, especially miR7i after 6hrs and 24hrs. In dendriticcells, heat inactivated Lactobacillus GG (LGG) and L.delbrueckii stimulated an enhanced expression of IL-6, TNFaand IL-10 mRNA and expression of surface molecules CD86,CD80,CD83,CD209,CD54 however to a different degree.Further, in dendritic cells the investigated strains effectedexpression of the analysed miRNAs. Discussion: Our resultsfrom CACO II cells and dendritic cells support the hypothesisthat heat inactivated cells from often closely related probioticstrains or bacteria from the gut differentially induce signalingpathways, gene expression and miRNAs which epigeneticallycontrol gene expression. Other work of our group, such as

the investigation of effects of probiotic intervention on fecalmicrobiota in antibiotic induced diarrhea, suggests quitestriking changes in the abundance of subgroups ofmicrobiota. Therefore, changes of subgroups of GI-microbiota as well as intervention with different probioticstrains should discriminate effects of bacteria on immune-or endothelial cells.

OC1.7SUPPLEMENTATION OF YOGURT BY COMMERCIALLYAVAILABLE BACILLUS CLAUSII ENDOSPORES Pal Karoly*[1], Szarvas Jozsef[4], Hilyakne Kadlott Maria[1],Szen Orsolya[5], Naar Zoltan[1], Kiss Attila[4]

[1]Eszterhazy Karoly College, Dept. of Microbiology and Food Technology- Eger, Hungary - [4]Eszterhazy Karoly College, Dept. of Food Chemistryand Biochemistry - Eger, Hungary - [5]Eszterhazy Karoly College,EGERFOOD Regional Knowledge Centre - Eger, Hungary

Development of probiotic products is based on the use ofmicrobes having numerous beneficial effects on the humanhealth. Majority of probiotic organisms belong to the groupof lactic acid bacteria, but rarely other microbes are shownto have probiotic effects, too. The endospore forming B.clausii is one of these exceptions and its endospores are usedin a commercially available product. Since no literature wasfound about the use of B. clausii in food, we investigated thesurvival of endospores and their impact on the starter cultureand ripening process of yogurt. We used fresh, warm (42 °C)yogurt samples that were inoculated by the starter cultures(Lactobacillus delbrueckii ssp. bulgaricus and Streptococcusthermophilus) 30 minutes before the experiment. Sporenumber of the B. clausii suspension was counted prior toaddition; 175 g yogurt was inoculated by 109 cfu of spores.After inoculation the yogurt cups were closed and treated thesame way as in the normal manufacturing procedure. Twoparameters of acidity, the Soxhlet-Henkel degree (°SH) andpH were measured during the 4 hours of ripening in everyhour. The yogurts were stored at 4 °C for 16 days; endosporeand total cell numbers were counted regularly. At the end ofthe experiment the yogurts were tasted and scored. Ourresults showed that addition of B. clausii endospores did nothave any significant effect on the progress of ripening. Aciditywas not altered, pH and °SH in the supplemented yogurtschanged in the same rate and degree as in the controlsamples. B. clausii endospores survived the ripening andstorage period very well. Taste, texture, colour and smell ofthe endospore enriched yogurts were very similar to that ofthe controls. This research was financed by Egerfood Ltd.and the NKTH research program.

ORA L C OMMUN I C AT I O N S 1

30

Page 33: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

OC1.8GALACTOOLIGOSACCHARIDES PRODUCTION FROM WHEYUSING ENZYME ISOLATED FROM STREPTOCOCCUSTHERMOPHILUS AND ASSESSMENT OF THEIR PREBIOTICPOTENTIALSangwan Vikas*[1], Tomar Sudhir Kumar[1], Ali Babar[1], SinghR.R.B.[1]

[1]National Dairy Research Institute - Karnal, India

1. Production of ß-galactosidase from isolated strains of S.thermophilus.2. Production of prebiotic Galactooligosaccharides (GOS)using whey as a substrate.3. Evaluation of immunomodulatory effect of GOS using miceas animal model. Materials and methods: 1. ß-galactosidase production: S. thermophilus strainsisolated from different dairy products were evaluated for theproduction of ß-galactosidase (intracellular). Five differentmethods viz. lysozyme treatment, SDS-chloroform,sonication, glass beads and microfluidizer were tried for theisolation of enzyme.2. Production of GOS: Lactose is the main substrate for theproduction of GOS. Different concentrations of lactose andß-galactosidase for varying time periods were used for theproduction of GOS. The HPLC was used for the detection ofGOS.3. Evaluation of immunomodulatory effect of GOS: CrudeGOS (produced in the lab) was then evaluated for the itsimmunomodulatory effect using mice as animal model. Alongwith the immunomodulation, it was also tested for its abilityto reduce the infection caused by Listeria monocytogenes. ResultsAmong the various methods used for the isolation of ß-galactosidase, lysozyme treatment was found to be the bestone. As it is a chemical method so cannot be used for theproduction of food grade GOS. Therefore, microfluidizer(found to be the best among mechanical methods) was usedfor the isolation of ß-galactosidase to be used in theproduction of GOS. The GOS was found to be produced at alactose conc. of 15% and at this conc. max production wasobserved after 10 hrs of incubation at 400C. The amount ofGOS production showed a gradual increase with increasinglactose concentration. In animal model GOS was found tomodulate the immune system (Increase in IgG and IgAconcentration) along with a significant reduction of L.monocytogenes infection in mice.Future projections: 1. Further standardization to increase the maximum amountof GOS production using whey as substrate is underway. 2. Evaluation of anticancerous effect of GOS.

OC1.9DOSE-RESPONSE EFFECT OF BIFIDOBACTERIUM LACTISHN019 ON WHOLE GUT TRANSIT TIME AND FUNCTIONALGASTROINTESTINAL SYMPTOMS IN ADULTSOuwehand Arthur*[1], Waller Philip[2], Gopal Pramod[3], LeyerGreg[4], Reifer Cheryl[5], Stewart Morgan[5], Miller Larry[5]

[1]Danisco Sweeteners - Kantvik, Finland - [2]Accurate Clinical Research- Huston, USA - [3]Fonterra - Palmerston North, New Zealand - [4]DaniscoUSA - Madison, USA - [5]SPRIM - San Fransisco, USA

Slow colonic transit is a common complaint in westernsocieties and manifests itself as constipation. It is, however,also associated with more serious diseases such as varioustypes of cancer, diverticulitis, gall stones, etc. Variousalternative remedies exist to relieve constipation and thesemay have various success. The present study aimed toassess the impact of Bifidobacterium lactis HN019supplementation on total colonic transit time (TCTT) andfrequency and severity of functional gastrointestinal (GI)symptoms in adults. We randomized 100 subjects (meanage: 44 years; 64% female) with functional GI symptoms toconsume a B. lactis HN019 (Danisco Cultures, Paris), at dailydoses of 17.2 billion colony forming units (CFU) (high dose;n = 33), 1.8 billion CFU (low dose; n = 33), or placebo (n =34) for 14 days. The primary endpoint of TCTT was assessedby X-ray on days 0 and 14 and was preceded by consumptionof radio-opaque markers once a day for 6 days. Thesecondary endpoint of functional GI symptom frequency wasrecorded with a subject-reported numeric (1–100) scalebefore and after supplementation. Decreases in mean TCTTover the 14-day study period were statistically significant inthe high dose group (49 ± 30 to 21 ± 32 h, p < 0.001) andthe low dose group (60 ± 33 to 41 ± 39 h, p = 0.01), but notin the placebo group (43 ± 31 to 44 ± 33 h). Time to excretionof all ingested markers was significantly shorter in thetreatment groups versus placebo. Of the nine functional GIsymptoms investigated, eight significantly decreased infrequency in the high dose group and seven decreased withlow dose, while two decreased in the placebo group. Noadverse events were reported in any group. Daily B. lactisHN019 supplementation is well tolerated, decreases TCTT ina dose-dependent manner, and reduces the frequency offunctional GI symptoms in adults. This is one of the fewdose-response studies with probiotics; showing a dosedependent reduction in colonic transit and functional GIsymptoms. Waller et al. 2011 Scand. J. Gastroenterol. DOI:10.3109/00365521.2011.584895

31

ORA L C OMMUN I C AT I O N S 1

Page 34: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

OC1.10MORPHOLOGY OF SEGMENTED FILAMENTOUS BACTERIAAND THEIR PATTERNS OF CONTACT WITH THE FOLLICLE-ASSOCIATED EPITHELIUM OF THE MOUSE TERMINALILEUM: IMPLICATIONS FOR THE RELATIONSHIP WITH THEIMMUNE SYSTEM[1]Caselli M., Cassol F., Boldrini P., Vaira D., Calò G.[1]School of Gastroenterology; Department of Experimental and ClinicalMedicine; University of Ferrara; Ferrara, Italy

Recent evidence indicates that segmented filamentousbacteria (SFB), "Candidatus Arthromitus", play a unique rolein different aspects of the maturation of the immunesystem, including T cell responses. Thus, it seemsparticularly relevant in this moment to shortly review theinformation on these bacteria and their relationship with theimmune system, and to actively investigate theirmorphological aspects. We distinguished a developmentalform from a vegetative form of these organisms. Thesedifferent forms have distinct roles in the life cycle: thedevelopmental form permits a rapid growth of theorganisms while the vegetative form permits the attachmentof SFB to the follicular epithelium. We have also givenspecial attention to the modes of contact between SFB andthe epithelial cells of the terminal ileum to better understandthe unique relationship between these bacteria and theimmune system.

OC1.11THE PIG MODEL TO STUDY IBD-ASSOCIATED INTESTINALINFLAMMATION AND DYSBACTERIOSIS: RESULTS FROM APRELIMINARY STUDYE. Grilli[1], B. Tugnoli[1], A. Zannoni[1], M.L. Bacci[1], M. Forni[1],A. Piva[1]

[1]DSMVET, Faculty of Veterinary Medicine, University of Bologna, Italy

Inflammatory Bowel Disease (IBD) is a chronic idiopathicinflammation of the gastrointestinal tract and includesCrohn’s Disease and Ulcerative Colitis. It is considered acomplex disorder caused by many factors among whichgenetic susceptibility, immune dysfunction, anddysbacteriosis.In recent years, many animal models of IBD have beendeveloped mainly in rats and small laboratory animals. Thepig, which is considered the animal specie most similar tohumans in terms of immunological development, intestinalmicrobiota, and physiology, is still under-investigatedcompared to other species. Therefore, the aim of the studywas to develop a swine model of diet-induced acute colitis. Six post-weaning pigs were assigned to two differentdietary treatments: the control group fed with a control dietand the challenged group fed with control dietsupplemented with 5% w/w of sodium dextran sulphate(DSS). Animals were daily inspected for health status, feedintake and fecal score, and were sacrificed after 7 days oftreatment. Intestinal mucosa and contents were collectedto perform histochemical analysis, inflammatory cytokinesexpression, and lactobacilli and coliforms analysis. Resultsshowed that pigs treated with DSS developed a pathologicalcondition characterized by a significant anorexia, weightloss, hyperthermia, bloody diarrhea, anemia and

leucocytosis (P<0.05). Histologically, DSS group showed amoderate inflammation, limited to the mucosal surface,associated with a damage of the basal portion of the crypts(in cecum, colon and rectum) and with infiltration ofinflammatory cells (lymphocytes, plasma cells andeosinophils; P<0.05). Microbiological analysis revealed asignificant reduction of lactobacilli and coliforms in ileum,cecum and colon contents of pigs treated with DSScompared to controls (1-3 Log CFU; P<0.05); alsoinflammatory cytokines expression patterns were altered inthe treated group compared to control. In conclusion, ourswine model showed clinical symptoms andhistopathological signs consistent with DSS-induced colitisin murine models. Further investigations will allow the useof the pig as a human model to better understand thecomplex interactions between intestinal inflammation,microbiota and intervention strategies.

ORA L C OMMUN I C AT I O N S 1

32

Page 35: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

OC2.1COLONIC MUSCLE CONTRACTILE ACTIVITY FOLLOWINGEXPOSURE TO LACTOBACILLUS RHAMNOSUS GGGuarino Michele[1], Cocca Silvia*[1], Altomare Annamaria[1],Ammoscato Francesca[2], Alloni Rossana[1], Severi Carola[2],Cicala Michele[1]

[1]Campus Bio-Medico - Roma, Italy - [2]Università degli Studi "LaSapienza" - Roma, Italy

INTRODUCTION: Clinical studies support the efficacy ofprobiotics in the treatment of acute and chronic intestinaldisorders, although mechanisms underlying are still unclear.Direct effect of probiotics on the intestinal muscle has beensuggested by preliminary findings showing that Lactobacillusstrains modulate intestinal contractility. AIMS & METHODS:We investigated the effect of Lactobacillus rhamnosus GG(LGG) on human colonic circular muscle strip (HCCMS)contractility. HCCMSs were obtained from disease-freemargins of resected segments for cancer. After removing themucosa and serosa layers, strips were mounted in separatechambers, oxygenated at 37°C. Isometric contractions weremeasured using force displacement transducers connectedwith a computer using MacLab system. The strips wereexposed to LGG obtained from the strain ATCC53103(Dicofarm spa, Rome, Italy) at different concentrations(36x109, 12x109 and 6x109 CFU/ml), to analyze contractilefrequency and amplitude and muscle response to Acetylcholine(Ach, 10-5M). LGG effects were also tested on a highly pureprimary smooth muscle cells (SMC) culture where Toll-likereceptor (TLR) 1, 2 and 6 expression was tested by real-timePCR. RESULTS: After 30 minute perfusion at 37°C, HCCMSsdeveloped a stable phasic contraction, with a significant Ach-elicited contractile response (50±8% compared to baseline).Compared to baseline, exposure to LGG induced a dose-dependent enhancement of contractile frequency and anincrease of 10.2±3%, 8±1.4% and 3±1% of mean contractileamplitude at the concentration of 36x109, 12x109 and 6x109CFU/ml of LGG, respectively. Maximum effect was reachedafter 3±2 minutes, then decreased to mean value till wash-out.Following washout, contractile frequency and amplitude as wellas response to Ach returned to baseline values. Similarly, directexposure of SMCs to LGG (120x106CFU/ml) caused dose-dependent cell shortening and inhibition of Ach contractileresponse (15%±7 and 59%±15, respectively; p<0.05). Indeed,PCR analysis showed SMC constitutive expression of TLR1, 2and 6. CONCLUSION: LGG exerts a direct and reversible effecton human colonic smooth muscle which could be related to theconstitutive expression of TLR1, 2 and 6 receptors, involved inrecognition of Gram+ bacteria, on SMCs. These effects couldpossibly explain the clinical beneficial effect of probiotics.

OC2.2DETECTION, IDENTIFICATION AND QUANTIFICATION OFDIFFERENT PROBIOTIC STRAINS IN FOOD BY USING AMULTIPLEX QPCR Herbel Stefan*[1], Günther Sebastian[3], Wieler Lothar H.[2]

[1]Stefan Roland Herbel - Berlin, Germany - [2]Lothar H. Wieler - Berlin,Germany - [3]Sebastian Günther - Berlin, Germany

Probiotic strains are often used in dairy products and inmedical pharmaceuticals. They are also fed to animals, inparticular since the European Union adopted a prohibition touse antibiotics to increase the immune defense of animalsagainst bacterial infections. In general, probiotics promotebeneficial influences on the gastrointestinal tract and therebyon health. So far the different strains of the generaLactobacillus and Bifidobacterium were isolated fromdifferent probiotic food samples and were compared withother reference strains from culture collections by phenotypiccriteria and by using the polymerase chain reaction (PCR).Present methods for the isolation of probiotic bacterials areusing selective media, utilizing different growth conditions.However, these methods are time-consuming, labor-intensive and the species differentiation via the phenotypiccharacterization is error-prone. To detect and quantifydifferent strains in a food sample, a DNA-isolation- and amolecular-based method has to be established for a species-specific characterization and quantification within a DNA-mixture of probiotic strains isolated from food or feedsamples. Therefore, the use of a usual PCR-detection methodis not feasible, as this method does not enable aquantification ability of strains in the samples analysed. Itwas shown that the formerly selected target-sequencessituated within the 23s-5s rRNA intergenic spacer regionwere not species-specific enough to differentiate close relatedprobiotic strains from each other. So other sequences likethe heat shock proteins (hsp60) had been chosen for thespecific detection and identification of different Lactobacillusstrains. For the species-specific detection of strainsbelonging to the genera Bifidobacterium we chose thesequences for the clpC, dnaJ1 and atpD proteins. In a finalstep we designed TaqMan-labelled primer pairs based on thealready existing specifically working gene sequences for thedetection of eight different probiotic species by qPCR – sixspecies of the genera Lactobacillus and two belonging to thegenera Bifidobacterium; they are all used in probiotic food.The advantage of a multiplex-qPCR method is a rapid andspecies-specific detection, identification and quantification ofdifferent probiotic strains within a single qPCR run.

ORA L COMMU N I C AT I O N S 2

33

Page 36: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

OC2.3EVALUTATION OF PROBIOTIC BACTERIAL ADHESION TONORMAL AND DISPLASTIC COLONIC MUCOSA BY AN EX-VIVO ORGAN CULTURE EXPERIMENTAL MODELPagnini Cristiano*[1], Corleto Vito[1], Di Giulio Emilio[1], DelleFave Gianfranco[1]

[1]Universita' di Roma "La Sapienza" - Roma, Italy

Background: Probiotic bacteria have shown to promotemucosal health in several pathologic conditions. The capacityto interact with the mucosa strictly depends on the adhesionof such bacteria to mucosal surface, but specific studies onadhesion properties of different bacterial species are scanty.Evidence of in-vivo colonization of specific probiotic bacteriain different colonic segments is lacking, but bacterial speciesare likely to distribute not univocally in the colon. Weintended to evaluate the adhesion property of differentprobiotic bacteria either in normal colonic mucosa and inadenomatous polyps. To this purpose, an ex-vivo organculture technique was developed. Bifidobacterium infantis andStreptococcus thermophilus, contained in a multiple probioticpreparation commercially available, were preliminary tested.Materials and methods: Biopsies of macroscopically normalmucosa from proximal and distal colon were collected uponendoscopic examination (n=6 each group). The samples werewashed and incubated with a multiple probiotic preparationfor 2h at 37°C, and then washed again. Total DNA wasextracted, and S. thermophilus and B. infantis mucosalconcentration was evaluated by real-time PCR with specificprimers. Analogue procedure was performed with biopsiesfrom adenomatous polyps and normal mucosa (n=8 each group),in the rectum-sigma. Results: After probiotic incubation, mucosa-associated concentration of B. infantis wassignificantly higher in the mucosa of proximal colon biopsiescompared with distal colon specimens (8.1±2.4 vs. 1.8±0.6,4.4-relative increment, p<0.05), while no significant differencein mucosal concentration was found for S. thermophilus.Adenomatous polyps mucosa had a significant reduction ofboth the probiotic bacteria concentration compared withnormal mucosa (B. infantis: 1.7±0.5 vs. 5.5±1.9, S.thermophilus: 13.6±8.4 vs. 70.9±24.4, p<0.01 for both), witha 3.2-fold decrement for B. infantis and a 5.2-fold decrementfor S. thermophilus. Conclusion: Preliminary results in ex-vivo organ culture experimental model indicate that differentprobiotic bacteria show peculiar adhesion to normal andpathologic colonic mucosa. Adhesion properties of probiotcbacteria appear to be important for clinical effectiveness andneed to be further investigated in experimental models.

