Network


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

Hotspot


Dive into the research topics where Alain L. Servin is active.

Publication


Featured researches published by Alain L. Servin.


Gut | 1994

Lactobacillus acidophilus LA 1 binds to cultured human intestinal cell lines and inhibits cell attachment and cell invasion by enterovirulent bacteria.

Marie-Françoise Bernet; Dominique Brassart; Jean-Richard Neeser; Alain L. Servin

Four human Lactobacillus acidophilus strains were tested for their ability to adhere onto human enterocyte like Caco-2 cells in culture. The LA 1 strain exhibited a high calcium independent adhesive property. This adhesion onto Caco-2 cells required a proteinaceous adhesion promoting factor, which was present in the spent bacterial broth culture supernatant. LA 1 strain also strongly bound to the mucus secreted by the homogeneous cultured human goblet cell line HT29-MTX. The inhibitory effect of LA 1 organisms against Caco-2 cell adhesion and cell invasion by a large variety of diarrhoeagenic bacteria was investigated. As a result, the following dose dependent inhibitions were obtained: (a) against the cell association of enterotoxigenic, diffusely adhering and enteropathogenic Escherichia coli, and Salmonella typhimurium; (b) against the cell invasion by enteropathogenic Escherichia coli, Yersinia pseudotuberculosis, and Salmonella typhimurium. Incubations of L acidophilus LA 1 before and together with enterovirulent E coli were more effective than incubation after infection by E coli.


Clinical Microbiology Reviews | 2006

The Front Line of Enteric Host Defense against Unwelcome Intrusion of Harmful Microorganisms: Mucins, Antimicrobial Peptides, and Microbiota

Vanessa Liévin-Le Moal; Alain L. Servin

SUMMARY The intestinal tract is a complex ecosystem that combines resident microbiota and the cells of various phenotypes with complex metabolic activities that line the epithelial wall. The intestinal cells that make up the epithelium provide physical and chemical barriers that protect the host against the unwanted intrusion of microorganisms that hijack the cellular molecules and signaling pathways of the host and become pathogenic. Some of the organisms making up the intestinal microbiota also have microbicidal effects that contribute to the barrier against enteric pathogens. This review describes the two cell lineages present in the intestinal epithelium: the goblet cells and the Paneth cells, both of which play a pivotal role in the first line of enteric defense by producing mucus and antimicrobial peptides, respectively. We also analyze recent insights into the intestinal microbiota and the mechanisms by which some resident species act as a barrier to enteric pathogens. Moreover, this review examines whether the cells producing mucins or antimicrobial peptides and the resident microbiota act in partnership and whether they function individually and/or synergistically to provide the host with an effective front line of defense against harmful enteric pathogens.


Gut | 2000

Bifidobacterium strains from resident infant human gastrointestinal microflora exert antimicrobial activity

V. Liévin; I. Peiffer; S. Hudault; Florence Rochat; Dominique Brassart; Jean-Richard Neeser; Alain L. Servin

BACKGROUND AND AIMS The gastrointestinal microflora exerts a barrier effect against enteropathogens. The aim of this study was to examine if bifidobacteria, a major species of the human colonic microflora, participates in the barrier effect by developing antimicrobial activity against enterovirulent bacteria. METHODS Antibacterial activity was examined in vitro against a wide range of Gram negative and Gram positive pathogens. Inhibition ofSalmonella typhimurium SL1334 cell association and cell invasion was investigated in vitro using Caco-2 cells. Colonisation of the gastrointestinal tract in vivo by bifidobacteria was examined in axenic C3/He/Oujco mice. Antimicrobial activity was examined in vivo in axenic C3/He/Oujco mice infected by the lethal S typhimurium C5 strain. RESULTS Fourteen human bifidobacterium strains isolated from infant stools were examined for antimicrobial activity. Two strains (CA1 and F9) expressed antagonistic activity against pathogens in vitro, inhibited cell entry, and killed intracellular S typhimurium SL1344 in Caco-2 cells. An antibacterial component(s) produced by CA1 and F9 was found to be a lipophilic molecule(s) with a molecular weight of less than 3500. In the axenic C3/He/Oujco mice, CA1 and F9 strains colonised the intestinal tract and protected mice against S typhimurium C5 lethal infection. CONCLUSION Several bifidobacterium strains from resident infant human gastrointestinal microflora exert antimicrobial activity, suggesting that they could participate in the “barrier effect” produced by the indigenous microflora.


Best Practice & Research in Clinical Gastroenterology | 2003

Adhesion of probiotic strains to the intestinal mucosa and interaction with pathogens

Alain L. Servin; Marie-Hélène Coconnier

Probiotic lactic acid strains are live micro-organisms that, when consumed in adequate amounts as part of food, confer a health benefit on the host. The scientific basis for the use of selected probiotic strains has only recently been firmly established, and appropriate and well-conducted experimental in vitro and in vivo studies, as well as clinical studies, are now beginning to be published, especially with regard to the effectiveness of probiotic strains in antagonizing pathogens. In particular, experimental data have allowed new insights into selected probiotic strains that express strain-specific probiotic properties and into the mechanism of action of these strains. The objective of this review is to analyse the in vitro or in vivo experimental studies in which the antimicrobial activity of selected Lactobacillus and Bifidobacterium strains has been documented.