OC2.4LACTOBACILLUS CASEI RHAMNOSUS STRAIN GG INHIBITSTHE OXIDATIVE STRESS INDUCED BY ROTAVIRUS IN HUMANENTEROCYTESBuccigrossi Vittoria*[1], Laudiero Gabriella[1], Sofia Morena[1],Oliva Valentina[1], Verrone Maria Antonietta[1], Wudy Anna[1],Guarino Alfredo[1]

[1]Dept. of Pediatrics University of Naples "Federico II°" - Naples, Italy

Background/Aim: Lactobacillus casei rhamnosus strain GG

(LGG) is an established probiotic for treatment of childhoodgastroenteritis. Rotavirus (RV) is the most severe agent ofgastroenteritis and induces a sequence of enterotoxic andcytotoxic effects in enterocytes, including oxidative stress.The aim of this study was to investigate the antioxidanteffects of LGG in RV infection in human enterocytes.Methods: We used a RV infection model developed in ourlaboratory, which consists in the infection of Caco-2 cellmonolayers with RV strain SA11 (1). Tissue integrity wasevaluated by the transepithelial resistance (TER). Reactiveoxygen species (ROS) and reduced (GSH)/oxidated (GSSG)glutathione ratio were assessed using respectivelydichlorofluorescein (DCF) and a colorimetric assay. DCF wasalso used to evaluate ROS increase by fluorescencemicroscope. We also tested the effects by the antioxidant N-acetylcysteine (NAC). Results: RV induced a significantincrease in ROS intracellular level (223±76 vs 25±19 DCFfluorescence units, p<.05) and a reduction of GSH/GSSGratio compared to controls (4.48 vs 0.08, p<.05) indicatingthat the virus alters the oxidative status and impairsantioxidant defences. The addition of NAC to Caco-2 cellsreduced RV-induced tissue damage by 68.9% (p<.05)indicating that tissue damage is oxidative stress-dependent.NAC completely inhibited RV-induced ROS increase andGSH/HSSH unbalance (p<.05). LGG prevented TER decreaseby 79.8% (p<.05). LGG counteracted RV-induced oxidativestress, reducing ROS increase by 42.8% and restoringGSH/GSSH ratio to the control level (p<.05). Microscopicevaluation confirmed the protective role of LGG in RV-induced oxidative stress. Conclusions: RV induces decreasein TER associated with ROS increase and a reduction ofGSH/GSSG ratio. LGG prevents tissue damage induced byRV by counteracting oxidative stress induced by the viralinfection. These data provide a new mechanisms for the highefficacy of LGG against childhood diarrhea observed inclinical trials.1. De Marco et al. J Infect Dis 2009;200:813

OC2.5MOLECULAR BIOLOGICAL METHODS FOR RAPIDDETERMINATION OF LACTIC ACID BACTERIA STRAINSISOLATED FROM FOOD AND ENVIRONMENTAL SAMPLESSzen Orsolya*[3], Pal Karoly[4], Naar Zoltan[4], Kiss Attila[5]

[3]Egerfood National Knowledge Centre - Eszterhazy Karoly College -Eger, Hungary - [4]Eszterhazy Karoly College - Dept. of Microbiology andFood Technology - Eger, Hungary - [5]Eszterhazy Karoly College - Dept.of Food Chemistry and Biochemistry - Eger, Hungary

The most popular methods for determination of themicrobiological composition of foods are the time consumingclassical microbiological techniques. Our primary aims were1. Building a collection of bacteria that show beneficial(probiotic) features and might be used for foodmanufacturing in the future; 2. Adapting and developingmolecular biology tools for rapid determination of isolatedbacterium strains. In our experiments we investigateddifferent samples: raw milk, fermented food products,stomach of bees and beebread. In order to select the lacticacid bacteria, we used BSM and MRS media as selectivemedium. DNA of 70 bacterial strains was extracted. At first

ORA L C OMMUN I C AT I O N S 2

34

Page 37: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

we identified the strains by traditional microbiologicalmethods and then we performed restriction analysis (RFLP)to confirm the results. We used the E8F-E1115R primer pairthat amplified an 1100 bp long fragment of the 16S rDNA.This product was digested simultaneously by the mix of threerestriction enzymes (AluI, HhaI and RsaI). Standardrestriction patterns were made by the use of known referencebacterial strains (from the DSMZ collection) and the patternsof the new isolates were compared to these. The referenceLactobacillus casei, Lb. paracasei and Lb. zeae strains (the‘Lb. casei group’) showed the same restriction pattern, so wehad to use species specific primers for the appropriatedifferentiation of these strains and the isolates having thesame pattern. In addition, we tried the ‘High ResolutionMelting’ (HRM) method for the identification of bacteria. Inour experiments we managed to identify bacterial strainsoriginating from raw milk, fermented food and bees bymeans of time-saving molecular techniques. Though theRFLP method is commonly used in laboratories, wecombined three restriction enzymes and got a much betterresolution of restriction fragments. Those bacteria that showed the restriction pattern of the ‘Lb.casei’ group were identified by species specific primers. TheHRM method was a useful tool for the separation of somestrains. We continue the search for new bacterium strains andthe improvement of our database and hope that it will help usin the rapid identification of bacteria. The research wasfinanced by Egerfood Ltd. and the NKTH research program.

OC2.6PROBIOTIC STRUCTURE FUNCTION ANALYSIS REVEALSPRTP-ENCODED LACTOCEPIN TO MEDIATE ANTI-INFLAMMATORY EFFECTS VIA SELECTIVE DEGRADATIONOF IP-10Szen Orsolya*[3], Pal Karoly[4], Naar HörmannspergerGabriele*[1], von Schillde Marie-Anne[1], Weiher Monika[1],Alpert Carl-Alfred[2], Hahne Hannes[3], Bäuerl Christine[4],Perez Gaspar[4], Haller Dirk[1]

[1]Biofunctionality, Technical University of Munich - Freising-Weihenstephan, Germany - [2]Gastrointestinal microbiology, GermanInstitute for Human Nutrition - Potsdam-Rehbrücke, Germany - [3]Chairfor Bioanalytics, Technical University of Munich - Freising-Weihenstephan, Germany - [4]Departamento de Biotecnología, Institutode Agroquímica y Tecnología de Alimentos - Valencia, Spain

Interferon-inducible protein (IP)-10 is a major chemokine forlymphocyte recruitment and known to be stronglyupregulated in the context of an array of chronicinflammatory diseases. We previously demonstrated thatVSL#3, a clinically relevant probiotic mixture in the contextof inflammatory bowel diseases (IBD), normalizes intestinalepithelial IP-10 levels via cell surface proteins ofLactobacillus paracasei (L.p). In the present study, we aimedto identify the probiotic structure-function relationshipunderlying this anti-inflammatory effect. In vitro studiesrevealed the observed loss of epithelial derived IP-10 to bedue to direct degradation of the chemokine via a highlyselective secreted and cell surface associated serine proteaseof L.p. Explant culture and intraperitoneal injectionexperiments using a murine ileitis model (TNFdeltaARE/+)

demonstrated that established IP-10 gradients in inflamedintestinal tissue can be selectively targeted by the probioticprotease, resulting in reduced ileal inflammation.Chromatographic fractionation and LC-MS/MS analysis ofthe active L.p supernatant indicated prtP-encoded lactocepinto be the protective probiotic protease, which was finallyproven by the generation of a lactocepin-negative mutant ofan analogously active, but transformable, humanLactobacillus casei (L.c) isolate (L.c lac-/-), which did nolonger show the protective anti-IP-10 activity. Physiologicallyimportant, oral probiotic treatment in a colitis model (T celltransferred RAG2-/- mice) revealed L.c to mediate reducedcecal inflammation, whereas the lactocepin negative isogenicL.c mutant is not able to exert anti-inflammatory effects. Thepresent study identified the selective degradation of IP-10 byprtP-encoded lactocepin to be a therapeutically relevantprobiotic structure-function relationship. As dysregulated IP-10 secretion is linked to an array of chronic inflammatorydiseases like IBD, allergy and arthritis, the development oflactocepin-based therapies might result in broadly applicable,safe and efficient anti-inflammatory treatment options.

OC2.7PROTECTION AGAINST SEPSIS BY PROBIOTIC THERAPY ISCORRELATED WITH STIMULATION OF A NOT PREVIOUSLYDESCRIBED BACTERIAL PHYLOTYPEGerritsen Jacoline*[1], Timmerman Harro M.[2], FuentesSusana[1], van Minnen L. Paul[2], Panneman Henk[3],Konstantinov Sergey R.[1], Rombouts Frans M.[1], GooszenHein G.[2], Akkermans Louis M. A.[2], Smidt Hauke[1], RijkersGer T.[2]

[1]Wageningen University - Wageningen, Netherlands - [2]UniversityMedical Center - Utrecht, Netherlands - [3]Dr. van HaeringenLaboratorium B.V. - Wageningen, Netherlands

Background: Prophylactic probiotic therapy has shownbeneficial effects in an experimental rat model for acutepancreatitis on the health status of the animals. Mechanismsby which probiotic therapy interfere with severity of acutepancreatitis and associated sepsis, however, are poorlyunderstood. Aims of this study were to identify the probiotic-induced changes in the intestinal microbiota and to correlatethese changes to disease outcome. Methods: Duodenum andileum samples were obtained from healthy and diseased ratssubjected to pancreatitis for seven days and prophylacticallytreated with either a multispecies probiotic mixture or placebo.Intestinal microbiota was characterized by terminal-restrictionfragment length polymorphism (T-RFLP) analyses of PCR-amplified 16S rRNA gene fragments. Results: These analysesshowed that during acute pancreatitis the host-specific ilealmicrobiota was replaced by an ‘acute pancreatitis associatedmicrobiota’. This replacement was not reversed byadministration of the probiotic mixture. An increase, however,was observed in the relative abundance of an unculturedbacterial phylotype most closely related to Clostridiumlituseburense, and referred to as commensal rat ileumbacterium (CRIB). Specific primers targeting the CRIB 16SrRNA gene sequence were developed to detect this phylotypeby quantitative PCR. An ileal abundance of CRIB 16S rRNAgenes of more than 7.5% of the total bacterial 16S rRNA gene

ORA L COMMU N I C AT I O N S 2

35

Page 38: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

pool was correlated with reduced duodenal bacterialovergrowth, reduced bacterial translocation to remote organs,improved pancreas pathology and reduced pro-inflammatorycytokine levels in plasma.Conclusion: Our current findings andfuture studies involving this uncharacterized bacterialphylotype will contribute to unraveling one of the potentialmechanisms of probiotic therapy.

OC2.8EFFECT OF A NOVEL TRANS-GALACOOLIGOSACCHARIDEMIXTURE (B-GOS) ON METABOLIC SYNDROME RISKFACTORS IN OVERWEIGHT ADULTS Vulevic Jelena*[1], Juric Aleksandra[2], Tzortzis George[2],Gibson Glenn[3]

[1]University of Reading/Clasado Ltd. - Reading, United Kingdom -[2]Clasado Ltd. - Reading, United Kingdom - [3]University of Reading -Reading, United Kingdom

Background: Metabolic syndrome is combination ofdisorders that increase the risk of developing cardiovasculardisease and diabetes. Prebiotics are known to support thegrowth of beneficial bacteria that can be reduced inoverweight adults and to also reduce cholesterol levels insome instances. However, their effects on the metabolicsyndrome risk factors in overweight adults have not beeninvestigated thus far. Objective/Design: Overweight adults(47) with three or more risk factors [increased fastingglucose and/or insulin, high blood pressure (BP),dyslipidaemia, waist circumference (>94cm in man; >84cmin women), BMI>25] associated with metabolic syndromewere randomised to receive a prebiotic GOS mixture (B-GOS)and placebo in a double-blind, placebo-controlled, crossoverstudy. They consumed both treatments daily for 12 wk witha 4 wk wash-out period in between. Blood, BP andanthropometric measurements were taken at the beginning,middle (6 wk), and end of each test period. Insulin, glucose,total cholesterol (TC), HDL-C, LDL-C and triacylglycerol(TAG) were measured. Results: B-GOS significantly reducedinsulin, TC and TAG compared with the baseline and placebo.No effect was observed on other measurements. Conclusion:B-GOS administration to overweight adults resulted inpositive effects on insulin, TC and TAG. Thus, B-GOS may beconsidered as a useful dietary candidate for the reduction ofthese metabolic syndrome risk factors in overweight adults.

OC2.9EFFECTS OF ANTIBIOTIC THERAPY ON THEGASTROINTESTINAL MICROBIOTA AND THE INTERVENTIONWITHL.CASEI Angelika Pirker[1], Berit Hippe[1], Christoph Kamhuber[2], FelixStockenhuber[2], Alexander Haslberger*[1]

[1]Univ. Vienna, Dep for Nutritional Sciences - Vienna, Austria -[2]Krankenhaus Oberpullendorf - Vienna, Austria

Background: Results from a clinical study reported reducedantibiotic associated diarrhoea (AAD, 18%:5%) byintervention with L.casei Shirota (Stockenhuber et al., 2008).Objectives: In the present pilot study effects of a combinationtherapy antibiotic and L.casei Shirota and controls weretested for AAD, C.difficile and changes of fecal microbiota in

56 patients.Study design: Stool samples from 4 groups/ 56 patients weretaken at one before and 2 timepoints after antibiotic treatment± intake of L.casei. Samples of control group were taken atsame time points. (A=antibiotic therapy; AP=antibiotic/probiotic therapy; P= control group receiving theprobiotic drink; C= Control)Methods: feacal samples were investigated for C.difficile toxinand changes of bacterial groups in GI microbiota by qPCRusing 16S rRNA group specific primers and probes,Euroclone® C.difficile A/B kit, PCR/DGGE and a C.difficileELISA test. Especially abundance and diversity of totalbacteria, Lactobacilli, Bifidobacteria, Bacteroides, Clostridiumcluster IV and XIV, C.difficile, and Enterobacteriaceae wereanalyzed.Results: 16S rRNA qPCR suggested some C.difficile in allgroups but only one C.difficile positive patient could bedetected by Euroclone® kit in group A. In group A and APwas a decrease of total bacteria following therapy.Also mean values of all bacteria were lower in A and APgroups compared to control groups.The results of PCR/DGGE showed a higher diversity in groupAP than in group A. The abundance of Enterobacteriaceaewas higher especially in group A than in control groups. Bycontrast, the abundance of Clostridium Cluster IV was lowerin both antibiotic receiving groups than in control groups.Within group P there was a significant increase ofLactobacilli. Discussion: Only qPCR tests addressing theC.difficile toxin may indicate CDI apropriately. CDI seems tobe a rare reason for AAD. To understand other pathogenicmechanisms for AAD, group specific shifts under antibiotictreatment must be better analysed. This will also lead to thedevelopment of improved probiotic approaches.

OC2.10GLYCOSAMINOGLYCANS OF HUMAN MILK DURING THEFIRST MONTH OF LACTATION: FURTHER POTENTIALPREBIOTICS FOR THE BREASTFED INFANTCoppa Giovanni Valentino*[1], Gabrielli Orazio[1], ZampiniLucia[1], Galeazzi Tiziana[1], Padella Lucia[1], Bertino Enrico[2],Maccari Francesca[3], Volpi Nicola[3]

[1]Univ. Vienna, Dep for Nutritional Sciences [1]Università politecnicaMarche - Ancona, Italy - [2]Università di Torino - Torino, Italy -[3]Università Modena e Reggio Emilia - Modena, Italy

In an our previous study (1) we defined the structure andcomposition of pooled human and bovine milkglycosaminoglycans (GAGs). Human milk GAGs (HMGs)were about 7 times higher compared to bovine milk;moreover their patterns were significantly different.Structural analyses of HMGs, revealing the presence ofglycosidic linkages resistant to the digestion by intestinalenzymes as in human milk oligosaccharides (HMOs),suggest a possible prebiotic role of such substances. As nodata are actually available, the aim of the present study wasto obtain data on HMGs concentration during the first monthof lactation.Quantitative agarose-gel electrophoresis wasperformed on milk samples collected at 4th, 10 th, 20 th and30 th day post partum from healthy mothers delivering atterm, according to (1).The mean HMGs content was 3.8, 1.0,

ORA L C OMMUN I C AT I O N S 2

36

Page 39: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

0.5 and 0.4 g/L at 4 th, 10 th, 20 th, and 30 th day of lactationrespectively. In conclusion consistent amount of GAGs wasfound during all first month of lactation; moreover a timerelated decrease of concentration have been observed, asalready described for HMOs. Because of their biochemicalcharacteristics HMGs can represent a further class of non-digestible carbohydrates with a potential prebiotic effect.

OC2.11L. PLANTARUM TENSIA COMPRISING PROBIOTIC CHEESEWITH HYPOTENSIVE EFFECTHütt Pirje*[1], Songisepp Epp[2], Rätsep Merle[2], Shkut Elena[2],Zilmer Mihkel [3], Ehrlich Kersti[3], Mikelsaar Marika [1]

[1]University of Tartu, Dept. of Microbiology - Tartu, Estonia - [2]Bio-Competence Centre of Healthy Dairy Products LLC - Tartu, Estonia -[3]University of Tartu, Dept. of Biochemistry - Tartu, Estonia

The prevalence of risk markers of atherosclerosis, incl.increased blood pressure are increasing among worldwidepopulation. Functional probiotic food could affect positivelythe metabolism of host. Some peptides, harbored in milkpossess blood pressure lowering ability through inhibition ofACE. Aim. To assess if the consumption of the L. plantarumTENSIA comprising cheese has any cardio-protective effect.Methods. The strain TENSIA (DSM 21380) originates from ahealthy Estonian child. The strain possesses some proteolyticability and is able to produce polyamines and NO in vitro. ACEinhibitory effect of the probiotic cheese extracts wasassessed spectrophotometrically at 228 nm. A DBPC cross-over study (ISRCTN15061552) was carried out with theprobiotic and control cheese. In clinically healthy adults(n=82; 37.7 ? 11.1 yrs, M/F: 33/49) the 3-week consumptionof probiotic or control cheese was followed with two-weekwashout period. Daily dose: 50 g of cheese (i.e. TENSIA 10.0log CFU). The self-reported questionnaire was applied. Fasting blood,faeces and urine were collected and blood pressure wasmeasured at recruitment, after probiotic treatment, afterwash-out and after control period. Haematological andbiochemical indices incl. serum glucose, hs-CRP, totalcholesterol, cholesterol fractions and triglycerides weredetermined. Results The consumption of cheese comprising L. plantarumTENSIA did not cause abdominal discomfort (abdominalpain, flatulence, bloating). The probiotic cheese of relativelyhigh fat content did not increase the body mass index ofvolunteers. At the end of the trial the systemic inflammationmarkers (hs-CRP, leukogram) did not show increased values,remaining within the normal range. The 3-week consumptionof probiotic cheese reduced significantly systolic anddiastolic blood pressure in adults (-3.4 mmHg and -2.4mmHg, p=0.0006, p=0.0004, respectively). Conclusion.Probiotic cheese expressing blood pressure loweringpeptides helps to maintain blood pressure supporting thecardiovascular system.

OC2.12STUDY OF PROBIOTIC WHEY BASED ORAL REHYDRATINGSOLUTION (BIO-ORS) AGAINST SHIGELLA DYSENTERIAEINFECTION IN MICE Goyal Nupur*[1]

[1]Amity Institute of Biotechnology,Amity University - Noida, India

Two BIO-ORS were prepared with whey, a dairy by-product,in combination with probiotic cultures, Lactobacillusparacasei 17 and Lactobacillus casei 299 separately.Osmolarity was maintained at 311mM/l (WHO standard).Swiss Albino mice were divided into 5 groups with twocontrols i.e. Infected (I) and Non-fermented whey (NFW)group. 109cfu/ml cells of Shigella could provoke diarrheawith soft fecal output on 2nd day and shedding at 103cfu/g.The BIO- ORS treatment in experimental groups started on2nd day. Fecal output was lowest in L.casei 299 groupswithin 5 days of the BIO-ORS treatment. On 2nd day, therewas a significant (p<0.01) increase in body weight in groups17 and 299 animals. Serum Na+ was normal (143mM/l) after3 days of drinking BIO-ORS. Higher serum K+ was observedin group 299. Loss of K+ ion in faeces was least in WHOgroup but also gets reduced to 20.7mM±5.09 and 19.5mM±1.90 in 17 and 299 group respectively. Na+ ion loss infeces was (p<0.05) lower in both BIO-ORS groups, indicatingcure against dehydration. The blood glucose level was non-significantly lowered in control groups but higher in BIO ORSgroups.On 6th day, there was a sharp increment in total lactobacillicount, although transient in feces of group 17 (8.229±0.07)and 299 (8.399±0.181). On 4th day, shedding of coliformswas less than 5 log cycle in groups 17 and 299 and highestin control groups’ i.e.7.5log cfu/g. On 5th day, there was anincrease in total LAB count in order as 299 > 17> WHO.Shedding of pathogen completely reduced (>1.5 log cfu/g)in groups 17 and 299. Colonization of aerobic bacteria insmall intestine was highest in group 17 (6.77±0.25) followedby 299. Colonization of pathogen in small intestine was least(2 log cfu/ml) in group 299 among all till 7th day.Colonization of aerobic bacteria in large intestine was highest(6.6 log cfu/ml) in 17 groups followed by 299 groups (5.60log cfu/ml). Significantly fewer viable Shigella cells weremeasured on 7th day in group 17 than 299. Colonization ofShigella in liver was reduced by 5 log units in group17. On7th day, anti-Shigella antibodies in the intestinal lumen werehigh in both BIO ORS groups facilitating the clearing ofpathogen. Moreover, lactic acid produced in the intestineadds to inhibitory effect.Nupur Goyal *(Noida,India)and D.N.Gandhi (Karnal,India)

ORA L COMMU N I C AT I O N S 2

37

Page 40: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

OC2.13THERAPEUTIC EFFECTS OF OAT AND MILK BASEDPROBIOTIC FERMENTED PRODUCT AGAINST TYPE 2DIABETESSangwan Seema*[1], Karasi Anbu K[1], Nanda Dhiraj K[2], PoplySarang[1], Singh Rameshwar[1]

[1]National Dairy Research Institute - Karnal, India - [2]National Bureauof Animal Genetic Resources - Karnal, India

In today’s modern lifestyle, where prevalence of diabetes isincreasing at an alarming rate, our aim is to develop an oatand milk based probiotic fermented product and to study itseffect on high fructose diet (HFD) induced type 2 diabetes.1st product was optimized by Response SurfaceMethodology and level of oat bran in milk, level of inoculumand incubation time for product were decided. Then oat branappropriately diluted in skim milk was steam treated forappropriate time, cooled to 370C and fermented for 7 hourswith L. rhamnosus GG (LGG) and Lactobacillus casei NCDC19 (~104 CFU/ml) separately. After fermentation, storagestudies were done for 10 weeks at 40C and it was found thatLGG fermentedproduct contain protein (2.49%), fat (0.96%),total dietary fibre (2.38%), ß-glucan (0.857%) and viabilityof Lactobacilli (1.5×107 CFU/gm). Similarly L. casei NCDC 19fermented product containprotein (2.26%), fat (1.24%), totaldietary fibre (2.43%), ß-glucan (0.831%) and viability ofLactobacilli (4.2×106 CFU/gm). Further evaluation of theseproducts was done to study their effect on progression andinduction of type 2 diabetes in HFD-fed wistar rats as animalmodel. Various blood parameters were evaluated afterfeeding for 9 weeks. Best results were shown by the ratsgroup fed with oat and milk based product fermented withLGG and it was found that this product results in a significantdecrease in blood glucose (74-76%), oxidative stress (SOD,catalase, glutathione), cholesterol (71-74%) and triglycerideslevel (63-64%) during progression study of type 2 diabetes.But during induction study, it was found that there wascomparatively less reduction in blood glucose level (52-55%), oxidative stress, cholesterol level (35-39%) andtriglycerides level (25-27%). Gene expression studiesregarding 3 genes (GLUT-4, IRS-1 and IRS-2, PPAR-?) areunderway and will be completed within 3 months.

OC2.14TANAGEL REDUCE COLITIS SEVERITY IN DEXTRAN SODIUMSULPHATE (DSS) MODEL OF MURINE ACUTE COLITISLopetuso Loris Riccardo*[1], Scaldaferri Franco[1], CufinoValerio[2], Petito Valentina[2], Gerardi Viviana[1], PizzoferratoMarco[1], Pecere Silvia[1], Laterza Lucrezia[1], StiglianoEgidio[2], Arena Vincenzo[2], Sgambato Alessandro[2],Gasbarrini Antonio[1]

[1]Internal Medicine, Catholic University of Rome - Roma, Italy -[2]Pathology, Catholic University of Rome - Roma, Italy

INTRODUCTION: Tanagel, a gelatin powder containing TannicAcids, is commonly used for diarrhea in children. Fewminformation exist on its mechanisms of action, involving gelformation and bacterial toxin sequestration, mostly obtaineby in vitro studies. No information, however, exist regardingin vivo studies and no animal model has been used to

confirm its efficacy or unravel further mechanism of action.AIMS & METHODS: Aim of this study was to evaluate thetherapeutic effect and mechanisms of action of Tanagel inthe murine model of acute colitis by DSS. C57BL/6 mice wereexposed to 2.5% Dextran Sodium Sulfate(DSS), given for 8days in tap water. At the 5th,6th, 7th, 8th day mice received1mg or 10mg of TANAGEL by gavage in 200 ul of drinkingwater; control mice received water only. Body weight, occultblood test and stool consistency were measured every dayand used to calculate the Disease Activity Index (DAI) toassess severity of colitis; survival was expressed as %. Micewere sacrificed at day 9 and colon length was measured, thencolon was opened and underwent microscopical analysis toassess the degree of inflammation. To explore the directeffects of Tanagel on intestinal epithelial cell proliferation, anMTT assay was used on CT26 and Caco2, a murine andhuman intestinal epithelial cell line, respectively. TNF –apretreated cells were incubated in presence of TNF-a(25ng/mL) or Tanagel (5, 10, 50 µg/ml) and cell vitality wasevaluated after 48 hours.RESULTS: Tanagel significantly reduced DAI in treated micecompared to controls in a dose dependent manner, being 10mg more efficacious than 1 mg dose. No differences wereassessed between the three groups about body weight loss.Neither differences in mortality were observed. At thesacrifice the lenght of colons was measured. Gelatine Tannatetreated mice showed a longer colon, compared to controls.Tanagel did not significantly affect the proliferation of Caco2and CT26 at MTT assay. Proliferation was reduced at higher(no physiological) concentrations.CONCLUSION: Taken together our preliminary observationsuggest that Tanagel decreased the clinical severity of colitisin mice. Gelatin tannate is an interesting product able to re-establish intestinal homeostasis in course of acute colitis asshown by human studies. Further analysis are required tobetter define mechanisms of action underlying these findingsand more indication for gelatin tannate could be developpedfollowing specific studies.

OC2.15MODULATION OF THE FAECAL MICROBIOTA PROFILEAND IMMUNE MARKERS BY A NOVEL TRANS-GALACOOLIGOSACCHARIDE MIXTURE (B-GOS) IN OVERWEIGHTADULTSVulevic Jelena*[1], Juric Aleksandra[3], Tzortzis George[2],Gibson Glenn[4]

[1]University of Reading/Clasado Ltd. - Reading, United Kingdom -[2]Clasado Ltd. - Reading, United Kingdom - [3]Clasado Ltd. - Reading ~United Kingdom - [4]University of Reading - Reading, United Kingdom

Background: Recent evidence suggests that gut microbiotais altered towards less beneficial one in overweightindividuals. Also, inflammation can be associated withincreased weight and altered by microbiota.Galactooligosaccharides (GOS) support the growth ofbeneficial bifidobacteria and can positively modify theimmune system, but little is known about their effect inoverweight adults.Objective/Design: We assessed the effect of a prebiotic GOSmixture (B-GOS) on faecal microbiota and immune markers

ORA L C OMMUN I C AT I O N S 2

38

Page 41: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

in a double-blind, placebo-controlled, crossover study with47 overweight subjects. They consumed both prebiotic andplacebo treatments daily for 12 wk with a 4 wk wash-outperiod in between. Blood, saliva and faeces were collected atthe beginning, middle (6 wk), and end of each test period.Predominant bacterial groups were quantified and cytokineproduction, faecal/saliva IgA, calprotectin and CRP weremeasured. Results: B-GOS significantly increased the numbers ofbeneficial bacteria, especially bifidobacteria, at the expenseof less beneficial groups compared with the baseline andplacebo. Significant decrease in CRP, calprotectin and IL-6and significant increase in faecal IgA levels were alsoobserved. Conclusion: B-GOS administration to overweightindividuals resulted in positive effects on both the microbiotacomposition and the immune response. Thus, B-GOS maybe a useful dietary candidate for the enhancement ofgastrointestinal health and immune function in overweightadults.

OC2.16OBESITY-INDUCED CHANGES IN GUT MICROBIOTA AREGENERATED BY MUCOSAL DEFENSINS AND MODULATED BYLACTOBACILLUS CRISPATUSM247DIET SUPPLEMENTATION Cavallo Donatella[1], Elli Marina[2], Morelli Lorenzo[3], MoratelliKetty[1], Castagliuolo Ignazio[1], Martines Diego[1], BrunPaola*[1]

[1]Università di Padova - Padova, Italy - [2]AAT - Piacenza, Italy -[3]Cattolic University - Piacenza, Italy

Changes in gut microbiota have been described in obesesubjects and associated to body weight gain. However it isnot clear which factor(s) are driving microbiota changes andwhether the modification of microbiota is a consequence ofobesity or rather a culprit. Taking advantage of a murine highfat diet-induced obesity model, the aims of this study wereto determine whether: 1) changes in gut microbiota precedeor follow body weight gain; 2) variation of ileal mucosaantimicrobial peptides (defensins) is associated tomodifications of intestinal microflora; 3) administration of aprobiotic strain (Lactobacillus crispatus M247) influencesdiet-induced gut microbiota alteration.Methods: male C57Bl/6 mice received either a regular chowdiet (RCD) or an high fat diet (HFD) for 9 weeks, with orwithout daily L. crispatus M247 (108 cfu) supplementation.Fecal microbiota was studied by quantitative real time PCRand Denaturing Gradient Gel Electrophoresis, whereasmucosal level of defensins, mRNA and peptides, wasdetermined by qRT-PCR and bactericidal assays,respectively.Results: A significantly increase in body weightgain was evident in mice receiving HFD as compared RCDafter the seventh week on diet. However significativechanges, both qualitative and quantitative, in gut microbiotastarted to be detectable as early as two weeks on diet. Weobserved a significant increase in Firmicutes phylumassociated to Bacteroidetes phylum reduction in mice fed aHFD, with a severe drop of Lactobacillus species. HFD-induced changes in gut microbiota paralleled a significantreduction in mucosal defensins mRNA (e.g. RegIII? andRegIIIß) that was associated to a significant loss of

antimicrobial activity on Gram+ and Gram– in vitro assays.Daily supplementation with L. crispatus M247 partiallycorrected HFD-induced changes in gut microbiota. Conclusions:changes in gut microbiota in mice receiving a HFD precede theonset of obesity and are associated to reduced defensinsproduction in the ileal mucosa,suggesting that diet can influencegut microbiota affecting the production of antimicrobial peptidesand dietary supplementation with a probiotic strain is effectiveto correct gut dysbiosis caused by an HFD.

OC2.17ADMINISTRATION OF BERBERINE IMPROVES HEPATICNECROINFLAMMATION IN MURINE STEATOHEPATITIS, BUTIS ASSOCIATED WITH INCREASED MORTALITYVivoli Elisa*[1], Provenzano Angela[1], Madiai Stefania[1], NovoErica[1], Parola Maurizio[1], Marra Fabio[1]

[1]University of Florence - Florence, Italy

Berberine (BRB) is an alkaloid present in several medicinalplant species. BRB has been shown to possess antibacterial,anti-inflammatory, and antidiabetic properties. The use ofBRB could be particularly appealing for the treatment ofnonalcoholic steatohepatitis, based on its hepatoprotectiverole during hepatotoxicity, and the ability to limit metabolicdamage and obesity. The aim of this study was to test theeffects of chronic administration of BRB in a dietary modelof murine steatohepatitis. Male C57BL/6 mice were dividedinto four groups and fed a diet deficient in methionine andcholine (MCD) or a control diet. BRB (5 mg/kg) or its vehiclewas administered intraperitoneally every day. Histology wasanalyzed by H&E staining followed by a semiquantitativescore. Intrahepatic gene expression was assayed byquantitative real time PCR. The experiment was set up toinvestigate two time points, 4 and 8 weeks. However, animalsadministered BRB together with MCD showed a remarkableexcess in mortality during the first weeks of experimentalobservation compared to mice administered MCD and vehicle(60% vs. 5% mortality at 4 weeks). Thus, all data are referredjust to the 4 week time point which included all survivinganimals. Mortality was similar comparing mice administereda control diet with or without BRB. Animals receiving MCDand BRB had significantly lower ALT levels than those onMCD+vehicle. BRB also caused a significant increase inliver/body weight ratio in MCD-fed mice. As well established,MCD induced hepatic steatosis and inflammation. The gradeof steatosis was not significantly affected by BRB. In contrast,necroinflammation was markedly and significantly reducedin mice treated with MCD+BRB vs. MCD+vehicle. MCD-induced steatohepatitis resulted in a significant increase inthe intrahepatic gene expression of monocytechemoattractant protein-1 and transforming growth factor ß.Co-administration of BRB normalized the expression levelsof these factors. Autoptic examination of animals died uponadministration of MCD+BRB demonstrated the presence ofbilateral effusions in the lungs, compatible with pneumonia.In conclusion, BRB administration effectively limits thedevelopment of necro-inflammation in mice withexperimental steatohepatitis, but this effect is overshadowedby increased mortality possibly due to deleterious actions onextrahepatic organs, such as the lungs.

ORA L COMMU N I C AT I O N S 2

39

Page 42: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

OC2.18FACTORS INFLUENCING CLINICAL EFFICACY OF VAGINALPROBIOTICSHeczko Piotr*[1], Strus Magdalena[1], Wiecek Grazyna[1],Kupka Anna[1], Kryczyk Jadwiga[1]

[1]Jagiellonian University Medical College - Krakow, Poland

Several clinical trials have been reported over the last yearsabout the efficacy of vaginal probiotic preparations. Althoughthe use of lactobacilli containing capsules appeared to begenerally beneficial for the treatment of patients with bacterialvaginosis or for prevention of its recurrences, results of thesestudies were heterogeneous mainly due to differentLactobacillus strains used and variations in study protocols.Selection of probiotic strains for vaginal delivery varied fromwell-characterized bacteria isolated from vaginal microbiotashowing strongly antagonistic properties against Gardnerellavaginalis and other vaginal pathogens to those used in dietarysupplements without marked specificity for vaginalecosystems. Also doses of the applied lactobacilli differedfrom study to study by several logarithms. Moreover, only ina few trials the presence of the applied lactobacilli in vaginaduring the observation period was proved by colonizationstudies. Protocols of the clinical studies were based on short-or long-term application of preparations regardless ofmenstrual cycles of the studied women. It is known frommicrobiological observations that vaginal microbiota issubject to great quantitative changes during and aftermenstruation. Our in vitro studies have shown that higherestrogen levels increase adherence of the vaginalLactobacillus to vaginal epithelium while those ofprogesterone negatively influence these interactions.Successful colonization of the vaginal epithelium in vitroprevented its subsequent colonization with vaginalpathogens. Thus, physiological hormonal changes may be afactor which probably strongly influences efficacy of vaginalprobiotics used to treat or prevent bacterial vaginosis inwomen at reproductive age. A critical analysis of alreadypublished clinical trials with respect to the above mentionedvariables will be made in this presentation.

OC2.19PROBIOTIC VSL#3 MAY BE EFFECTIVE TO CHANGE THEPROFILE OF CYTOCHINES AND IMMUNOGLOBULINS INBREAST MILK?Baldassarre Mariella*[1], FanelliMargherita[2], Tafaro Angela[3],Laforgia Nicola[1]

[1]Ospedale Policlinico-Neonatology and NICU, University of Bari - Bari,taly - [2]Dept of Internal Medicin and Public Health, Section of DiagnosticImaging, Medical Statistics, University of Bari - Bari, Italy - [3]IRCCSOspedale "S.De Bellis" - Castellana Grotte, Italy

Aim of study. To examine the specific effects of maternalsupplementation with the oral probiotic VSL#3 on breast milkhealth promoting agents (cytokines and immunoglobulins).Patients and methods. 18 healthy pregnant women wereincluded in the study.Half of the mothers received daily VSL#3 probiotic (VSLPharmaceuticals, Inc, Towson, MD: sachets containing 900billion live lyophilized bacteria of 4 strains of Lactobacillus,

3 strains of bifidobacteria, and 1 strain of Streptococcusthermophilus) starting from 1 month before delivery until 1month after delivery, the others were used as control group.Milk and stool samples were collected 3 days (T0) and onemonth (T30) after delivery in order to assess cytokines values(Il-6,IL-1ß, Il-10 and TGFß-1) and immunoglobulin values(IgA, IgG, IgM) in breast milk of healthy mothers andimmunoglobulins values (IgA, IgG, IgM) in stool samples oftheir newborns,.The concentrations of TGF-ß1, IL-6, IL-10and IL-1ß were analyzed using commercial kits (R & DSystem Inc., Minneapolis, MN, USA; Research DiagnosticsInc., Flandern, NJ, USA). Statistical analysis. Analysis was done by SPSS. A nonparametrical test for independent samples (U-Mann-Whitney)was performed for each variable examined.Results.TGF-ß values (pg/ml) were higher at T0 in VSL#3group (mean: 637 +/- 520 vs 381 +/-143,31, (p<0.006). andincreased significantly at T30 in the VSL#3 group (1754 +/-1151,16) but not in control group (273 +/-53,81) (p<0.001).IL10: in both groups, there was significant decrease betweenT0 and T30. No group difference was observed at T0 but atT30, IL10 levels were significantly higher in the VSL#3 group(85+/- 9,81 vs 72 +/-17,93)( p= 0.01).IL-1ß: significantlyhigher at T0 in VSL#3 group (284 +/- 448,63 vs 46 +/- 68,01)(p=0.02). Immunoglobulins. IgA and IgM levels were significantlyhigher (p<0.05) at T30 and IgG levels were significantlyhigher at both T0 and T30 in the milk samples of the VSL#3group (p<0.05). Regarding the stool samples, IgM levelswere significantly higher at T0 and T30 and IgA and IgG weresignificantly higher at T30 in the VSL#3 group (p<0.05)These data indicate that VSL#3 may be a powerfulimmunomodulator in terms of cytokines andimmunoglobulin production, mostly at mucosal surface.Preliminary experiments also indicate that VSL#3 canrepresent a good supplement in the diet of pregnant women.

OC2.20EFFECT OF DIETARY SUPPLEMENTATION OF A LACTOBACILLUSPLANTARUM STRAIN IN AN ARTIFICIALLY INDUCEDNECROTIZING ENTEROCOLITIS MODELCastro Erica*[1], Jofre Jaime[1], Vera Rodrigo[1], MonsalvezElizabeth[1], Pardo Karen[1], Aguayo Maria[1], Soza Francisco[1],Stillfried Nicolas[1], Medina Rossi[1], Labra Alan[1], MontecinosHernan[1]

[1]Universidad De Concepciòn - Concepciòn, Chile

Necrotizing enterocolitis (NEC) is the principal cause of deathfrom gastrointestinal disease in premature neonates and isassociated with long-term complications in survivingpatients. The primary focus of research in NEC has shifted tothe prevention and treatment of the disease. Our hypothesisproposed that one Lactobacillus plantarum strain coulddecreases the intestinal injuries caused for an artificiallyinduced NEC´s model.The experimental protocol was approved by the EthicalCommittee of the University of Concepción. Twelve newbornSprague Dawley pups were used in this study. Theexperimental groups were: i) Control group: Pups artificiallyfed with infant formula (Similac NeosureTM, Abbott); ii)

ORA L C OMMUN I C AT I O N S 2

40

Page 43: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

Probiotic group: Pups artificially fed with the same infantformula containing 109 CFU/dose of probiotic strain. Eachgroup was fed with 300 µl of food formula every three hours,by using an adapted urethral catheter. All experimentalgroups were exposed to asphyxia/cold stress, 60 seconds in100% N2, followed by cold stress 4°C for 10 minutes (twicedaily) to develop experimental NEC. At signs of illness or atthe end of the experimental period (96 hrs), animals wereeuthanized and their small intestines carefully removed andevaluated for typical signs of NEC, microbiological andhistologically. A standardized scoring system was developedto describe the NEC´s severity.The histological evaluation showed severe damage in bothtreatments. The entire control group showed transmuralnecrosis and hereby classified as grade 4. The probioticgroup, even if showed severe damage too, the grade of NECwas comparatively lower, given that three probiotic fed pupsshowed mid or complete villous necrosis and herebyclassified as grade 2 and 3. One animal had a completerecovery. The mortality was similar in both groups. SGR werenegative and not significantly differences between groupswere observed. The LABs counts in the probiotic were higherthan the control group (2,76x108 and 2,32x106 CFU/grespectively).We conclude that the artificially induced NEC model waseffectively established in all pups, and the probiotic strainslightly decreases the injury´s grade in newborn pups.However, we propose some improvements to our modelmainly in apply more effective feeding methods that ensurethe correct incorporation of the probiotic in the animals withlesser stress levels. Research financed by INNOVA Chileproject 09CN14-5919.

ORA L COMMU N I C AT I O N S 2

41

Page 44: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

P1INTERFENCES OF A SYMBIOTIC FORMULATION ON A GASTRICAND INTESTINAL PERMEABILITY IN RATS WITHEXPERIMENTALLY INDUCED CHRONIC LIVER DAMAGECariello Rita[1], Tuccillo Concetta[1], Mazzone Giovanna[2],Ribecco Maria Teresa[2], Federico Alessandro[1], IadevaiaMaddalena[1], De Magistris Laura[1], D'Argenio Giuseppe*[2],Grossi Enzo[3], Caporaso Nicola[2], Loguercio Carmela[1]

[1]Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale,SUN - Napoli, Italy - [2]Dipartimento di Medicina Clinica e Sperimentale,Univ. Federico II - Napoli, Italy - [3]Bracco Spa, Italy

Background & Aim: we have previously reported that liverdamage was significantly reduced by treatment with asymbiotic preparation (Lactobacillus paracasei FB, Bracco,Spa) in a rat model of liver fibrosis induced by CCl4.This effectwas associated with improvement of biochemical parametersand reduced liver inflammation. Aim of the present study was to go deeper inside into theeffects of FB on liver fibrosis with particular interest in thegastrointestinal permeability. Methods: We examined 10Wistar rats with liver fibrosis induced by twice a week CCl4i.p injection for 7 weeks, 10 rats with liver fibrosis treated dailywith symbiotic FB (200mg/kg/die, by gavage), and 10 normalrats receiving symbiotic treatment alone. A further group ofnormal rats was considered as control. In all animals gastro-intestinal permeability was assessed by means of multipleprobes test (urinary recovery of sucrose-mannitol-lactulose);liver damage and inflammation were documented by routinelylaboratory tests and histology for tissue collagen depositionwas also assessed. Results: The administration of CCl4significantly increased gastric permeability in respect to basalvalue (p<0.001), while the treatment with FB significantlydecreased it (p<0.01). Unexpectedly, CCl4 reduced intestinalpermeability compared to normal rats, and this reduction waseven more marked when these rats were treated withsymbiotic preparation. Interestingly, FB administered tonormal rats significantly reduced intestinal permeability. Inother terms CCl4 induces liver damage without enhancingintestinal permeability, that, on the contrary, results loweredby it, and is further reduced by symbiotic treatment. The otherparameters were all improved by FD treatment.Conclusions. The treatment with CCl4 induces, in rats, achronic liver damage, and also less known effects, such asincrease of gastric and decrease of intestinal permeability,through mechanisms not well defined. Bacterial translocation(BT) is one of the mechanisms involved both in the inductionand progression of chronic liver disease. BT isn’t synonimusof altered intestinal permeability and in course of cirrhosisexperimentally induced in rats with CCl4, BT may be due tochanges on the surfactant and brush border intestinalmembranes, changes in the luminal gut flora, which mightalter adherence to the mucosa, and/or by a significantoxidative stress in the intestine that precedes the damage atthe cellular level and that affects the mucosal barrier. Asymbiotic such as FB per se modifies gastric and intestinalmucosa, without effects on liver function. In presence of liverdamage, a chronic treatment with this symbiotic formulationimproves liver fibrosis and inflammation and also affects bothgastric and intestinal permeability.

P2EVALUATION AND GENETIC VALIDATION OF MEDIA SELECTIVEFOR BIFIDOBACTERIUMRebecchi Annalisa*[1], Pisacane Vincenza[1], Callegari Maria L.[1],Morelli Lorenzo[1]

[1]Centro Ricerche Biotecnologiche- Università Cattolica del Sacro Cuore -Cremona, Italy

The selective enumeration of Bifidobacteria is of great interestfor the study of intestinal microbiota. During the routine analysisthe differentiation of bifidobacteria is made difficult by the highpresence of lattobacilli.In our study three selective media for the detection andenumeration of bifidobacteria were compared and evaluated fortheir efficiency in the recovery of bifidobacteria from pigletstools. The used media were: Beerens agarsupplemented withnalidixic acid (15mg/ml), Bifidus selective medium (BSM, Fluka)supplemented with a mixture of antibiotics andTransoligosaccharide propionate agar medium (TOS, Merck)modified by addition of mupirocin (50 mg/ml). On Beerens andTOS media the bifidobacteria colonies shown a white/creamcolour whereas on BSM medium the colonies of bifidobacteriaare violet/brown. Supplementation used for all these mediashould inhibit the lactobacilli growth.Thirty-five stools samples were tested and 380 colonies wereisolated following the indications of the manufacture’sinstructions. For example on BSM medium only violet colonieswere picked up. Difference in count number on the three differentmedia: in particular the Beerens supplemented medium gave thehigher number of colonies (ranging from 108 to 109 cfu/g),around one log less was detected on BSM mediumand the lowervalue was obtained on TOS modified medium (ranging from 105to106 cfu/g). In order to confirm that isolated colonies belongedto the Bifidobacterium genera a PCR reaction with primers Bif164-Bif 662 described by Satokari et al. 2001, were performed.On Beerens medium we detected only presence oflactobacilli, onBSM and TOS medium colonies of bifidobacteria represented the20% and 75 % of the total colonies respectively. Moreover TOSmedium allowed the isolations of several species of bifidobacteriaindeed after identifications isolated colonies belonged to thefollowing species in order to frequency: Bifidobacteriumpseudolongum, Bifidobacterium tsurumiense, Bifidobacteriumthermophilum, Bifidobacterium choerinum, Bifidobacteriumthermacidophilum, Bifidobacterium coryneforme. On BSMmedium only the first two species were isolated. On the basis ofour results the TOS medium could be a good candidate for theisolation of bifidobacteria from a complex ecosystem such as thefaecal microbiota.

P3IN VITRO PROBIOTIC EVALUATION USING A MICROBIALENGINEERING APPROACH WITH THE 3S-ECSIM, A 3-STAGES ENVIRONMENTAL CONTROL SYSTEM FORINTESTINAL MICROBIOTADavid Féria-Gervasio[1], William Tottey*[1], PascalVandekerckove[2], Monique Alric[1], Jean-François Brugère[1]

[1]ERT-CIDAM - Clermont-Ferrand, France - [2]Lesaffre International Sarl- Marcq En Baroeul, France

A microbial engineering approach was applied to evaluate themetabolic impact of probiotics Bacillus subtilis (Bs) and

pos t e r s

42

Page 45: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

Lactobacillus plantarum (Lp) on the human gut microbiota,using an alternative in-vitro fermentation system calledEnvironmental Control System for Intestinal Microbiota(ECSIM). This system can simulate the human gut microbiotaunder various nutritional and environmental conditions. Thestrategy applied to evaluate probiotics using ECSIMconsisted of the introduction, during microbiota anaerobicmetabolism with/without substrate limitation, of a localperturbation by the addition of probiotics, and in the analysisof the consequences of such a perturbation on the humangut microbiota’s metabolism and structure. The probioticsconcentration feeding in the bioreactor was set at 2x109CFU/day added once in batch cultures and in continuous flowin the proximal section of the 3S-ECSIM at D=0.004h-1(eachprobiotic was studied individually). Feeding the cultures withprobiotics Bs and Lp led to modifications of the microbialmetabolism in-vitro. Results showed a perturbation of themetabolism caused by the introduction of two probiotics inanaerobic batch and continuous culture systems. Differenceswere observed in the biomass production, specific biomassproduction, gas production, short chain fatty acids (SCFA)and bacteria concentration. Those results were confirmed byvarying profiles observed with molecular fingerprintingtechniques. The probiotics used did not show implantationin the human microbiota in-vitro. However, a decrease of thespecific biomass and of the gas production was observed inthe presence of both probiotics. The probiotic Bs in cultureswithout substrate limitation led to a decrease in the shortchain fatty acids (SCFA) production and also affected theBacteroides population. On the other hand, in cultures withsubstrate limitation in presence of Lp and Bs, Bacteroidesand Bifidobacteria population increased and SCFA observeddecreased. In conclusion, this microbial engineeringapproach combined with appropriate analytical techniquesprovides an alternative and efficient means to study theeffects of probiotics in the human microbiota in itsconstitution, metabolism and adaptation to environmentaland particularly nutritional changes.

P4INVESTIGATION OF ANTIBACTERIAL ACTIVITY OFLACTOBACILLUS SPECIESSzen Orsolya*[1], Pal Karoly[2], Hilyakne Kadlott Maria[2], NaarZoltan[2], Kiss Attila[3]

[1]Egerfood National Knowledge Centre - Eszterhazy KarolyCollege - Eger, Hungary - [2]Eszterhazy Karoly College - Dept.of Microbiology and Food Technology - Eger, Hungary - [3]EszterhazyKaroly College - Dept. of Food Chemistry and Biochemistry - Eger, Hungary

Research of lactic acid bacteria gets more and more attentionfrom the consumers and producers, as well. In addition totheir well-known effects, some Lactobacillus strains haveanother useful feature: production of antibacterial proteins,which prevent the growth and multiplication of food spoilageGram-positive bacteria, thus may represent alternatives toartificial preservatives. Lactic acid bacteria are widely used inthe production of fermented products. The primary use oflactic acid bacterial strains that might produce bacteriocin-like proteins is to supplement the commonly used startercultures in order to produce antimicrobial agents. Weinvestigated the antimicrobial activity of nearly 100 lactic acid

bacteria strains that were isolated from food products andenvironmental samples. After identification we tested theseisolates on lawn of sensitive bacteria (related LAB strains anddifferent species that cause food spoilage). Two strains, a L.plantarum and a L. acidophilus were selected, since theseshowed the strongest inhibition against the most sensitivestrains. In the next step we investigated whether antibacterialsubstrates or pH-lowering activity of organic acids wereresponsible for the inhibition effect. By PCR we could detectthe plantaricin gene in four amongst the six isolated L.plantarum. We did not find evidence for the presence ofantimicrobial peptides produced by the L. acidophilus, so inthis case presumably the low pH was the reason of theinhibition. Production of bacteriocins requires certainenvironmental conditions and most likely induction by signalmolecules. At the present we are working on thedetermination of those parameters that affect thebiosynthesis of plantaricin.

P5LACOBACILLUS PLANTARUM TENSIA AND LACOBACILLUSPLANTARUM INDUCIA ANTILISTERIAL ACTIVITY INEXPERIMENTAL CHEESERätsep Merle*[1], Smidt Imbi [2], Songisepp Epp [1]

[1]Bio-Competence Centre of Healthy Dairy Products LLC - Tartu, Estonia -[2]University of Tartu - Tartu,Estonia

Background: the food-borne illnesses are one of the mostwidespread health problems. The most common bacteria thatcause food-borne infections are Salmonella, Listeria andCampylobacter. Some studies show inhibition of these entericpathogens by indigenous lactobacilli of human origin andprobiotic Lactobacillus strains. Objective: the aim was toinvestigate the influence of Lactobacillus plantarum Tensia DSM21380 and L. plantarum Inducia DSM 21379 as adjunct startersin Edam-type cheese to the viability of L. monocytogenes andSCFA profile. The probiotic strains Tensia and Inducia possessstrong antagonistic activity against food-borne pathogens invitro and produce plantaricins. Material and methods: The semi-hard Edam-type cheeses were prepared from cow milk with0.8-1 % of the precultured starter C92 (CSK Food EnrichmentNederlands) Different additives (adjunct probiotical starterand/or Listeria monocytogenes) were used: one with onlyprobiotic adjunct (in count of 5 log10CFU/g); with probioticstarter and Listeria monocytogenes; and control cheese withonly Listreria monocytogenes (in count of 4-5 log10CFU/g).Antilisterial activity of probiotic strains was assessed by viabilityof added listeria. The SCFA profile was determined by gaschromatography (HP 6890 Series GC System). Results: Thecount of added lactobacilli strains Tensia and Inducia increasedby two logarithms during the four week ripening period. ThepH decrease of experimental cheeses was characteristic for thattype of cheese. Listeria count in cheese with probiotic adjunctwas significantly lower (p=0.03) in both probiotic cheeses thanin the control cheese No differences were found in SCFA profilein experimental cheeses with dfferent additives, but succinicacid amount was significantly higher (p<0,01) in probioticcheese compared to cheeses comprising only listeria.Conclusions: Both probitic strains have good viablility in cheeseenvironment and are able to sustain the antimicrobial activityin food matrix.

pos te r s

43

Page 46: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

P6LACTOBACILLUS REUTERI IMPROVES THE ERADICATIONRATE OF HELICOBACTER PYLORIEfrati Cesare*[1], Nicolini Giorgia[1], Cannaviello Claudio[1]

[1]Ospedale israelitico - Roma, Italy

Several studies report an inhibitory effect of probiotics onHelicobacter pylori. In particular, Lactobacillus Reuteri (LR)exerts a competition of binding to glycolipid receptor of andsuppresses H. pylori urease activity. The aim of this study isto evaluate whether adding LR to antibiotic therapy mayimprove the H. pylori eradication rate. The H. pylori infectionwas diagnosed in 90 adult dyspeptic patients. Patients wereexcluded if previously treated for H. pylori infection and ifthey were taking PPI, H2-receptor antagonist or antibioticsin the four weeks preceding the study. Patients were assignedto receive one of the following therapies: (a) 7- day triple (PPIplus clarithromycin and amoxicillin or metronidazole) plusLR supplementation during the antibiotic treatment; (b) 7-day triple plus LR supplementation after the antibiotictreatment; (c) sequential regimen (5-day PPI plus amoxicillintherapy followed by a 5-day PPI, clarithromicin andtinidazole) plus LR supplementation during the antibiotictreatment; (d) sequential regimen plus LR supplementationafter the antibiotic treatment. Successful eradication therapywas defined as a negative UBT performed at least 4 weeksfollowing the eradication treatment. Eighty-three patients (30males and 53 females; mean age 57 ± 13 years) completedthe study. Nineteen patients were administrated a 7-day tripletreatment: 11 with LR supplementation during and 8 aftertherapy. Sixty-four patients were administered sequentialregimen: 32 with LR supplementation during and 32 aftertherapy. At the end of the study, the whole eradication ratewith the LR supplementation was 82% versus 74-76 %reported in literature without probiotics. Furthermore, in ourpopulation the eradication rate was significantly higher in thesequential group compared with the 7-day triple (88 % vs 63%; p = 0.01). The results of our study confirmed that as thefirst-line, the sequential therapy seems more efficiency thanstandard 7-day, in the H.pylori eradication. The LRsupplementation appears improve the H.pylori eradicationrate, although large, double-bind, controlled studies areneeded to confirm these results.

P7LACTOBACILLUS RHAMNOSUS LR06 DSM 21981,LACTOBACILLUS PENTOSUS LPS01 DSM 21980,LACTOBACILLUS PLANTARUM LP01 LMG P-21021 ANDLACTOBACILLUS DELBRUECKII SUBSP. DELBRUECKIILDD01 DSM 22106 IN VITRO STRONGLY INHIBIT DIFFERENTESCHERICHIA COLI SEROTYPES, INCLUDED E. COLIO157:H7 Del Piano M.*[1], Strozzi G.P.[2], Deidda F.[3], Allesina S.[3],Barba M.[3], Soattini L.[4], Sforza F.[4], Mogna G.[2]

[1]Gastroenterology Independent Operating Unit, Maggiore della CaritàHospital, Novara, Italy - [2]Probiotical SpA, Novara, Italy - [3]BiolabResearch Srl, Novara, Italy - [4]Casa di Cura I Cedri, Novara, Italy

P8PROBIOTICS FOR PREVENTION OF NECROTIZINGENTEROCOLITIS IN PRETERM INFANTS Al Faleh Khaled*[1], Anabrees Jasim[1], Bassler D[2], Al-Kharfi T[1]

[1]King Saud University - Riyadh, Saudi Arabia - [2]U - Germani, Germany

Background: Necrotizing enterocolitis (NEC) and nosocomialsepsis are associated with increased morbidity and mortalityin preterm infants. Through prevention of bacterialmigrationacross themucosa, competitive exclusion of pathogenicbacteria, and enhancing the immune responses of the host,prophylactic enteral probiotics may play a role in reducingNEC and associated.morbidity. Objectives: To compare the efficacy and safety ofprophylactic enteral probiotics administration versus placeboor no treatment in the prevention of severe NEC and/or sepsisin preterm infants. Methods: Searches were made ofMEDLINE (1966 to October 2010), EMBASE (1980 to October2010), CENTRAL (Cochrane Library, Issue 2, 2010), andabstracts of annual meetings of SPR (1995 to 2010).Only randomized or quasi-randomized controlled trials thatenrolled preterm infants < 37 weeks gestational age and/or< 2500 g birth weight were considered. Trials were includedif they involved enteral administration of any live microbialsupplement (probiotics) and measured at least oneprespecified clinical outcome. Standard methods of theCochrane Collaboration and its Neonatal Group were used toassess the methodologic quality of the trials, data collectionand analysis. Results Sixteen eligible trials randomizing 2842infants were included. Included trials were highly variablewith regard to enrollment criteria (i.e. birth weight andgestational age), baseline risk of NEC in the control groups,timing, dose, formulation of the probiotics, and feedingregimens. Data regarding extremely low birth weight infants(ELBW) could not be extrapolated. In a meta-analysis of trialdata, enteral probiotics supplementation significantly reducedthe incidence of severe NEC (stage II or more) (typical RR0.35, 95% CI 0.24 to 0.52) and mortality (typical RR 0.40,95% CI 0.27 to 0.60). There was no evidence of significantreduction of nosocomial sepsis (typical RR 0.90, 95% CI 0.76to 1.07). The included trials reported no systemic infectionwith the probiotics supplemental organism. The statisticaltest of heterogeneity for NEC, mortality and sepsis wasinsignificant. Conclusions: Enteral supplementation ofprobiotics prevents severe NEC and all cause mortality inpreterm infants. Our updated review of available evidencesupports a change in practice. More studies are needed toassess efficacy in ELBW infants and assess the most effectiveformulation and dose to be utilized.

P9QUANTIFICATION OF LACTIC ACID AND ENTERIC BACTERIABY MEANS OF QPCRPal Karoly*[1], Szen Orsolya[2], Naar Zoltan[1], Kiss Attila[3]

[1]Eszterhazy Karoly College, Dept. of Microbiology and Food Technology- Eger, Hungary - [2]Eszterhazy Karoly College, EGERFOOD RegionalKnowledge Centre, Eger, Hungary - [3]Eszterhazy Karoly College, Dept.of Food Chemistry and Biochemistry - Eger, Hungary

Beneficial features of probiotic bacteria have been well knownsince Metchnikoff published his results one hundred years

pos t e r s

44

Page 47: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

ago. However, a couple of decades elapsed until the industrydiscovered the potential in the production of probioticproducts, and nowadays the probiotic industry is blooming.Discovery of prebiotics boosted the use of probiotic microbesand the most innovative food products try to combine theadvantageous effects of these ingredients.In order to have the proper effect, probiotic bacteria must bepresent in the colon in an adequately high number.Enumeration of microbes is traditionally made by thetraditional microbiological method, on selective plates.Though this technique is very efficient, it might be timeconsuming, depending on the species.Our aim was to adapt existing species-species primers anda TaqMan probe for quantitation of four bacteria. Thesespecies were used as model organisms and cultivated in twoways: in the presence of and without various substances (e.g.inulin, a prebiotic ingredient). In our experiments we usedthree bacteria, which probiotic strains are present in pills andfood supplements (Bifidobacterium bifidum, Enterococcusfaecium, Lactobacillus acidophilus) and the Escherichia coli,as member of the normal human colon flora. We isolatedDNA from the bacteria by three different kits, in order tocompare the efficacy of the two basic DNA extractionmethods (precipitation vs. adsorption on column).The precipitation based kit was efficient in those cases wherefloating particles were present in the liquid cultivationmedium, because the sediment occluded the column atcentrifugation. The other two kits resulted clean DNA in ashorter time, but only from those suspensions, which did notcontain supplementing substances as floating particles.The PCR based identification and quantification of bacteriawas made by primers that were available in the literature. Wemanaged to identify and quantify the E. coli, but we need todo more experiments and optimization to get the same resultwith the other three bacteria, however, we got reassuringresults up to now.The expected result of our work is the elaboration of such aquick and reliable practical method that can at least partiallyreplace the work- and time-consuming microbiologicaltechnique and might be used for validation of it.This research was financed by Egerfood Ltd. and the NKTHresearch program.

P10THE USE OF LACTOBACILLUS GG IN CHILDREN WITHFUNCTIONAL ABDOMINAL PAIN: A DOUBLE-BLINDRANDOMIZED CONTROL TRIALSabbi Tamara*[1], Palumbo Massimo[1]

[1]Belcolle Hospital Viterbo - pediatric unit, Italy

OBJECTIVE: To determine whether oral administration of theprobiotic Lactobacillus GG under randomized, double-blinded, placebo-controlled conditions would improvesymptoms of recurrent abdominal pain in children.PATIENTS AND METHODS: 61 children with functionalabdominal pain were given Lactobacillus GG or placebo for6 weeks and entered follow-up for 4 weeks. Children entereda randomized, double-blind, placebo-controlled trial.RESULTS: LGG, but not placebo, caused a significantreduction of both frequency (P < .01) and severity (P < .01)of abdominal pain. These differences still were significant at

the end of follow-up (P < .02 and P < .001, respectively). CONCLUSIONS: Lactobacillus GG was superior to placebo inthe treatment of recurrent abdominal pain in children, LGGsignificantly reduces the frequency and severity of functionalabdominal pain and may help relieve such symptoms asperceived abdominal distention.

P11IN VITRO EFFECT OF FOUR NOVEL FLOURS FERMENTATIONON GUT MICROBIOTA PARAMETERSChitarrari Roberto*[2], Carnevali Paola[3], Costabile Adele[2]

- [2]Food Microbial Sciences, School of Food and Nutritional Sciences,University of Reading - Reading, United Kingdom - [3]Barilla G. e R.Fratelli - Parma, Italy

The prebiotic concept has attracted much interest as anapproach for the modulation of the colonic microbiota. Theaim of the present study was to evaluate the potentialprebiotic properties of four novel selected flours (wholegrainrye -WR, nutriwheat -NW, pulses -PS, 50% lentils + 50%chickpeas and barley milled grains -BMG) using in vitrohuman gut models, which represent different anatomicalareas of the large intestine. Prior to introduction into the gutmodel, the test ingredients were currently being digestedunder conditions that resemble the gastric (HCl, pepsin) andsmall intestinal (bile salts, pancreatic enzymes)environments, at appropriate pH and incubation times.Studies on the development of the microbiota in the gutmodels have been performed and bacteria identified bymolecular characterization techniques (FISH). Short chainfatty acids (SCFAs), as principal end products of gut bacterialmetabolism, have been measured along with a quantitativeassessment of predominant microbiota. Furthermore, weinvestigated the impact of the tested carbohydrates on thegrowth and survival of the human colon carcinoma cell lineHT29 using the growth curve assay. No statisticallysignificant cytotoxicity effects were observed for each testedflours. Significant results were observed in modulation of theoverall composition of the gut microbiota and on the overallSCFA production. In this context the results of our study onthe four selected flours provided new insight into potentialchanges undergone due to digestion and fermentation in thegastrointestinal tract. We demonstrated that WR and NWshowed higher prebiotic properties compare to PS and BMG.In details, both WR and NW caused an increase ofbifidobacteria, lactobacilli and Desulfovibrionales, whereas adifferent shift in microbial metabolism was observed by anincrease in the concentration of acetate and propionaterespectively. In conclusion, this study provides findingssupporting the utilization of WR and NW for the productionon new functional food as pasta and bakery products.However, in order to classify these flours as prebiotic, humanstudies are necessary to prove that these ingredientswithstands digestion in the upper gastrointestinal tract andhas a selective effect within faecal microbiota.

pos te r s

45

Page 48: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

P12SURVIVAL OF LACTOBACILLUS RHAMNOSUS LR06 DSM21981, LACTOBACILLUS PENTOSUS LPS01 DSM 21980,LACTOBACILLUS PLANTARUM LP01 LMG P-21021 ANDLACTOBACILLUS DELBRUECKII SUBSP. DELBRUECKIILDD01 DSM 22106 IN DIFFERENT PARTS OF THE DIGESTIVETRACT OF PATIENTS CHRONICALLY TREATED WITH PPIDel PianoM.*[1], Ballarè M.[1], Pagliarulo M.[1], Anderloni A.[1],Balzarini M.[1], Orsello M.[1], Carmagnola S.[1], Tari R.[1],Deidda F.[3], Allesina S.[3], Barba M.[3], Strozzi G.P.[2], Mogna G.[2], Mogna L.[2], Sforza F.[4]

[1]Gastroenterology Independent Operating Unit, Maggiore della CaritàHospital, Novara, Italy - [2]Probiotical SpA, Novara, Italy - [3]BiolabResearch Srl, Novara, Italy - [4]Casa di Cura I Cedri, Novara, Italy

P13STRUCTURAL CHARACTERIZATION OF CHONDROITINSULFATE FROM ITALIAN CHEESE PARMIGIANO REGGIANOCoppa Giovanni[1], Maccari Francesca[2], Zampini Lucia[1],Santoro Lucia[1], Galeazzi Tiziana[1], Gabrielli Orazio[1], Volpi Nicola*[2]

[1]Polytechnic University of the Marche, Ospedali Riuniti, Presidio Salesi -Ancona, Italy - [2]University of Modena and Reggio Emilia - Modena Italy

Chondroitin sulfate (CS) was purified for the first time fromItalian cheese Parmigiano-Reggiano and analyzed to evaluateits structure and properties. Two main polysaccharides wereidentified as CS, ~72%, and fast moving-heparin/heparansulfate, ~28%. Quantitative analyses yielded ~1.5-3.0 µg oftotal GAGs per gram of Parmigiano-Reggiano (0.15-0.30%).By means of specific chondroitinases and HPLC separationof generated unsaturated disaccharides, CS was found to becomposed of ~9% on nonsulfated disaccharide, ~26% ofdisaccharide monosulfated in 6 of the GalNAc, ~65% ofdisaccharide monosulfated in 4 of the GalNAc, with a chargedensity of ~0.91 and a 4/6-sulfated ratio of 2.45. The ratio of4/6 sulfated residues was confirmed by 13C-NMRexperiments. Chondroitinase B confirmed that the purifiedParmigiano-Reggiano CS contained mainly GlcA (~94%) asuronic acid. PAGE analysis showed a CS having a molecularmass with an average value of 15400. Along with otherpossible functions, such as glycomimetic playing a role assoluble receptors able to inhibit the binding of differentpathogens to the intestinal mucosa, CS may perform aprebiotic role compared to other well known complexoligo(poly)saccharides such as inulin, fructo- and isomalt-oligosaccharides. Studies are in progress to deeplyunderstand the physiological role of complexglycosaminoglycans and CS in human and bovine milks andderivatives.

P14FLAVONOIDES AND GASTROINTESTINAL TRACT: FROM THEBENCH TO CLINICAL PERSPECTIVESMarotta Francesco*[1], Tomella Claudio[1], Polimeni Ascanio[1],Joyal Steven[2]

[1]ReGenera Res Group - Milano, Italy - [2]Life Extension Foundation - Ft.Lauderdale, USA

There is increasing interest in the potential health benefits ofdietary flavonoids. Recent evidence suggests that significant

quantities of quercetin and possibly myricetin and kaempferolare absorbed in the gut. A larger fraction probably remainsin the lumen, and thus a substantial proportion of thegastrointestinal mucosa is exposed to biologically significantconcentrations of these compounds. Until recently the majorfocus of interest in flavonoids has been their putativeantioxidant properties in the plasma, but other biologicaleffects at the cellular level are now receiving greater attentionand may yet turn out to be even more important. This aspectof polyphenolic biochemistry is particularly relevant to thealimentary tract because the mucosal surfaces of the gut arethe most rapidly proliferating epithelial cells in the humanbody. In this context, it needs to be critically evaluatedwhether positive properties of flavonoid-rich diets can bereplaced by purified flavonoids as dietary supplements. Plantsources of flavonoids contain a complex mixture ofsecondary plant metabolites and not only flavonoids per se.Like the flavonols, catechins are biologically active andabundant in commercially relevant food products such asgreen tea. Unabsorbed catechins may exert biologicallyimportant effects on the metabolism of the colonicmicroflora. Cathechins have also been positively involved ina number of lipid metabolic pathways especially in obesesubjects. We conducted a study to ascertain whether an high-purity green tea extract [decaffeinated Camellia sinensis leafextract std. to 98% polyphenols by UV (710.5 mg), 45%EGCG by HPLC (326.25 mg), Mega Green Tea, LEF, USA]could affect body fat accumulation in normal-weight healthyrats fed a regular diet. Green tea extract-supplemented ratsshowed a significant lower body weight increase whileshowing an increased fluid intake. Green tea extractsupplementation brought about also a significant decreaseof either hepatic and mesenteric lipid content together withlower triglyceride and cholesterol liver, but not plasma, level.However, these rats showed a significant decreased plasmalevel of total bile acids. These preliminary data further enforcethe prospective clinical application towards a pro-activehealthy-promoting approach of medicine.

P15SUPPLEMENTATION WITH LACTOBACILLUS HELVETICUS ANDBIDIFOBACTERIUM LONGUM INDUCED IMMUNOLOGICALCHANGES IN MODERATE MALNOURISHED ELDERLYSUBJECTSFinamore Alberto*[1], Roselli Marianna[1], Brasili Elisa[1],Donini Lorenzo M [2], Neri Barbara [3], Carnevali Paola [4],Mengheri Elena[1]

[1]National Research Institute on Food and Nutrition (INRAN) - Roma, Italy- [2]Sapienza University - Rome, Italy - [3]Villa delle Querce RehabilitationInstitute - Nemi, Italy - [4]Barilla G&R f.lli SpA - Parma, Italy

The immune system undergoes age-associated alterations,termed immunosenescence, which is a process where somefunctions are reduced, others unchanged or increased. Agingis also characterized by changes in gut microbiota with adecrease of probiotic bacteria. The principal mechanism ofthe probiotic protective activity is the immunomodulation.Based on these considerations, probiotic supplementationhas been suggested to improve the immunosenescence andpromising results have been obtained up to now, althoughsome studies reported contrasting results on the

pos t e r s

46

Page 49: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

effectiveness of probiotics, depending on several factorsincluding probiotic strain, dose, time of treatment, andphysiological conditions of elderly.In this study we assessed whether of Lactobacillus helveticusBar13 and Bifidobacterium longum Bar33 could improve theimmune function of moderate malnourished elderly. Arandomized placebo controlled study was conducted onhospitalized elderly aged 75 ? 11.1 years with a moderatemalnutrition (BMI < 17 Kg/m2), that received daily a biscuitcontaining L. helveticus and B. longum (109 CFU each;provided by Barilla) or the same product without probioticsas placebo, for 30 days. Peripheral blood lymphocyte subsetswere analyzed by cytofluorimetric assay. After the probioticsupplementation, the main changes occurred in the femalepopulation, where naïve T cells (CD4+CD45RA+CCR7+)increased, whereas central memory T (CD4+CD45RA-CCR7+), effector memory T (CD4+CD28-CD95+), and naïveCD8+ CD28+ cells decreased. In the male population only adecrease in effector memory T cells was found. A stronginduction of Treg (CD4+CD25+CD127-) was observed in bothfemales and males. In conclusion, L. helveticus and B.longum were able to improve the immunosenescence byreducing age-induced changes in lymphocyte subpopulationsand increasing Treg cell population which may contribute toimprove the defense against the age associated inflammatorydiseases. This work was supported by an Italian grant fromMiPAAF, project Qualifu (Alieta)

P16CAN LACTOBACILLUS RHAMNOSUS LR06 DSM 21981,LACTOBACILLUS PENTOSUS LPS01 DSM 21980,LACTOBACILLUS PLANTARUM LP01 LMG P-21021 ANDLACTOBACILLUS DELBRUECKII SUBSP. DELBRUECKIILDD01 DSM 22106 RESTORE THE “GASTRIC BARRIEREFFECT” IN PATIENTS CHRONICALLY TREATED WITH PPI?Del PianoM.*[1], Ballarè M.[1], Pagliarulo M.[1], Anderloni A.[1],Balzarini M.[1], Orsello M.[1], Carmagnola S.[1], Tari R.[1],Deidda F.[3], Allesina S.[3], Barba M.[3], Strozzi G.P.[2],Mogna G.[2], Mogna L.[2], Sforza F.[4]

[1]Gastroenterology Independent Operating Unit, Maggiore della CaritàHospital, Novara, Italy - [2]Probiotical SpA, Novara, Italy - [3]BiolabResearch Srl, Novara, Italy - [4]Casa di Cura I Cedri, Novara, Italy

P17CHARACTERISATION OF THE CYANOBACTERIAL TOXINREMOVAL PROCESS IN THE PRESENCE OF PROBIOTICBACTERIA Nybom Sonja*[1], Dziga Dariusz[2], Salminen Seppo[3],Meriluoto Jussi[1]

[1]Åbo Akademi University/Department of Biosciences - Turku, Finland -[2]Jagiellonian University - Krakow, Poland - [3]University of Turku -Turku, Finland

Toxic cyanobacteria have been reported in lakes andreservoirs in several countries. The presence of toxins indrinking water creates a potential risk of toxin transferencefor water consumers. Besides chemical and physicalmethods of cyanotoxin removal from water and raw materialsfor food industry, biodegradation methods should beproposed.The aim of the current study was to assess the ability of

probiotic bacteria to remove cyanotoxins, such asmicrocystins, from aqueous solutions. Furthermore, theremoval process was investigated by testing the hypothesisof enzymatic degradation of microcystin-LR in the presenceof probiotic lactic acid and bifidobacterial strains and theparticipation of a proteolytic system in the toxin removal.Specific strains of probiotic bacteria, including Lactobacillusrhamnosus, Bifidobacterium lactis and Bifidobacteriumlongum, were studied. A maximum removal of 70-90% of microcystins wasobserved (100 µg toxin/L, 1E+10 CFU/mL, 37°C, 24 h). Theremoval of microcystins was shown to be dependent ontemperature, bacterial cell density and cell viability. Theresults suggest that extracellularly located cell-envelopedproteinases are involved in the decomposition ofmicrocystins. In particular, a correlation between proteolyticactivity and microcystin removal was found. In addition,EDTA, which was indicated as a main inhibitor of proteinasesof the investigated strains, was shown to limit the rate ofmicrocystin removal.Toxin removal by probiotic bacteria may open newopportunities for detoxification and decontamination ofmicrocystin-contaminated water and also to provide humanhealth protection by eliminating toxins from thegastrointestinal tract.

P18PROBIOTIC LACTOBACILLUS RHAMNOSUS LGA UPREGULATESSS-DEFENSIN 9 EXPRESSION IN CULTURED CHICKENSMALL INTESTINAL EPITHELIAL CELLSLi Guanhong*[1], Liu Siguo[2], Hong Zhimin[1], Jia Yongjie[1],You Jinming[1], Qu Minreng[1]

[1]College of Animal Science and Technology, Jiangxi AgriculturalUniversity - Nanchang, China - [2]Harbin Veterinary Research Instituteof Chinese Academy of Agricultural Sciences - Harbin, China

The avian beta-defensin 9 (AvBD9), an inducible antimicrobialpeptide expressed in the epithelial cells of chicken smallintestine, plays an important role in maintaining thehomeostasis of gastrointestinal microflora and intestinalimmune system. The probiotic Lactobacilli, predominantlyclonized in the chicken small intestine, interact with epithelialcells and can modulate the physiological function of theepithelial cells. We hypothesize that the probiotic Lactobacillicould stimulate the antimicrobial peptides expression duringthe Lactobacilli-epithelial cell interaction. However, there havebeen no studies regarding the effects of Lactobacilli-epithelialcell interactions on the antimicrobial peptide expression inchicken.Probiotic Lactobacillus rhamnosus LGA (L. rhamnosus LGA)was selected to investigate the effects of lactobacillus stimulion the AvBD9 mRNA expression in cultured chicken smallintestinal epithelial cells. The time-and dose-response ofAvBD9 gene expression upon stimulation of epithelial cellswith L. rhamnosus LGA was also examined. The AvBD9mRNA expression was determinted by fluorescencequantitative PCR. AvBD9 expression was upregulated bystimulation of L. rhamnosus LGA at different concentrations(2×105 cfu/ml, 2×106 cfu/ml, 2×107 cfu/ml), and expressiondifference was observed between treatments at threebacterial concentrations. Heat-inactivated L. rhamnosus LGA

pos te r s

47

Page 50: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

also stimulated the expression of AvBD9, and showedstronger induction response than live bacteria (P<0.05). TheL. rhamnosus LGA promoted AvBD9 mRNA transcription intime-dependent manner. The AvBD9 mRNA expressionpeaked at 12 h of incubation upon treatment of epithelial cellswith lactobacillus. AvBD9 peptide was detected by Westernblot in the supernatants of cultured epithelial cells treatedwith L. rhamnosus LGA, indicating that AvBD9 peptidereleases into extracellular medium and exerts bactericidalaction. Probiotic L. rhamnosus LGA stimulates AvBD9expression in the epithelial cells of chicken small intestineduring lactobacillus-epithelial cell interaction. This studyrevealed the new possible functional mechanism by whichthe probiotic Lactobacilli exert their beneficial effects to thehost through the antimicrobial peptide expression in thesmall intestinal epithlium, which will contribute to furtherstudies and understanding on the molecular mechanismunderlying the modulation of intestinal function by probiotics.

P19SELENIUM AND ZINC INTERNALIZED BY LACTOBACILLUSBUCHNERI LB26 DSM 16341 AND BIFIDOBACTERIUMLACTIS BB1 DSM 17850: DEVELOPMENT OF A NEWBIOLOGICAL METHOD TO EVALUATE THE BIOAVAILABILITYOF THE TWO MINERALSPane M.*[2], D'Andrea M.[3], Nicola S.[3], Strozzi G.P.[2], Mogna G.[2], Del Piano M.[1]

[1]Gastroenterology Independent Operating Unit, Maggiore della CaritàHospital, Novara, Italy - [2]Probiotical SpA, Novara, Italy - [3]BiolabResearch Srl, Novara, Italy

P20SAFETY OF A PROBIOTIC CHEESE COMPRISING L.PLANTARUM TENSIA ACCORDING VARIETY OF HEALTHINDICES IN DIFFERENT AGE GROUPSSongisepp Epp*[1], Hütt Pirje [2], Rätsep Merle [1], Shkut Elena[1], Zilmer Mihkel[2], Kõljalg Siiri[2], Truusalu Kai[2], SmidtImbi[2], Kolk Helgi[2], Zagura Maksim [3], Mikelsaar Marika [2]

[1]Bio-Competence Centre of Healthy Dairy Products LLC - Tartu, Estonia- [2]University of Tartu - Tartu, Estonia - [3]Tartu University Clinics - Tartu,Estonia

Safety of L. plantarum TENSIA (DSM 21380) was tested invitro, in composition of semi-hard cheese, in an animalmodel and in human intervention studies with different agegroups.The susceptibility of L. plantarum TENISA to 8 antibiotics,presence of tet (M, S, O, K, L) genes and class 1 integronwas assessed by E-test and PCR methods. Production ofbiogenic amines (BA) in decarboxylation medium with 1% ofL-histidine, L-glutamine, L-ornithine, L-arginine or L-lysineand in cheese was tested by gas chromatograph. The bio-safety of L. plantarum TENSIA was evaluated on NIH linemice fed with probiotic cheese comprising TENSIA inconcentration of 4x 109 CFU/g for 30 consecutive days.In human trials in adults and elderly the impact of differentdoses of Edam-type cheese and probiotic strain on bodyweight, gut functionality indices and host metabolism wasevaluated. The strain TENSIA was susceptible to all testedantibiotics, did not possess tetracycline resistance genestet(L), tet(S) and tet(O) nor contained the IntI gene. However,

the strain possessed tet(K) and tet(M) genes. L. plantarum TENSIA did not produce potentially harmful BA. In animal model no translocation of administrated strain orother microbes into blood or organs of mice was detected.No harmful impact on body mass index, inflammatorymarkers or serum lipidogram was registered during humantrials in daily dose of 2x1010 CFU or 1.5x 108 CFU perserving for 3-weeks.No negative impact on gastrointestinal welfare was registeredbut the consumption of 100g/d for 3 weeks caused hardstools from the second week of the trail. The content of totallactobacilli increased in feces and the presence of ingestedprobiotic strain was confirmed after consumption of cheese. Thus, L. plantarum strain TENSIA strain did not harbor anyundesirable characteristic. The regular semi-hard Edam-typecheese (fat content 26%) with probiotic additive in daily doseof 50 g or in excess (100 g) and with probiotic daily dose of1010 CFU during 3-weeks is was proved to be safe.

P21EFFECT OF LAB ON CYTOKINE SECRETION BY THP-1 CELLSSTIMULATED BY LPSHacin Biljana*[2], Citar Manuela[2], Tompa Gorazd[1], Rogelj Irena[1]

[1]University of Ljubljana - Ljubljana, Slovenia - [2]Medis, d.o.o.,Ljubljana, Slovenia

Inflammatory bowel syndrome is regarded as a chronicillness that can dramatically affect the quality of a sufferer'slife. Lactic acid bacteria have shown to alleviate the syptomsof IBD presumably due to their anti-inflammatory properties.To investigate the role of cytokines in interactions betweenlactic acid bacteria and the immune system, production ofTNF-a and IL-6 by THP-1 cells after stimulation with LPS,lactic acid bacteria or both was measured. Lactobacillus gasseri K7, a strain of human origin, waspreviously found to exhibit several probiotic properties suchas survival in simulated conditions of the gastrointestinaltract, antimicrobial activity, adhesion to porcine intestinalmucosa ex vivo, as well as adhesion to human and porcinemucus and Caco-2 cell line in vitro. Additional investigationof its anti-inflammatory properties will further establish theprobiotic properties of the strain. Due to their well knownprobiotic effects including modulation of immune response,Lactobacillus casei DN 114 001 and Bifidobacterium animalissubsp. lactis BB12 were used as positive controls.THP-1 cells were grown in RPMI medium supplemented withFBS and antibiotics. Prior to the experiment the cells werestimulated with PMA (phorbol ester) for 48 hours to inducedifferentiation into macrophages. Cells were then incubated either with selected lactic acidbacteria (DN 114 001, K7 or BB12), LPS (lipopolysaccharidesof E. coli O111:B4) or combination of LPS and LAB. Samplesof the cell line supernatant were collected after 2, 4, 6, 8, 12and 24 hours of incubation to determine cytokine production(ELISA).High concentrations of TNF-a in the THP-1 cell supernatantcould already be measured after 2h of incubation and highlevels of IL-6 were measured after 4h of incubation with LPS.When LPS and LAB were applied simultaneously, LABreduced the concentration of inflammatory cytokinesmeasured.

pos t e r s

48

Page 51: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

THP- 1 cell line macrophages stimulated with LPS proved tobe a good model for assessing cytokine release and could beused as a tool for screening anti-inflammatory properties ofvarious LAB strains.

P22SLOW RELEASE EFFERVESCENT TABLETS WITH L.FERMENTUM LF10 DSM 19187 AND L. ACIDOPHILUS LA02 G. Mogna[1], F. Deidda[2], S. Allesina[2], M. Pane[2], M. Barba[2],M. D’Andrea[2], P. Lorenzini[2], S. Nicola[2], E. Raiteri[2], G.P.Strozzi[1], L. Mogna[2], F. Vicariotto[3]

[1]Probiotical SpA - Novara, Italy - [2]Biolab Research Srl - Novara, Italy[3] -Department of Obstetrics and Gynecology, San Pio X Hospital - Milan, Italy

Candida is an opportunistic fungus that normally inhabits themouth, throat, gastrointestinal tract and vagina, but Candidaovergrowth can cause infections such as candidiasis andthrush. Both physiological and pathological affections can leadto mucosal microbiota imbalance, conditioning the growth ofopportunistic “putrescent flora”. The composition of vaginalmicroflora is known to be critical for woman‘s health andwellbeing. In a healthy vagina, the dynamic micro-environmentis mainly composed of “Döderlein’s bacilli”, which generallycomprise different species of lactobacilli. Imbalance of thevaginal microbiota leads to a prevalence of opportunisticmicrorganisms that may cause vaginitis or forms of vaginosis.It is known from literature that some lactic acid bacteria havethe capacity to inhibit the overgrowth of Candida spp. The aim of this in vitro study was to evaluate the ability of aninnovative dosage form of effervescent tablets releasing CO2,containing two select probiotics, L. fermentum LF10 and L.acidophilus LA02, to inhibit different species of Candida andputrescent opportunistic flora. In particular, L. fermentum LF10DSM 19187 has shown a strong in vitro antimycotic activityagainst different Candida biotypes. L. acidophilus LA02 DSM21717 may help to rebalance the microbial vaginal flora.Lactobacilli with antimycotic activity in the form of innovativeeffervescent tablets which release CO2 may represent a neweffective tool for long term treatment of altered microflora inthe vaginal lumen.

P23SYMPTOM RESOLUTION AND IMMUNE MATURATION ININFANTS WITH ATOPIC DERMATITIS RECEIVINGHYDROLYZED FORMULA WITH LACTOBACILLUS GG (LGG)Nermes Merja[1], Salminen Seppo[1], Isolauri Erika*[1]

[1]University of Turku - Turku - Finland

Background: Gradual intestinal colonization promotesmaturational events in the gut barrier. Abnormal barrierfunction in atopic dermatitis (AD) could lead to a vicious circleof sensitization to antigens and further barrier compromise.We previously showed that LGG enhances gut barrier andhastens recovery of symptoms in infants with AD andchallenge-proven food allergy (FA). This study assesses theeffect of LGG on antigen absorption and symptom resolutionin intrinsic AD not related to FA. Methods: In a double-blindrandomized study, 39 infants with AD received extensivelyhydrolyzed casein formula (Nutramigen®) with (H-LGG,n=19) or without (Control, n=20) LGG 5.0 x 107 cfu/g for 3months.

Severity of AD was determined by SCORAD and blood wasdrawn at study entry, 1, and 3 months. Immunoglobulin-secreting cells by ELISPOT and the proportions of B cells(CD19+) and memory B cells (CD27+) among peripheralblood leukocytes by flow cytometry were determined. Datawere analyzed by ANCOVA adjusted for baseline.Results: Baseline characteristics were similar betweengroups, including age at onset of AD and at study entry,mode of delivery, and duration of breastfeeding. There wereno significant differences in SCORAD between the twogroups during the study. However, the greatest meandecrease in SCORAD occurred at 1 month in the H-LGG [from27.9 (95%CI 22.3–33.5) to 17.6 (95%CI 10.6–24.6)] and at3 months in the Control group [from 30.2 (95%CI 23.6–36.8)to 17.8 (95%CI 10.3–25.3)]. The proportions of IgA- and IgM-secreting cells decreasedsignificantly in the H-LGG group (Figure); the baseline-adjusted ratios for H-LGG vs. Control at 1 month were 0.59(95%CI 0.36–0.99, P = 0.044) for IgA- and 0.53 (95%CI0.29–0.96, P = 0.036) for IgM-secreting cells. Theproportions of CD19+CD27+ B cells increased in H-LGG butnot in the Control group.Figure. Ig-secreting cells in peripheral blood may reflectlower antigen uptake in H-LGG group Conclusion: Ahydrolyzed formula with LGG may strengthen gut barrier,thus reducing antigen uptake and immunoglobulin secretion,potentially causing faster immune maturation and symptomresolution in infants with intrinsic AD.

P24THE GLOBAL PHENOTYPIC ANALYSIS OF PUTATIVEANTIALLERGIC POTENTIAL OF THREE LACTOBACILLUSSTRAINSAleksandrzak-Piekarczyk Tamara*[1], Koryszewska-BaginskaAnna[1], Bardowski Jacek[1]

[1]Institute of Biochemistry and Biophysics Polish Academy of Sciences- Warsaw, Poland

The increase in incidence of allergy in the last few decadeshas been linked to the high hygienic standards connected toa reduced microbial stimulation of the mucosal immunesystem. Hence, this demands the development of newprevention strategies to inhibit the rapidly increasingprevalence of allergic diseases.

pos te r s

49

Page 52: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

There is much evidence concerning the role of probiotics inthe prevention and therapy of allergic diseases. Variousstrains of lactic acid bacteria (LAB), especially lactobacilli andbifidobacteria, are considered as probiotics and have beenreported to suppress the allergic reactions. Recently, twostrains of Lactobacillus casei LOCK 0900 and LOCK 0908 andone strain of Lactobacillus paracasei LOCK 0919 wereselected according to their antagonistic activity againstpathogenic bacteria, tolerance to low pH and bile acids(demonstrated by Cukrowska et al. 2009). Preliminarystudies have shown that ingestion of that the mixture of L.casei LOCK 0900, L. casei LOCK 0908 and L. paracasei LOCK0919 strains affects the immune system by inducing TH1 andregulatory cytokine production and by suppressing pro-allergic response (Cukrowska et al. 2010). In this work, we present an additional phenotypiccharacterization, as it is considered to be essential in betterdefining the role and use of specific probiotics Lactobacillusstrains. Using Biolog Phenotype Microarrays we investigated,which of metabolic substrates (various carbon and nitrogensources) could or could not be used by these Lactobacillusstrains. The same technique was used to examine theresponse of tested strains to various osmolytes and pH.Furthermore, API ZYM (BioMerieux) test was used tocomplete phenotypic identification as each strain wasexamined for 19 enzymatic reactions.Cukrowska B., Motyl I., Kozáková H., Schwarzer M., GóreckiR.K., Klewicka E., Slizewska K., Libudzisz Z.: ProbioticLactobacillus strains: in vitro and in vivo studies. FoliaMicrobiol. 54, 533–537 (2009).Cukrowska B.,Rosiak E., Klewicka E., Motyl I., Schwarzer M.,Libudzisz Z., Kozáková H.:Impact of heat-inactivated Lactobacillus casei andLactobacillus paracasei strains on cytokine responses inwhole blood cell cultures of children with atopic dermatitis.Folia Microbiol. 55, 277–280 (2010).This work is partly supported by the grant from the NationalCentre for Research and Development No. N R12 0101 10.

P25DOES CRANBERRY USEFUL FOR PREVENTION RECURRENTURINARY TRACT INFECTIONS IN CHILDREN?Dessì Angelica*[1], Fanos Vassilios[1]

[1]Department of Paediatrics, Neonatal Intensive Care Unit, NeonatalPathology, Puericultura Institute and Neonatal Section, University ofCagliari - Cagliari, Italy

Urinary tract infections (UTI) are common in childhood. In30-50% of children with UTI the infections occur recurrently,especially in those with vesicoureteral reflux (VUR),neurogenic bladder (NB), previous cystitis or pyelonephritisand malformative uropathies. To reduce the likelihood of UTI,antibiotic prophylaxis has been regarded as the therapeuticstandard for many years. However, the disadvantage of long-term antibiotic therapy is the potential for development ofcollateral effects and resistant organisms in the host. Suchreasons have induced scientists to search for alternativemodalities of UTI prevention and have contributed todetermining the increasing desire for “naturalness” of thepopulation and preventing excessive medication. Cranberry,alone or in combination with probiotics has been suggested

by literature. The use of cranberry fulfils these needs bypotentially replacing or enhancing traditional procedures. Thepurpose of this study was to assess the effectiveness ofcranberry in preventing UTI in pediatric populations. Wesearched PUBMED, the Cochrane Central Register ofControlled Trials and Internet. Cranberry in patients withprevious UTI was evaluated in three studies, cranberry inpatients with VUR in three studies and four studies analyzedthe efficacy of cranberry in children with NB. In seven of ninestudies cranberry had a significant effect in preventing UTI.References Fanos V, Atzei A, Zaffanello M, Piras A, Cataldi L2006. Cranberry and prevention of urinary tract infections inchildren. J Chemother 3: 21-24. Dessì A, Atzei A, Fanos V.2010. Cranberry in pediatria: efficacia clinica e revisione dellaletteratura. SILAE - Abstract Book of XIX Congress “FernandoCabieses Molina” - ISBN: 88-8160-218-0; 56-57. Keywords:Cranberry, UTI, VUR, pediatrics, children, neurogenic bladder.

P26LACTOBACILLUS SPP. STRAINS OF CULTURE COLLECTIONOF TARTU UNIVERSITY, ESTONIAŠtšepetovaJelena*[1], Rööp Tiiu[1], Mändar Reet[1], Sepp Epp[1],Mikelsaar Marika[1]

[1]University of Tartu - Tartu, Estonia

Culture collection of Lactobacillus spp. isolates (more than1000, LB) of University of Tartu, Estonia has been establishedin 1994. LB were collected from fecal samples of healthy anddiseased infants, children, adults, elderly persons during 8different projects (1994-2009). First level of testing of LBincludes in house preliminary identification by specific colonyand cell morphology, gram-positive, catalase negative,fermentation groups. Next, for more precise identification ofsome LB isolates the molecular methods have been used.However, to know which of the advanced methods is the bestfor species identification, the selection should be based oncomparative studies. Aim: To compare the suitability ofdifferent methods for identification of LB strains from Tartucollection. Material and methods: A total 120 LB isolates and21 reference strains of 20 species were analysed bybiochemical (API 50CHL) and molecular methods internal-transcribed spacer polymerase chain reaction (ITS-PCR) and sequencing of 16S rRNA. ITS-PCR was performedaccording to Jacobsen et al. (1999) with primers(GATTCTGGCTCAGGATGAACG and AGGTCCTAACAAGGTA).The PCR products were restricted with TaqI restrictionenzyme. Two primers, CO1: AGTTTGATCCTGGCTCAG andCO2: TACCTTGTTACGACT, were used to generate a 1.5-kb16S rRNA product for sequencing (Simpson et al. 2003). The16S rRNA sequences were compared with GeneBankdatabase by BLAST algorithm.Results: The concordancebetween API 50CHL and ITS-PCR was found in 10 out of 21(48%) reference strains. The concordance between API50CHL and ITS-PCR for LB isolates belonging to 14 specieswas 85/112 (75.8%). Some strains remained unidentified byboth methods. In the case of sequencing the concordancewas high. Both molecular methods (ITS-PCR andsequencing) assigned the isolates of L. acidophilus (API50CHL) to either L. acidophilus or L. gasseri. Differentfunctional properties (metabolites, antimicrobial andantioxidative activity) of the possible probiotic strains have

pos t e r s

50

Page 53: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

been evaluated. Conclusion: The preliminary identificationmethods of Lactobacillus spp. serve as basis for obtaininglarge collections. The molecular identification methods arenecessary for selection of the strains with health effects.

P27ASSESSMENT OF SAFETY AND TOLERANCE OF AFERMENTED DAIRY FOOD CONTAINING NOVEL,POTENTIALLY PROBIOTIC STRAINSChambaud Isabelle*[1], Jeansen Stéphanie[1], Elfakir Anissa[1],Banning Federike[2], Queudot Jean-Christophe[3], BouchezElodie[3], Bourlioux Pierre[4], Marteau Philippe[5], SchrezenmeirJuerguen[6][1]Danone Research - Palaiseau, France - [2]Harrison Clinical ResearchDeutschland GmbH - Munich, Germany - [3]CIT Safety and HealthResearch Laboratories - Evreux, France - [4]Faculty of Parmacy - Paris-Sud University - Paris, France - [5]University Denis Diderot, Paris 7 &AP-HP, Lariboisière hospital - Paris, France - [6]Gutenberg-UniversityMainz - Kiel, Germany

Lactobacilli have a long history of use as probiotics, withoutestablished risks in humans (Bernardeau et al., 2006). Therecurrently is no regulation or consensus for the safetyassessment of live micro-organisms added in food products ascultures or ingredients. However, there are a number ofguidelines, recommendations and expert reviews on possiblesteps to document and validate the safety of live micro-organisms used in foods (Borriello et al., 2003; O'Brien et al.,1999; de Vrese & Schrezenmeir, 2008; FAO and WHO, 2002 and2006). The safety and tolerance of a dairy drink containingpotentially probiotic strain were investigated in a monocentricrandomised double-blind clinical trial following FAO/WHOguidelines. 96 healthy adults, aged from 18 to 55 years wererandomly allocated to 4 groups of 24 subjects each. Subjectsconsumed either 1 bottle (100 g) or 3 bottles (3 x 100 g) of studyproducts (either a fermented dairy product or a control product)per day. The total duration of the study was 10 weeks, includinga 2-week wash-out period, a 4-week product consumptionperiod and 4-week follow-up period. The recorded outcomeswere: adverse events, bowel movements, faeces consistency,and frequency of Digestive Symptoms (Questionnaire),physicalexamination, blood cell counts, creatinin, CRP, glycaemia, bloodlipids, calprotectin in faeces,). Based on descriptive statisticsincluding Cohen’s d approach for quantitative and probabilitiesof an event in the verum/control group for qualitative binaryparameters, confidence intervals, and case by case evaluationnone of these parameters appeared to be influenced by theproduct consumption, compared to the test product. Thenumber of adverse events during the whole study period were:89 in the verum and 88 in the control group at 1 bottle per dayand 71 in the verum group versus 83 in the control group at 3bottles per day. In conclusion the results of this study do notindicate at this step safety concerns for the product ingestion inthe dosages of 1 to 3 times per day. In view of the limited samplesize and power of the study monitoring of safety parameters andadverse events is still recommended in future intervention trialsdedicated to efficacy and other goals.In addition, Repeated Dose90-day Oral Toxicity Study in Rodents, following OECD408guidelines were conducted on the probiotic strains. No signs oftoxicity were observed at the clinical, laboratory and pathologyinvestigations.

P28PROBIOTIC TREATMENT INDUCED AGE DEPENDENTMETABOLIC CHANGESBrasili Elisa[1], Tomassini Alberta [2], Finamore Alberto*[1],Roselli Marianna [1], Mengheri Elena [1], Capuani Giorgio [2],Sciubba Fabio [2], Miccheli Alfredo [2]

[1]National Research Institute on Food and Nutrition, INRAN - Roma,Italy - [2]Sapienza University - Roma, Italy

The intestinal microflora plays an important role in hostmetabolism. The aging process is characterized by changesin gut microbiota community and metabolic system. Lacticacid bacteria can ameliorate several biological functions. Upto now only few studies were conducted on the regulation ofmetabolism by probiotics and no data are available duringaging processes. This work was conducted to assess theeffects of a probiotic mixture on the metabolic phenotype ofadult and aged mice, and to find possible biomarkers of theprobiotic induced metabolic changes in different ages. Aged(16 months old) and adult (3 months old) BALB/c micereceived Lactobacillus acidophilus (La5) and Bifidobacteriumlactis (BB12) (1.5×109 CFU) or PBS, daily for 4 weeks. Urineand feces were collected for 48h at the beginning and at theend of treatment, and analyzed by high-resolution 1H-NMRspectroscopy combined with partial least squares-discriminant analysis. In the old PBS treated mice, fecesNMR-based metabolomic analysis identified a gut flora ageassociated metabolic phenotype (metabotype) that differedfrom that of the adult mice. Discriminant metabolites werebile salts, isoleucine, a-keto-ß- metil N-valerate, histidine andtyrosine, a-keto-isovalerate, 4-OH phenyalcetate andsuccinate. Urine NMR-based metabolomic analysis in the oldPBS treated mice identified an aged metabotype that differedfrom that of adult mice, and the altered metabolites weresuccinate, citrate, 3-OH-isovalerate and taurine. Probioticadministration induced several metabolic changes in fecesand urine of aged and adult mice. Among the changes, thecommon alterations occurring in aged and adult mice werean increase in 4-OH phenylacetate in fecal water and anincrease in dimethylglycine in urine. Urine metabolomicanalysis identified a metabotype in probiotic treated agedmice that suggested a systemic effect on trans-methylationand trans-sulfuration processes. In conclusions, the usedapproach has proved useful to study the interactions betweenhost and probiotics and to identify biomarkers of theprobiotic induced age dependent metabolic changes.

pos te r s

51

Page 54: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

P29COMPARATIVE STUDY ON THE CELL SURFACE PROPERTIESAND STRUCTURE OF EXOPOLYSACCHARIDES PRODUCEDBY LACTOBACILLUS CASEI AND LACTOBACILLUSPARACASEI STRAINSGórska-Fraczek Sabina*[1], Gamian Andrzej[1], Kozakova Hana[2],Schwarzer Martin[2]

[1]Institute of Immunology and Experimental Therapy, Polish Academyof Sciences, - Wroclaw, Poland - [2]Department of Immunology andGnotobiology, Institute of Microbiology of the Academy of Sciences of theCzech Republic, v. v. i - Novy Hradek, Czech Republic

Probiotics are defined as live microorganisms which, whenadministrated in adequate amounts, confer a health benefiton the host. There is much evidence concerning the role ofprobiotics in the prevention or therapy of allergic diseases[1]. Novel probiotics used in allergy treatment must meetseveral criteria: be safe for the host organism, be resistantto gastric acids and bile salts in order to survive into the gut,and be able to modulate Th1/Th2 balance in favor of anti-allergic Th1 immune responses [2]. Three strains presentingantagonistic activity against pathogenic bacteria, tolerance tolow/high pH, which in functional animal studies inducedcytokine production towards Th1 anti-allergic response wereselected. The strain specificity may be related to the structureof cell-wall components such as exopolysaccharides (EPSs),the major components of lactobacilli biofilm, however theirrole in the “cross-talk” between bacteria and the immunesystem is poorly understood. In the present work we reportedthe surface properties and structure of the EPS produced byLactobacillus casei LOCK 0900, L. casei LOCK 0908 and L.paracasei LOCK 0919 strains. Methods: The biopolymerswere isolated from human intestinal flora. The polysaccharidematerials were prepared by TCA extraction of bacterial cellmass, purified by anion-exchange and gel permeationchromatography and characterized using chemical andenzymatic methods. Surface properties were characterizedby salt aggregation test, adherence to xylene, adhesion toCaco-2 cell line and slime production.Results: On the basis of sugar and methylation analysis theEPS were shown to be composed of glucose, galactose,mannose, galactosamine, glucosamine, mannosamine andfucose in different configuration and ratio. Strains appearedto be a different entities and their surface properties weredistinct. The project is supported by grant co-funded by theEuropean Regional Development Fund under OperationalProgramme Cross-border Cooperation “Czech Republic -Republic of Poland 2007 -2013'”.Programme under theEuropean Territorial Cooperation Objective. „Probiotics:scientific cooperation, transfer of knowledge and education”No CZ.3.22/2.1.00/09.015741. BORCHERS A.T., SELMI C., MEYERS F.J., KEEN C.L.,GERSCHWIN M.E.: Probiotics and immunity. J.Gastroenterol.44, 26–46 (2009). 2. ISOLAURI E.: Probiotics in humandisease. Am.J.Clin.Nutr. 73, 1142–1146 (2001)

P30PREBIOTIC AND ANTIMICROBIAL EFFECT OF KEFIRANVardjan Tinkara*[1], Canžek Majhenic Andreja[2], Rogelj Irena[2]

[1]Kele & Kele, d.o.o. - Logatec, Slovenia - [2]University of Ljubljana,Biotechnical Faculty - Domžale, Slovenia

Kefiran is water soluble exoploysaccharide embedding lacticacid bacteria and yeasts inside kefir grains. This symbioticcommunity of microbiota and kefiran has been described toposses many health-promoting properties. Especially kefiranhas attracted considerable interest because of itsantimicrobial, anti-inflammatory, antifungal and antitumouralactivity. It has been also shown, that kefiran lowers bloodpressure and reduces serum cholesterol levels in rats. In ourstudy, the prebiotic and antimicrobial effects of kefiran,extracted either from pure lactobacilli cultures or kefir grains,were investigated. Preliminary, lactobacilli were isolated fromkefir grains and identified by partial sequencing of the 16SrDNA. To optimize their ability for kefiran or kefiran-likepolysaccharide production lactobacilli were cultivated indifferent media at different pH. Exopolysaccharides wereextracted from both, pure lactobacilli suspensionsand kefir grain. To determine their sugar composition,extracted exopolysaccharides were subjected to capillaryelectrophoresis. Antimicrobial activity of kefiran was evaluatedusing disk diffusion method. To establish the prebiotic effectof kefiran, the growth of different lactobacilli andbifidobacteria, common to gastrointestinal tract, was testedon plain agar medium or agar medium, supplemented withkefiran. Lactobacilli, isolated from kefir grains were identifiedas members of Lactobacillus kefiranofaciens subsp.kefirgranum, Lactobacillus parakefiri and Lactobacillus kefirispecies. Since these species are not recognized as typicalkefiran produces, their exopolysaccaharides were alsoidentified as kefiran-like polysaccharides. Furthermore, onlyexopolysaccharide, extracted from kefir grains was confirmedto be kefiran due to its structure composed of equal amountsof D-glucose and D-galactose. Moreover, this kefiran exertedantimicrobial activity against some potentially pathogenicstrains and prebiotic effect stimulating the growth of certainlactobacilli and bifidobacteria. Findings obtained in this studysuggest that kefiran is present in kefir grains, but none of theisolated strains was able to produce kefiran in pure cultureindicating that kefiran production by strains in pure culture islimited. To conclude the antimicrobial activity and prebioticeffect were demonstrated for kefiran extracted from kefir grains.

pos t e r s

52

Page 55: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

53

faculty

Page 56: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

f a cu lt y

AAggett Peter (United Kingdom) p. 13Agostoni Carlo (Italy) p. 13Andriulli Angelo (Italy) p. 6, 14Anti Marcello (Italy) p. 7Arlorio Marco(Italy) p. 10Attili Adolfo Francesco (Italy) p. 10Aureli Paolo (Italy) p. 19

BBalsano Clara (Italy) p. 4Barbara Giovanni (Italy) p. 4, 6Bastos Deborah (Brazil) p. 10Belzer Clara (The Netherlands) p. 6Bezirtzoglou Eugenia (Greece) p. 15Bienenstock John (Canada) p. 6Brandi Maria Luisa (Italy) p. 14Brigidi Patrizia (Italy) p. 4, 14Bruzzese Eugenia (Italy) p. 12Buonocore Giuseppe (Italy) p. 12

CCalvani Menotti (Italy) p. 11Camera Emanuela (Italy) p. 19Cani Patrice D. (Belgium) p. 15Caporaso Nicola (Italy) p. 8Caprilli Renzo (Italy) p. 7Capurso Gabriele (Italy) p. 4, 8Capurso Lucio (Italy) p. 5Carding Simon (United Kingdom) p. 7Caselli Michele (Italy) p. 4Castagliuolo Ignazio (Italy) p. 7Castellazzi Annamaria (Italy) p. 6Catassi Carlo (Italy) p. 14Cicero Arrigo F.G. (Italy) p. 19Collins Stephen M. (Canada) p. 5Corazza G. Roberto (Italy) p. 14Corazziari Enrico (Italy) p. 7Crespi Massimo (Italy) p. 14Cricelli Claudio (Italy) p. 6Crittenden Ross (Finland) p. 6Cucchiara Salvatore (Italy) p. 6, 7

DDanese Silvio (Italy) p. 7D'Argenio Giuseppe (Italy) p. 10De Giorgio Roberto (Italy) p. 4Del Piano Mario (Italy) p. 14

Del Rio Daniele (Italy) p. 14Delle Fave Gianfranco (Italy) p. 5Devirgiliis Chiara (Italy) p. 7de Ridder Lissy (The Netherlands) p. 12de Vos Willem M. (The Netherlands) p. 6Di Giorgi Gerevini Valeria (Italy) p. 13Dorè Joel (France) p. 5Drago Lorenzo (Italy) p. 19Dupont Christophe (France) p. 12

EElli Marina (Italy) p. 6

FFabiano Valentina (Italy) p. 19Fanos Vassilios (Italy) p. 12Fatati Giuseppe (Italy) p. 15Festi Davide (Italy) p. 4, 15Fogliano Vincenzo (Italy) p. 10Francavilla Ruggiero (Italy) p. 19

GGasbarrini Antonio (Italy) p. 4, 12Gassull Miguel Angel(Spain) p. 7Genazzani Alessandro D. (Italy) p. 11 Ghelardi Emilia (Italy) p. 6Gianfranceschi Gian Luigi (Italy) p. 15, 19Giovannini Marcello (Italy) p. 13Gleeson Michael (United Kingdom) p. 14Grilli Ester (Italy) p. 4Guandalini Stefano (USA) p. 12 Guarino Alfredo (Italy) p. 5, 12Guarner Francisco (Spain) p. 5, 7, 14 Guillemard Eric (France) p. 6

HHaller Dirk (Germany) p. 5Hojsak Iva (Croatia) p. 12

IIndrio Flavia (Italy) p. 12, 13Isolauri Erika (Finland) p. 12

JJirillo Emilio (Italy) p. 14Justen Peter (France) p. 7

54

Page 57: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

KKarakan Tarkan (Turkey) p. 7Kekkonen Riina (Finland) p. 6Koch Maurizio (Italy) p. 6

LLavermicocca Paola (Italy) p. 19Lionetti Paolo (Italy) p. 12Loguercio Carmelina (Italy) p. 4Louis Petra (United Kingdom) p. 7

MMarabelli Romano (Italy) p. 5Margolles Abelardo (Spain) p. 7Marigliano Vincenzo (Italy) p. 11 Marignani Massimo (Italy) p. 15Marotta Francesco (Italy) p. 11Matteuzzi Diego (Italy) p. 8Mengheri Elena (Italy) p. 6Miraglia del Giudice Michele (Italy) p. 10Morelli Lorenzo (Italy) p. 5Morisco Filomena (Italy) p. 10Mosca Fabio (Italy) p. 12

NNicoletti Claudio (United Kingdom) p. 6Nicoletti Marcello (Italy) p. 8

OOngaro Filippo (Italy) p. 15Orlando Antonella (Italy) p. 19

PPallone Francesco (Italy) p. 6Patrignani Paola (Italy) p. 14Peluso Gianfranco (Italy) p. 10Perozzi Giuditta (Italy) p. 7Picardo Mauro (Italy) p. 19Piraccini Bianca Maria (Italy) p. 19Polimeni Ascanio (Italy) p. 11

QQuigley Eamon (Ireland) p. 14

RRescigno Maria (Italy) p. 6, 7Ricciardiello Luigi (Italy) p. 10Riezzo Giuseppe (Italy) p. 19

Roberfroid Marcel (Belgium) p. 15Rossi Maddalena (Italy) p. 8Rossi Mauro (Italy) p. 14Russo Francesco (Italy) p. 19

SSaggioro Alfredo (Italy) p. 8, 15Salonen Anne (Finland) p. 6Salvini Filippo (Italy) p. 13Sanz Yolanda (Spain) p. 5, 14 Scapagnini Giovanni (Italy) p. 8, 19Scarpellini Emidio (Italy) p. 4Scott Karen P. (United Kingdom) p. 10Serafini Michele (Italy) p. 5, 10Serino Matteo (France) p. 8Severi Carola (Italy) p. 4Shamir Raanan (Israel) p. 12, 13Silano Marco (Italy) p. 14Sirtori Cesare (Italy) p. 19Swidsinski Alexander (Germany) p. 12Szajewska Hania (Poland) p. 13, 19

TTilg Herbert (Austria) p. 7Tuohy Kieran (United Kingdom) p. 14

VVandenplas Yvan (Belgium) p. 13Vanderhoof Jon (USA) p. 12 van Goudover J.B. Hans (United Kingdom) p. 12, 13Vannucci Luca (Czech Republic) p. 10 Verduci Elvira (Italy) p. 13Vincenzi Colombina (Italy) p. 19Vitaglione Paola (Italy) p. 10Virgili Fabio (Italy) p. 7

WWalker Alan (United Kingdom) p. 7

55

fa cu lt y

Page 58: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The
Page 59: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

57

index of authors

index o f a u t hor s

Page 60: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

i n d e x o f a u t hor s

58

AAguayo Maria p. 40Akkermans Louis M. A. p. 35Al Faleh Khaled p. 44Aleksandrzak-Piekarczyk Tamara p. 49Ali Babar p. 31Al-Kharfi T p. 44Allesina S. p. 44, 46, 47Alloni Rossana p. 33Alpert Carl-Alfred p. 35Alric Monique p. 29, 42Altomare Annamaria p. 33Ammoscato Francesca p. 28, 33Anabrees Jasim p. 44Anderloni A. p. 46, 47Arena Vincenzo p. 38Ascione Barbara p. 28Aumueller Eva p. 30

BBacci M.L. p. 32Baldassarre Mariella p. 40Ballarè M. p. 46, 47Balzarini M. p. 46, 47Banning Federike p. 50Barba M. p. 44, 46, 47Bardowski Jacek p. 49Bassler D p. 44Bäuerl Christine p. 35Bertino Enrico p. 36Blanquet-Diot Stéphanie p. 29Boldrini P. p. 32Bouchez Elodie p. 50Bourlioux Pierre p. 50Brasili Elisa p. 46, 51Brugère Jean-François p. 42Brun Mary p. 29Brun Paola p. 39Buccigrossi Vittoria p. 34

CCallegari Maria L. p. 42Calò G. p. 32Cannaviello Claudio p. 44Canžek Majhenic Andreja p. 52Caporaso Nicola p. 42Capuani Giorgio p. 51Cariello Rita p. 42Carmagnola S. p. 46, 47Carnevali Paola p. 45, 46Caselli M. p. 32Cassol F. p. 32Castagliuolo Ignazio p. 39Castro Erica p. 40Cavallo Donatella p. 39Chambaud Isabelle p. 50Chassaing Benoit p. 29Chitarrari Roberto p. 45Cicala Michele p. 33Cirillo Carla p. 28

Citar Manuela p. 48Cocca Silvia p. 33Coppa Giovanni Valentino p. 36, 46Corleto Vito p. 34Costabile Adele p. 45Cripps Allan p. 29Cufino Valerio p. 38Cuomo Rosario p. 28

DD'Alessandro Alessandra p. 28D'Andrea M. p. 48Darfeuille-Michaud Arlette p. 29D'Argenio Giuseppe p. 42De Magistris Laura p. 42Deidda F. p. 44, 46, 47Del Piano Mario p. 44, 46, 47, 48, 49Delle Fave Gianfranco p. 34Denis Sylvain p. 29Dessì Angelica p. 50Di Giulio Emilio p. 34Di Natale Giuseppe p. 28Donini Lorenzo M p. 46

EEfrati Cesare p. 44Ehrlich Kersti p. 37Elfakir Anissa p. 50Elli Marina p. 39Elmadfa Ibrahim p. 30Etienne-Mesmin Lucie p. 29

FFanelli Margherita p. 40Fanos Vassilios p. 50Farina Virginia p. 28Federico Alessandro p. 42Féria-Gervasio David p. 42Finamore Alberto p. 46, 51Fricker Peter p. 29Fuentes Susana p. 35

GGabrielli Orazio p. 36, 46Galeazzi Tiziana p. 36, 46Gamian Andrzej p. 51Gasbarrini Antonio p. 38Gerardi Viviana p. 38Gerritsen Jacoline p. 35Gibson Glenn p. 36, 38Gooszen Hein G. p. 35Gopal Pramod p. 31Górska-Fraczek Sabina p. 51Goyal Nupur p. 37Grilli Ester p. 32Grossi Enzo p. 42Guarino Alfredo p. 34Guarino Michele p. 33Günther Sebastian p. 33

Page 61: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

i n d e x o f a u t hor s

59

HHacin Biljana p. 48Hahne Hannes p. 35Haller Dirk p. 35Haslberger Alexander p. 30, 36Heczko Piotr p. 40Herbel Stefan p. 33Hilyakne Kadlott Maria p. 30, 43Hippe Berit p. 36Hong Zhimin p. 47Hopkins Will p. 29Hörmannsperger Gabriele p. 35Horn Peggy p. 29Hütt Pirje p. 37, 48

IIadevaia Maddalena p. 42Isolauri Erika p. 49

JJeansen Stéphanie p. 50Jia Yongjie p. 47Jofre Jaime p. 40Joyal Steven p. 46Juric Aleksandra p. 36, 38

KKamhuber Christoph p. 36Karasi Anbu K. p. 38Kiss Attila p. 30, 34, 43, 44Kõljalg Siiri p. 48Kolk Helgi p. 48Konstantinov Sergey R. p. 35Koryszewska-Baginska Anna p. 49Kozakova Hana p. 51Kryczyk Jadwiga p. 40Kupka Anna p. 40

LLabra Alan p. 40Ladan Giahi p. 30Laforgia Nicola p. 40Laterza Lucrezia p. 38Laudiero Gabriella p. 34Leyer Greg p. 31Li Guanhong p. 47Liu Siguo p. 47Livrelli Valérie p. 29Loguercio Carmela p. 42Lopetuso Loris Riccardo p. 38

MMaccari Francesca p. 36, 46Madiai Stefania p. 39Malorni Walter p. 28Mändar Reet p. 50Mango Annamaria p. 28Marignani Massimo p. 28Marotta Francesco p. 46

Marra Fabio p. 39Marteau Philippe p. 50Martines Diego p. 39Matarrese Paola p. 28Mazzone Giovanna p. 42Medina Rossi p. 40Mengheri Elena p. 46, 51Miccheli Alfredo p. 51Mikelsaar Marika p. 37, 48, 50Miller Larry p. 31Mogna Giovanni p. 44, 46, 47, 48, 49Mogna Luca p. 46, 47, 49Monsalvez Elizabeth p. 40Montecinos Hernan p. 40Moratelli Ketty p. 39Morelli Lorenzo p. 39, 42

NNaar Zoltan p. 30, 34, 35, 43, 44Nanda Dhiraj K. p. 38Nasti Anna p. 28Neri Barbara p. 46Nermes Merja p. 49Nestlberger Manuela p. 30Nicola S. p. 48Nicolini Giorgia p. 44Novo Erica p. 39

OOliva Valentina p. 34Orsello M. p. 46, 47Ouwehand Arthur p. 28, 29, 31

PPadella Lucia p. 36Pagliarulo M. p. 46, 47Pagnini Cristiano p. 34Pal Karoly p. 30, 34, 35, 43, 44Palumbo Massimo p. 45Pane M. p. 48Panneman Henk p. 35Pardo Karen p. 40Parola Maurizio p. 39Pecere Silvia p. 38Perez Gaspar p. 35Petito Valentina p. 38Petitta Chiara p. 28Pirker Angelika p. 36Pisacane Vincenza p. 42Piva A. p. 32Pizzoferrato Marco p. 38Polimeni Ascanio p. 46Poply Sarang p. 38Privat Maud p. 29Provenzano Angela p. 39Pyne David p. 29

Page 62: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

i n d e x o f a u t hor s

60

QQu Minreng p. 47Queudot Jean-Christophe p. 50

RRätsep Merle p. 37, 43, 48Rebecchi Annalisa p. 42Reifer Cheryl p. 31Ribecco Maria Teresa p. 42Rijkers Ger T. p. 35Rogelj Irena p. 48, 52Rombouts Frans M. p. 35Rööp Tiiu p. 50Roselli Marianna p. 46, 51

SSabbi Tamara p. 45Salminen Seppo p. 49Sangwan Seema p. 38Sangwan Vikas p. 31Santoro Lucia p. 46Sarnelli Giovanni p. 28Scaldaferri Franco p. 38Schrezenmeir Juerguen p. 50Schwarzer Martin p. 51Scirocco Annunziata p. 28Sciubba Fabio p. 51Sepp Epp p. 50Severi Carola p. 28, 33Sforza F. p. 44, 46, 47Sgambato Alessandro p. 48Shkut Elena p. 37, 38Singh R.R.B. p. 31Singh Rameshwar p. 38Smidt Hauke p. 35Smidt Imbi p. 43, 44Soattini L. p. 44Sofia Morena p. 34Songisepp Epp p. 37, 43, 48Soza Francisco p. 40Stewart Morgan p. 31Stigliano Egidio p. 38Stillfried Nicolas p. 40Stockenhuber Felix p. 36Strozzi G.P. p. 44, 46, 47, 48, 49Strus Magdalena p. 40Szarvas Jozsef p. 30Szen Orsolya p. 30, 34, 35, 43, 44

ŠŠtšepetova Jelena p. 50

TTafaro Angela p. 40Tari R. p. 46, 47Timmerman Harro M. p. 45Tomar Sudhir Kumar p. 31Tomassini Alberta p. 51Tomella Claudio p. 46

Tompa Gorazd p. 48Tottey William p. 42Truusalu Kai p. 48Tuccillo Concetta p. 42Tugnoli B. p. 32Turco Fabio p. 28Tzortzis George p. 36, 38

VVaira D. p. 32van Minnen L. Paul p. 35Vandekerckove Pascal p. 42Vardjan Tinkara p. 52Vera Rodrigo p. 40Verrone Maria Antonietta p. 34Vivoli Elisa p. 39Volpi Nicola p. 36, 46von Schillde Marie-Anne p. 35Vulevic Jelena p. 36, 38

WWaller Philip p. 31Warren Hilary p. 29Weiher Monika p. 35West Nic p. 29Wiecek Grazyna p. 40Wieler Lothar H. p. 33Wu Fan p. 29Wudy Anna p. 34

YYou Jinming p. 47

ZZagura Maksim p. 48Zampini Lucia p. 36, 46Zannoni A. p. 32Zilmer Mihkel p. 37, 48

Page 63: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

g e n er a l i n f ormat i o n

61

VenueUniversità Urbaniana - Terminal GianicoloVia Urbano VIII, 16 - 00165 Rome, Italy(Tel. +39 06 69889611 - Fax +39 06 69881871)www.urbaniana.edu

DatesSeptember 11-13, 2011

LanguageThe official language of the Meeting is English

ClothingInformal for all occasions except for the gala dinner

ClimateSeptember in Rome is still hot but unpredictable. For all the excursions we suggest casual wear and outdoorshoes

BadgesAll participants, accompanying persons and exhibitors are kindly requested to wear their badges throughout theMeeting area in order to be admitted to the scientific sessions and to all the activities of the Meeting

Registration FeesFees (20% VAT included)

Participants € 600,00Under 35* € 324,00Accompanying Persons € 290,00Pediatric Day** € 240,00Daily Registration € 240,00Gala dinner € 150,00

* the applicant’s registration form must be accompanied by a copy of an official document** If not registered to the general Meeting

Registration fees include:

Participants:- Admission to scientific sessions, technical exhibition- Final programme- Proceedings and abstract book- Coffee corners and lunches- Opening ceremony and welcome cocktail- Certificate of attendance- Italian CME certificate (to whom entitled)

Accompanying Persons- Opening ceremony and welcome cocktail- Two half day tours (on request)

Participants are kindly requested to wear their badges during all meeting activities and social events: No badge,no entry

Banking and ExchangeThe Italian monetary system is the Euro. Foreign currency may be exchanged at banks during normal bankinghours, at hotels, at airports and in exchange offices. All major credit cards are accepted in most hotels, restaurantsand shops

Liability and InsuranceThe Meeting Organisers cannot accept liability for personal injuries or for loss of, or damage to, property belongingto meeting participants (or their accompanying persons), either during or as a result of the meeting. Please checkthe validity of your own insurance

Page 64: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

62

g e n e r al i n f ormat i o n

Visa RequirementsAll visitors entering Italy must be in possession of a valid passport. For European Community citizens an IdentityCard is sufficient. Participants should check the Italian Embassy or Consulate in their country regarding visa andhealth certificate requirements.

Official Letter of InvitationOfficial letters of invitation designed to help overcome administrative difficulties in certain countries are availableon the Meeting website. It must be understood that such letters do not represent a commitment on behalf of theOrganising Committee or Conference to provide any financial assistance

Certificate of AttendanceCertificates of attendance will be available for all registered participants at the registration desk from September 13, 2011

Food and BeveragesBusiness lunches during breaks and coffee/tea service (as indicated in the programme) are included in theregistration fee

ParkingCars can be parked at terminal Gianicolo. Participants to the Meeting will have a special rate. To obtain it, pleasecontact the Organising Secretariat

About RomeRome is the capital city of Italy and of the Lazio region, as well as the country's largest and most populouscomune, with more than 2.7 million residents. The metropolitan area has a population of about 4 million. It islocated in the central-western portion of the Italian peninsula, where the river Aniene joins the Tiber. The Mayorof Rome is Gianni Alemanno. An enclave of Rome is the State of the Vatican City, the sovereign territory of theHoly See. It is the smallest nation in the world, and the capital of the only religion to have representation in theUnited Nations (as a non-member observer state). Rome, Caput mundi ("capital of the world"), la Città Eterna("the Eternal City"), Limen Apostolorum ("threshold of the Apostles"), la città dei sette colli ("the city of the sevenhills") or simply l'Urbe ("the City"), is thoroughly modern and cosmopolitan. As one of the few major Europeancities that escaped World War II relatively unscathed, central Rome remains essentially Renaissance and Baroquein character. The Historic Centre of Rome is listed by UNESCO as a World Heritage Site

Airport InformationRome can easily be reached by plane and is served by two international airports.Participants can fly into Rome via Leonardo da Vinci Airport, located in Fiumicino, 34 km from Rome's historiccity centre or via Ciampino Airport, situated 15 km southeast of central Rome

Access to Rome from the AirportsAccess from Leonardo da Vinci Airport:The airport is served by the Leonardo Express train operated by Trenitalia, available at the airport terminal. Thetrip takes 30 minutes (nonstop) to Termini Station in Rome - there are two such connections per hour.Alternatively, local trains leave once every 15 minutes, stopping at all train stations. You may have to change atTrastevere, Ostiense (Metro Piramide) or Tuscolana.Rental cars are available in the airport terminal from all the usual companies

Access from Ciampino Airport:There is no rail transport at Ciampino Airport. The options are to take a bus to a rail station (either metro orregular train) or to take a bus or taxi all the way

All the way by road transport:• Terravision runs a direct bus service to Termini. The price is € 8,00 c.a. one-way or € 13,50 c.a. return, taking 40

minutes (about 20 services a day). Despite timing buses to connect with flights, passengers on the return trip fromTermini are asked to board the bus 2.5 hours before their flight's departure time. The last bus is at 07:20 p.m.Terravision also offers buses from Fiumicino airport to Termini, and a transfer bus between the two airports.

• Schiaffini also runs direct buses to Termini station for € 5,00 one-way, taking 40 minutes, but with far fewerdepartures than Terravision (see above). These buses are not mentioned on the airport website but they can befound on Schiaffini's own site

• BusShuttle runs a service similar to Terravision. Their stop near Termini is about 20 metres up the road fromTerravision's. Cost is € 6,00 for a single

• The fixed fare for a taxi ride to the city centre (inside the Aurelian Walls) is € 30,00, according to the officialagreement between Roman taxi driver associations and Rome municipality. It is advisable to negotiate the totalprice including luggage supplements before boarding the taxi

Rental cars are available in the airport terminal from all the usual companies

Page 65: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

63

g e n er a l i n f ormat i o n

How to get to the Meeting venue

From Termini Rail Station:By taxi - We recommend you to only use licensed taxis available outside the station.

Telephone number main taxis companies:06 - 3570 Radio Taxi06 - 5551 Samarcanda06 - 4994 La Capitale

By public transport - Arriving from Termini Railways Station - BUS 64 stop at Lgt. Sassia (S. Spirito Hospital) -350 metres walking

From Leonardo da Vinci Airport:By taxi – We recommend you to only use licensed taxis available outside the airport.

Telephone number main taxis companies:06 - 3570 Radio Taxi06 - 5551 Samarcanda06 - 4994 La Capitale

By public transport - Follow the signs for “Station” of Leonardo express.Take the train for Stazione Termini and get off at the Station. Take the BUS 64 stop at Lgt. Sassia (S. SpiritoHospital) - 350 metres walking

From Ciampino Airport:We advice to take a taxi available outside the airport. We recommend you to only use licensed taxis availableoutside the airport

Transportation in the CityRome has a very efficient transportation system that services the entire city, which includes the Metro networkas well as buses, trains and taxis

Organising Secretary Desk at the Meeting venue will be open as follows:

DAY DATE FROM TOSunday September 11 12.00 p.m. 7.00 p.m. Monday September 12 8.00 a.m. 7.00 p.m.Tuesday September 13 8.00 a.m. 3.00 p.m.

Organising SecretariatPlease do not hesitate to contact the Organisers if you require any additional information or assistance. Please address all correspondence to:

e MEETING&CONSULTINGVia M. Mercati, 3300197 Rome, ItalyPhone: +39 06 80693320 Fax: +39 06 3231136www.emec-roma.come-mail: [email protected]: www.probiotics-prebiotics-newfood.org

Page 66: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

s c i en t i f i c i n f ormat i o n

64

Oral communicationsOral communications sessions are scheduled as follows:

- ORAL COMMUNICATIONS 1:September 12, 2011 AULA C from 10.00 a.m. to 11.30 a.m.

- ORAL COMMUNICATIONS 2:September 13, 2011 AULA C from 10.30 a.m. to 01.00 p.m.

PostersPoster authors are kindly requested to hang the poster at the poster area from 3.00 p.m. on September 11 andremove it after 1.00 p.m. on September 13. Your position will be indicated in the poster area

Slide centersAll speakers and authors must deliver their presentation (CD Rom, USB) 2 hours in advance or the day beforetheir speech to the slide centers

CME for Italian delegatesGli attestati riportanti i crediti ECM, dopo attenta verifica della partecipazione e dell'apprendimento, saranno inviation-line dopo la chiusura dell'evento. Il partecipante riceverà una password che gli permetterà di scaricarel'attestato con i crediti conseguiti (10,5) per la professione di Medico Chiurugo, Infermiere, Infermiere pediatrico

Page 67: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

65

T HE M E E T I N G WAS MADE POS S I B L E T H A N KS TO T H E C ON TR I B U T I ON P R OV I D E d B Y:

m a j o r s pon sors

AG PHARMAANGELINIDANONE

ITALCHIMICIPROBIOTICAL

SANOFI AVENTISYAKULT

ACQUA E TERME DI ULIVETOAIIPA

BIOACTIVALBIOFARMA

BRACCOCEC EDITORE

DKSHFARMACEUTICI PROCEMSA

GIULIANIGNOSIS

LESAFFRE HUMAN CARE MEAD JOHNSON NUTRITION ITALIA

NATHURANÓOS

NUTRICIAPARMALATSIGMA-TAU

S.I . I .T .SOFAR

TEKNOSCIENCEVITIS PHARMA

ZAMBON ITALIA

Page 68: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

e x h i b i t i o n a r e a

66

Floor plan, ground level

Floor plan, level 1

MAIN ENTRANCE

AULA MAGNA

ORGANISINGSECRETARIAT

EXHIBITORS

1) YAKULT2) NÓOS3) ITALCHIMICI4) LESAFFRE5) ANGELINI6) SOFAR7) ZAMBON7bis) PROBIOTICAL 8) AG PHARMA/VITIS PHARMA9) AG PHARMA/VITIS PHARMA10) FARMACEUTICI PROCEMSA11) BIOFARMA12) BRACCO13) SANOFI AVENTIS14) SANOFI AVENTIS

A) MEAD JOHNSON NUTRITION

SLIDE CENTER

COFFEE CORNER

AREA

EXHIBITORS

15) DKSH16) S.I.I.T.17) BIOACTIVAL

AULA C

AULA NEWMAN

COFFEE CORNER/LUNCH AREA

7 6 5 4 3 2 1

7bis

9 8

13 1411

12

10

17 16 15

SLID

ECE

NTE

R

A

Page 69: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

67

no te s

Page 70: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

68

n ot e s

Page 71: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The
Page 72: Università Urbaniana probiotics prebiotics · 08.30 - 10.30 a.m. MICROBIOTA AND INTESTINAL HEALTH Chairman: R. Crittenden (Finland) Microbiota in Health and Disease W.M. de Vos (The

Recommended