Microbiology | 1992

Adhesion of human Lactobacillus acidophilus strain LB to human enterocyte-like Caco-2 cells

Gilles Chauvière; Marie-Hélène Coconnier; Sophie Kernéis; Jacky Fourniat; Alain L. Servin

Twenty-five strains of lactobacilli were tested for their ability to adhere to human enterocyte-like Caco-2 cells in culture. Seven Lactobacillus strains adhered well to the Caco-2 cells, of which three possessed calcium-independent adhesion properties. A high level of calcium-independent adhesion was observed with the human stool isolate Lactobacillus acidophilus strain LB. Scanning electron microscopy revealed that this strain adhered to the apical brush border of the cells. Adhesion increased in parallel with the morphological and functional differentiation of the Caco-2 cells. Two Lactobacillus components were involved in this adhesion. One was protease-resistant and bacterial-surface-associated; the other was heat-stable, extracellular and protease-sensitive.


Clinical Microbiology Reviews | 2005

Pathogenesis of Afa/Dr Diffusely Adhering Escherichia coli

Alain L. Servin

SUMMARY Over the last few years, dramatic increases in our knowledge about diffusely adhering Escherichia coli (DAEC) pathogenesis have taken place. The typical class of DAEC includes E. coli strains harboring AfaE-I, AfaE-II, AfaE-III, AfaE-V, Dr, Dr-II, F1845, and NFA-I adhesins (Afa/Dr DAEC); these strains (i) have an identical genetic organization and (ii) allow binding to human decay-accelerating factor (DAF) (Afa/DrDAF subclass) or carcinoembryonic antigen (CEA) (Afa/DrCEA subclass). The atypical class of DAEC includes two subclasses of strains; the atypical subclass 1 includes E. coli strains that express AfaE-VII, AfaE-VIII, AAF-I, AAF-II, and AAF-III adhesins, which (i) have an identical genetic organization and (ii) do not bind to human DAF, and the atypical subclass 2 includes E. coli strains that harbor Afa/Dr adhesins or others adhesins promoting diffuse adhesion, together with pathogenicity islands such as the LEE pathogenicity island (DA-EPEC). In this review, the focus is on Afa/Dr DAEC strains that have been found to be associated with urinary tract infections and with enteric infection. The review aims to provide a broad overview and update of the virulence aspects of these intriguing pathogens. Epidemiological studies, diagnostic techniques, characteristic molecular features of Afa/Dr operons, and the respective role of Afa/Dr adhesins and invasins in pathogenesis are described. Following the recognition of membrane-bound receptors, including type IV collagen, DAF, CEACAM1, CEA, and CEACAM6, by Afa/Dr adhesins, activation of signal transduction pathways leads to structural and functional injuries at brush border and junctional domains and to proinflammatory responses in polarized intestinal cells. In addition, uropathogenic Afa/Dr DAEC strains, following recognition of β1 integrin as a receptor, enter epithelial cells by a zipper-like, raft- and microtubule-dependent mechanism. Finally, the presence of other, unknown virulence factors and the way that an Afa/Dr DAEC strain emerges from the human intestinal microbiota as a “silent pathogen” are discussed.


Applied and Environmental Microbiology | 2005

pH-, Lactic Acid-, and Non-Lactic Acid-Dependent Activities of Probiotic Lactobacilli against Salmonella enterica Serovar Typhimurium

Domitille Fayol-Messaoudi; Cedric N. Berger; Marie-Hélène Coconnier-Polter; Vanessa Liévin-Le Moal; Alain L. Servin

ABSTRACT The mechanism(s) underlying the antibacterial activity of probiotic Lactobacillus strains appears to be multifactorial and includes lowering of the pH and the production of lactic acid and of antibacterial compounds, including bacteriocins and nonbacteriocin, non-lactic acid molecules. Addition of Dulbeccos modified Eagles minimum essential medium to the incubating medium delays the killing activity of lactic acid. We found that the probiotic strains Lactobacillus johnsonii La1, Lactobacillus rhamnosus GG, Lactobacillus casei Shirota YIT9029, L. casei DN-114 001, and L. rhamnosus GR1 induced a dramatic decrease in the viability of Salmonella enterica serovar Typhimurium SL1344 mainly attributable to non-lactic acid molecule(s) present in the cell-free culture supernatant (CFCS). These molecules were more active against serovar Typhimurium SL1344 in the exponential growth phase than in the stationary growth phase. We also showed that the production of the non-lactic acid substance(s) responsible for the killing activity was dependent on growth temperature and that both unstable and stable substances with killing activity were present in the CFCSs. We found that the complete inhibition of serovar Typhimurium SL1344 growth results from a pH-lowering effect.


Gut | 2001

Escherichia coli strains colonising the gastrointestinal tract protect germfree mice against Salmonella typhimurium infection.

S Hudault; J Guignot; Alain L. Servin

BACKGROUND Escherichia coli is part of the normal gastrointestinal microflora which exerts a barrier effect against enteropathogens. SeveralE coli strains develop a protective effect against other Enterobacteriaceae. AIMS TwoE coli strains, EM0, a human faecal strain, and JM105 K-12 were tested for their ability to prevent in vivo and in vitro infection by Salmonella typhimurium C5. METHODS Inhibition of C5 cell invasion by E coli was investigated in vitro using Caco-2/TC7 cells. The protective effect ofE coli was examined in vivo in germfree or conventional C3H/He/Oujco mice orally infected by the lethal strain C5. RESULTS EMO expresses haemolysin and cytotoxic necrotising factor in vitro. In vitro, the two strains did not prevent the growth of C5 by secreted microcins or modified cell invasion of C5. In vivo, establishment of EM0 or JM105 in the gut of germfree mice resulted in a significant increase in the number of surviving mice: 11/12 and 9/12, respectively, at 58 days after infection (2×106/mouse) versus 0/12 in control germfree group at 13 days after infection. Colonisation level and translocation rate of C5 were significantly reduced during the three days after infection. In contrast, no reduction in faecal C5 excretion was observed in C5 infected conventional mice (1×108/mouse) receiving the EM0 or JM105 cultures daily. CONCLUSIONS Establishment of E coli strains, which do not display antimicrobial activity, protects germfree mice against infection and delays the establishment of C5 in the gut. Possible mechanisms of defence are discussed.


Gut | 2002

Lactobacillus acidophilus (strain LB) from the resident adult human gastrointestinal microflora exerts activity against brush border damage promoted by a diarrhoeagenic Escherichia coli in human enterocyte-like cells

V.Lievin‐Le Moal; Raymonde Amsellem; Alain L. Servin; Marie-Hélène Coconnier

Background and aims: The normal gastrointestinal microflora exerts a barrier effect against enteropathogens. The aim of this study was to examine whether lactobacilli, a minor genus of the resident gut microflora, exerts a protective effect against the cellular injuries promoted by the diarrhoeagenic Afa/Dr diffusely adhering Escherichia coli (Afa/Dr DAEC) C1845 strain in human intestinal cells. Methods: Cultured human intestinal fully differentiated enterocyte-like Caco-2/TC7 cells were used. Antibacterial activity was examined by measuring the viability of the adhering C1845 bacteria. The distribution of brush border associated cytoskeleton and functional proteins was examined by immunofluorescence labelling coupled to confocal laser scanning microscopy analysis. Results: The activity of Lactobacillus acidophilus strain LB isolated from the resident human gastrointestinal microflora was examined. A dose dependent decrease in viability of C1845 bacteria was observed after both direct contact in vitro between the spent culture supernatant (LB-SCS) and the bacteria, and when the bacteria were adherent on Caco-2/TC7 cells. Protection against the C1845 induced alterations in expression of F-actin, sucrase-isomaltase, dipeptidylpeptidase IV, alkaline phosphatase, and fructose transporter alterations was observed when the cells were exposed to LB-SCS. Conclusion:L acidophilus strain isolated from the resident adult human gastrointestinal microflora, together with its antimicrobial activity, exerts a protective effect against the brush border lesions promoted by the diarrhoeagenic Afa/Dr DAEC strain C1845.


Applied and Environmental Microbiology | 2000

Antagonistic activity of Lactobacillus acidophilus LB against intracellular Salmonella enterica serovar Typhimurium infecting human enterocyte-like Caco-2/TC-7 cells

Marie-Hélène Coconnier; Vanessa Lievin; Mathie Lorrot; Alain L. Servin

ABSTRACT To gain further insight into the mechanism by which lactobacilli develop antimicrobial activity, we have examined howLactobacillus acidophilus LB inhibits the promoted cellular injuries and intracellular lifestyle of Salmonella entericaserovar Typhimurium SL1344 infecting the cultured, fully differentiated human intestinal cell line Caco-2/TC-7. We showed that the spent culture supernatant of strain LB (LB-SCS) decreases the number of apical serovar Typhimurium-induced F-actin rearrangements in infected cells. LB-SCS treatment efficiently decreased transcellular passage ofS. enterica serovar Typhimurium. Moreover, LB-SCS treatment inhibited intracellular growth of serovar Typhimurium, since treated intracellular bacteria displayed a small, rounded morphology resembling that of resting bacteria. We also showed that LB-SCS treatment inhibits adhesion-dependent serovar Typhimurium-induced interleukin-8 production.

Collaboration


Dive into the Alain L. Servin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isabelle Beau

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge