Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Gut Microbes ; 14(1): 2110821, 2022.
Article in English | MEDLINE | ID: mdl-35960855

ABSTRACT

There is now strong evidence to support the interest in using lactic acid bacteria (LAB)in particular, strains of lactococci and lactobacilli, as well as bifidobacteria, for the development of new live vectors for human and animal health purposes. LAB are Gram-positive bacteria that have been used for millennia in the production of fermented foods. In addition, numerous studies have shown that genetically modified LAB and bifodobacteria can induce a systemic and mucosal immune response against certain antigens when administered mucosally. They are therefore good candidates for the development of new mucosal delivery strategies and are attractive alternatives to vaccines based on attenuated pathogenic bacteria whose use presents health risks. This article reviews the most recent research and advances in the use of LAB and bifidobacteria as live delivery vectors for human and animal health.


Subject(s)
Gastrointestinal Microbiome , Lactobacillales , Vaccines , Animals , Bifidobacterium/genetics , Genetic Vectors/genetics , Humans , Lactobacillales/genetics
2.
Microorganisms ; 10(5)2022 May 21.
Article in English | MEDLINE | ID: mdl-35630507

ABSTRACT

Functional foods are classified as traditional or staple foods that provide an essential nutritional level and share potentially positive effects on host health, including the reduction of disease by optimizing the immune system's ability to prevent and control infections by pathogens, as well as pathologies that cause functional alterations in the host. This chapter reviews the most recent research and advances in this area and discusses some perspectives on what the future holds in this area.

3.
Biomolecules ; 11(10)2021 09 24.
Article in English | MEDLINE | ID: mdl-34680035

ABSTRACT

The characteristics of innate immunity have recently been investigated in depth in several research articles, and original findings suggest that innate immunity also has a memory capacity, which has been named "trained immunity". This notion has revolutionized our knowledge of the innate immune response. Thus, stimulation of trained immunity represents a therapeutic alternative that is worth exploring. In this context, probiotics, live microorganisms which when administered in adequate amounts confer a health benefit on the host, represent attractive candidates for the stimulation of trained immunity; however, although numerous studies have documented the beneficial proprieties of these microorganisms, their mechanisms of action are not yet fully understood. In this review, we propose to explore the putative connection between probiotics and stimulation of trained immunity.


Subject(s)
Epigenesis, Genetic/immunology , Immunity, Innate/immunology , Probiotics/therapeutic use , Epigenesis, Genetic/drug effects , Humans , Immunity, Innate/drug effects
4.
Microorganisms ; 9(8)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34442713

ABSTRACT

The intestinal epithelium serves as an effective barrier against the external environment, hampering the passage of potentially harmful substances (such as pathogenic microbes) that could trigger an exacerbated host immune response. The integrity of this barrier is thus essential for the maintenance of proper intestinal homeostasis and efficient protective reactions against chemical and microbial challenges. The principal consequence of intestinal barrier defects is an increase in intestinal permeability, which leads to an increased influx of luminal stressors, such as pathogens, toxins, and allergens, which in turn trigger inflammation and immune response. The fine and fragile balance of intestinal homeostasis can be altered by multiple factors that regulate barrier function, many of which are poorly understood. This review will address the role of gut microbiota as well as food supplements (such as probiotics, prebiotics, and synbiotics) in modulating gut health and regulating intestinal barrier function. In particular, we will focus on three human pathologies: inflammatory bowel disease, irritable bowel syndrome, and food allergy.

5.
Vaccines (Basel) ; 9(5)2021 May 06.
Article in English | MEDLINE | ID: mdl-34066443

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 virus (SARS-CoV-2) infection, the causative agent of COVID-19, now represents the sixth Public Health Emergency of International Concern (PHEIC)-as declared by the World Health Organization (WHO) since 2009. Considering that SARS-CoV-2 is mainly transmitted via the mucosal route, a therapy administered by this same route may represent a desirable approach to fight SARS-CoV-2 infection. It is now widely accepted that genetically modified microorganisms, including probiotics, represent attractive vehicles for oral or nasal mucosal delivery of therapeutic molecules. Previous studies have shown that the mucosal administration of therapeutic molecules is able to induce an immune response mediated by specific serum IgG and mucosal IgA antibodies along with mucosal cell-mediated immune responses, which effectively concur to neutralize and eradicate infections. Therefore, advances in the modulation of mucosal immune responses, and in particular the use of probiotics as live delivery vectors, may encourage prospective studies to assess the effectiveness of genetically modified probiotics for SARS-CoV-2 infection. Emerging trends in the ever-progressing field of vaccine development re-emphasize the contribution of adjuvants, along with optimization of codon usage (when designing a synthetic gene), expression level, and inoculation dose to elicit specific and potent protective immune responses. In this review, we will highlight the existing pre-clinical and clinical information on the use of genetically modified microorganisms in control strategies against respiratory and non-respiratory viruses. In addition, we will discuss some controversial aspects of the use of genetically modified probiotics in modulating the cross-talk between mucosal delivery of therapeutics and immune system modulation.

6.
Microorganisms ; 9(5)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068653

ABSTRACT

The gut microbiota plays an important role in maintaining homeostasis in the human body, and the disruption of these communities can lead to compromised host health and the onset of disease. Current research on probiotics is quite promising and, in particular, these microorganisms have demonstrated their potential for use as adjuvants for the treatment of colorectal cancer. This review addresses the possible applications of probiotics, postbiotics, synbiotics, and next-generation probiotics in colorectal cancer research.

8.
J Gastroenterol ; 51(9): 862-73, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26749362

ABSTRACT

BACKGROUND: Chronic intestinal inflammation alters host physiology and could lead to colorectal cancer (CRC). We have previously reported beneficial effects of the probiotic strain of Lactobacillus casei BL23 in different murine models of intestinal inflammation. In addition, there is an emerging interest on the potential beneficial effects of probiotics to treat CRC. We thus explored whether L. casei BL23 displays protective effects on CRC. METHODS: Mice were subcutaneously injected with 1,2-dimethylhydrazine (DMH) weekly during 10 weeks and orally administered with L. casei BL23 in the drinking water until the 10th week. Multiple plaque lesions in the large intestine were observed macroscopically and counted and intestinal tissues were also histologically analyzed. Finally, T-cell populations and cytokine production were evaluated after co-incubation of L. casei BL23 with spleen cells from non-treated mice to determine the immuno-modulatory effects of this bacterium. RESULTS: Our results show that oral treatment with this probiotic bacterium modulates host immune responses and significantly protect mice against DMH-induced CRC. This protection may be associated with the modulation of regulatory T-cells towards a Th17-biased immune response accompanied by the expression of regulatory cytokines (IL-6, IL-17, IL-10 and TGF-ß), as demonstrated in L. casei BL23-treated splenocytes, but also with the colonic expression of IL-22 observed in vivo on L. casei BL23-treated mice; suggesting the induction of a fine-tune Th17-biased response. CONCLUSIONS: Altogether our results reveal the high potential of L. casei BL23 to treat CRC and opens new frontiers for the study of immunomodulatory functions of probiotics.


Subject(s)
Colorectal Neoplasms/prevention & control , Lacticaseibacillus casei , Probiotics/therapeutic use , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , 1,2-Dimethylhydrazine , Animals , Biomarkers/metabolism , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Cytokines/metabolism , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
9.
BMC Microbiol ; 15: 112, 2015 May 25.
Article in English | MEDLINE | ID: mdl-26003173

ABSTRACT

BACKGROUND: Mechanisms underlying the transition from commensalism to virulence in Enterococcus faecalis are not fully understood. We previously identified the enterococcal leucine-rich protein A (ElrA) as a virulence factor of E. faecalis. The elrA gene is part of an operon that comprises four other ORFs encoding putative surface proteins of unknown function. RESULTS: In this work, we compared the susceptibility to phagocytosis of three E. faecalis strains, including a wild-type (WT), a ΔelrA strain, and a strain overexpressing the whole elr operon in order to understand the role of this operon in E. faecalis virulence. While both WT and ΔelrA strains were efficiently phagocytized by RAW 264.7 mouse macrophages, the elr operon-overexpressing strain showed a decreased capability to be internalized by the phagocytic cells. Consistently, the strain overexpressing elr operon was less adherent to macrophages than the WT strain, suggesting that overexpression of the elr operon could confer E. faecalis with additional anti-adhesion properties. In addition, increased virulence of the elr operon-overexpressing strain was shown in a mouse peritonitis model. CONCLUSIONS: Altogether, our results indicate that overexpression of the elr operon facilitates the E. faecalis escape from host immune defenses.


Subject(s)
Bacterial Proteins/genetics , Enterococcus faecalis/physiology , Operon , Peritonitis/microbiology , Phagocytosis , Animals , Bacterial Adhesion , Bacterial Proteins/metabolism , Cell Line , Disease Models, Animal , Enterococcus faecalis/genetics , Enterococcus faecalis/pathogenicity , Gene Expression Regulation, Bacterial , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/veterinary , Macrophages/metabolism , Mice , Virulence
10.
Mol Cell Proteomics ; 12(12): 3935-47, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24002364

ABSTRACT

Surface proteins of Gram-positive bacteria play crucial roles in bacterial adhesion to host tissues. Regarding commensal or probiotic bacteria, adhesion to intestinal mucosa may promote their persistence in the gastro-intestinal tract and their beneficial effects to the host. In this study, seven Lactococcus lactis strains exhibiting variable surface physico-chemical properties were compared for their adhesion to Caco-2 intestinal epithelial cells. In this test, only one vegetal isolate TIL448 expressed a high-adhesion phenotype. A nonadhesive derivative was obtained by plasmid curing from TIL448, indicating that the adhesion determinants were plasmid-encoded. Surface-exposed proteins in TIL448 were analyzed by a proteomic approach consisting in shaving of the bacterial surface with trypsin and analysis of the released peptides by LC-MS/MS. As the TIL448 complete genome sequence was not available, the tryptic peptides were identified by a mass matching approach against a database including all Lactococcus protein sequences and the sequences deduced from partial DNA sequences of the TIL448 plasmids. Two surface proteins, encoded by plasmids in TIL448, were identified as candidate adhesins, the first one displaying pilin characteristics and the second one containing two mucus-binding domains. Inactivation of the pilin gene abolished adhesion to Caco-2 cells whereas inactivation of the mucus-binding protein gene had no effect on adhesion. The pilin gene is located inside a cluster of four genes encoding two other pilin-like proteins and one class-C sortase. Synthesis of pili was confirmed by immunoblotting detection of high molecular weight forms of pilins associated to the cell wall as well as by electron and atomic force microscopy observations. As a conclusion, surface proteome analysis allowed us to detect pilins at the surface of L. lactis TIL448. Moreover we showed that pili appendages are formed and involved in adhesion to Caco-2 intestinal epithelial cells.


Subject(s)
Bacterial Proteins/genetics , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Gene Expression Regulation, Bacterial , Lactococcus lactis/genetics , Proteome/genetics , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Amino Acid Sequence , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Bacterial Adhesion , Bacterial Proteins/metabolism , Caco-2 Cells , Chromatography, Liquid , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Humans , Intestines/cytology , Intestines/microbiology , Lactococcus lactis/metabolism , Lactococcus lactis/ultrastructure , Microscopy, Electron , Molecular Sequence Annotation , Molecular Sequence Data , Multigene Family , Peptide Fragments/analysis , Plasmids , Probiotics/chemistry , Proteolysis , Proteome/metabolism , Tandem Mass Spectrometry , Trypsin/chemistry
11.
Mol Microbiol ; 89(3): 518-31, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23772975

ABSTRACT

Most bacteria of the genus Streptococcus are opportunistic pathogens, and some of them produce extracellular DNases, which may be important for virulence. Genome analyses of Streptococcus agalactiae (GBS) neonate isolate NEM316 revealed the presence of seven genes putatively encoding secreted DNases, although their functions, if any, are unknown. In this study, we observed that respiration growth of GBS led to the extracellular accumulation of a putative nuclease, identified as being encoded by the gbs0661 gene. When overproduced in Lactococcus lactis, the protein was found to be a divalent cation-requiring, pH-stable and heat-stable nuclease that we named Nuclease A (NucA). Substitution of the histidine(148) by alanine reduced nuclease activity of the GBS wild-type strain, indicating that NucA is the major nuclease ex vivo. We determined that GBS is able to degrade the DNA matrix comprising the neutrophil extracellular trap (NET). The nucA(H148A) mutant was impaired for this function, implicating NucA in the virulence of GBS. In vivo infection studies confirmed that NucA is required for full infection, as the mutant strain allowed increased bacterial clearance from lung tissue and decreased mortality in infected mice. These results show that NucA is involved in NET escape and is needed for full virulence.


Subject(s)
Bacterial Proteins/metabolism , Deoxyribonucleases/metabolism , Neutrophils/immunology , Streptococcal Infections/immunology , Streptococcus agalactiae/pathogenicity , Amino Acid Sequence , Amino Acid Substitution , Animals , Bacterial Proteins/genetics , Deoxyribonucleases/genetics , Humans , Immune Evasion , Lung/microbiology , Mice , Molecular Sequence Data , Neutrophils/microbiology , Streptococcal Infections/microbiology , Streptococcus agalactiae/enzymology , Streptococcus agalactiae/genetics , Toll-Like Receptor 9/immunology , Virulence
12.
FEMS Microbiol Lett ; 344(1): 1-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23600579

ABSTRACT

Lactic acid bacteria (LAB) represent a heterogeneous group of microorganisms naturally present in many foods and those have proved to be effective mucosal delivery vectors. Moreover, some specific strains of LAB exert beneficial properties (known as probiotic effect) on both human and animal health. Although probiotic effects are strain-specific traits, it is theoretically possible, using genetic engineering techniques, to design strains that can exert a variety of beneficial properties. During the two past decades, a large variety of therapeutic molecules has been successfully expressed in LAB, and although this field has been largely reviewed in recent years, approximately 20 new publications appear each year. Thus, the aim of this minireview is not to extensively assess the entire literature but to update progress made within the last 2 years regarding the use of the model LAB Lactococcus lactis and certain species of lactobacilli as live recombinant vectors for the development of new safe mucosal vaccines.


Subject(s)
Lactobacillus/genetics , Mucous Membrane/microbiology , Probiotics/therapeutic use , Animals , Genetic Vectors , Humans , Mucous Membrane/immunology , Probiotics/administration & dosage , Vaccines/administration & dosage , Vaccines/immunology
13.
J Biomed Biotechnol ; 2010: 317545, 2010.
Article in English | MEDLINE | ID: mdl-20414353

ABSTRACT

The delivery of bioactive molecules directly to damaged tissues represents a technological challenge. We propose here a new system based on virus-like particles (VLP) from rotavirus, with a marked tropism for the gut to deliver bio-active molecules to intestinal cells. For this, nonreplicative VLP nanoparticles were constructed using a baculovirus expression system and used to deliver an exogenous biomolecule, the green fluorescent protein (GFP), into either MA104 cells or intestinal cells from healthy and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-treated mice. Our results show that expression of rotavirus capsid proteins in baculovirus led to the auto assembly of VLP that display similar properties to rotavirus. In vitro experiments showed that VLP were able to enter into MA104 cells and deliver the reporter protein. Intragastric administration of fluorescent VLP in healthy and TNBS-treated mice resulted in the detection of GFP and viral proteins in intestinal samples. Our results demonstrate an efficient entry of non-replicative rotavirus VLP into the epithelial cell line MA104 and provide the first in vivo evidence of the potential of these nanoparticles as a promising safe candidate for drug delivery to intestinal cells.


Subject(s)
Drug Delivery Systems/methods , Nanoparticles/virology , Rotavirus/physiology , Virion/physiology , Virus Internalization , Analysis of Variance , Animals , Baculoviridae/genetics , Cell Line , Colitis/chemically induced , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Male , Mice , Mice, Inbred BALB C , Spodoptera/metabolism , Trinitrobenzenesulfonic Acid , Virion/genetics
14.
BMC Res Notes ; 2: 167, 2009 Aug 24.
Article in English | MEDLINE | ID: mdl-19703307

ABSTRACT

BACKGROUND: The expression of vaccine antigens in lactic acid bacteria (LAB) is a safe and cost-effective alternative to traditional expression systems. In this study, we investigated i) the expression of Human papillomavirus type 16 (HPV-16) L1 major capsid protein in the model LAB Lactococcus lactis and ii) the ability of the resulting recombinant strain to produce either capsomer-or virus-like particles (VLPs). RESULTS AND CONCLUSION: HPV-16 L1 gene was cloned into two vectors, pCYT and pSEC, designed for controlled intra- or extracellular heterologous expression in L. lactis, respectively. The capacity of L. lactis harboring either pCYT:L1 or pSEC:L1 plasmid to accumulate L1 in the cytoplasm and supernatant samples was confirmed by Western blot assays. Electron microscopy analysis suggests that, L1 protein produced by recombinant lactococci can self-assemble into structures morphologically similar to VLPs intracellularly. The presence of conformational epitopes on the L. lactis-derived VLPs was confirmed by ELISA using an anti-HPV16 L1 capsid antigen antibody. Our results support the feasibility of using recombinant food-grade LAB, such as L. lactis, for the production of L1-based VLPs and open the possibility for the development of a new safe mucosal vector for HPV-16 prophylactic vaccination.

15.
Int Arch Allergy Immunol ; 150(1): 25-31, 2009.
Article in English | MEDLINE | ID: mdl-19339799

ABSTRACT

BACKGROUND: In the last years, the use of probiotics such as lactic acid bacteria (LAB) has been proposed as an attractive alternative for the management of allergic diseases. A partial prevention from sensitization to bovine beta-lactoglobulin (BLG), one of the major cows' milk allergens, could be achieved in mice after intranasal administration with a recombinant LAB strain, Lactococcus lactis, producing BLG (LL-BLG). This study aimed to evaluate the effects of the LL-BLG strain in a therapeutic protocol. METHODS: Three groups of mice were first orally sensitized to cows' milk and then intranasally administered with either the LL-BLG strain, BLG protein alone or saline solution. Serum samples were collected to analyze BLG-specific IgE, IgG1 and IgG2a, and mice were further intranasally challenged with BLG to elicit a specific allergic reaction. RESULTS: Treatment with LL-BLG, but not with BLG alone, contributed to diminish IgG1 production in serum and bronchoalveolar lavage fluids. This was associated with decreased IL-4 production and enhanced IFN-gamma production by BLG-reactivated splenocytes, suggesting a switch from Th2- to Th1-immune response. Furthermore, we observed that administration of LL-BLG or LL locally reduced the allergic reaction induced after intranasal challenge, as evidenced by decreased release of IL-4 in bronchoalveolar lavage fluids. CONCLUSION: These preliminary results demonstrate the efficiency of the intranasal administration of LL-BLG for specific therapy against cows' milk-related allergy.


Subject(s)
Desensitization, Immunologic/methods , Lactococcus lactis/immunology , Lactoglobulins/administration & dosage , Milk Hypersensitivity/prevention & control , Administration, Intranasal , Animals , Cattle , Female , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Lactoglobulins/immunology , Mice , Mice, Inbred BALB C , Milk Hypersensitivity/immunology
16.
Vaccine ; 26(46): 5778-83, 2008 Oct 29.
Article in English | MEDLINE | ID: mdl-18790708

ABSTRACT

Chemokines have been described as essential mediators in leukocytes migration to inflammatory sites and to secondary lymphoid organs. Mig and IP-10 are two CXC chemokines that recruit mononuclear cells in vivo and inhibit angiogenesis. In addition to their chemotactic roles, Mig and IP-10 have also an important role in the adaptative immune response. In this study, we asked whether a food-grade bacterium, Lactococcus lactis, is able to produce a fusion protein comprising Mig and IP-10 (Mig::IP-10). The activity of the recombinant Mig::IP-10 produced by the genetically engineered L. lactis (LL-Mig::IP-10) was confirmed in a murine spleen cells chemotaxis assay. Moreover, the adjuvant properties of LL-Mig::IP-10 strain were evaluated in mice by the co-expression of a model antigen, the human papillomavirus type 16 E7 protein. Our data show that LL-Mig::IP-10 can produce a genetic fusion of Mig::IP-10 biologically active. This recombinant strain represents a potential candidate for the development of new strategies for mucosal vaccination.


Subject(s)
Adjuvants, Immunologic , Chemokines, CXC/biosynthesis , Chemokines, CXC/pharmacology , Immunity, Mucosal/immunology , Lactococcus lactis/metabolism , Administration, Intranasal , Animals , Antibody Formation/immunology , Chemotaxis, Leukocyte/drug effects , DNA, Bacterial/genetics , Escherichia coli/genetics , Female , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Lactococcus lactis/genetics , Mice , Mice, Inbred C57BL , Plasmids/genetics , Protein Engineering
17.
FEMS Microbiol Lett ; 280(2): 144-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18248432

ABSTRACT

IFN-gamma is a cytokine produced primarily by both T lymphocytes and natural killer cells and it is considered to be an attractive therapeutic molecule. In the present study, a DNA sequence encoding the mature murine IFN-gamma (muIFN-gamma) protein was cloned and expressed in the food-grade lactic acid bacterium Lactococcus lactis. The activity of recombinant muIFN-gamma produced by genetically engineered L. lactis was confirmed in an antiviral assay using MoV cells infected with Vesicular Stomatitis Virus. The data provide the first demonstration that a Gram-positive bacterium, L. lactis, is able to produce functional muIFN-gamma. This recombinant strain could lead to the development of a new, well-tolerated vector to deliver active muIFN-gamma at the mucosal level.


Subject(s)
Interferon-gamma/biosynthesis , Lactococcus lactis/metabolism , Animals , Cell Line , Genetic Vectors/metabolism , Interferon-gamma/genetics , Interferon-gamma/pharmacology , Lactococcus lactis/genetics , Macrophages/immunology , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Vesicular stomatitis Indiana virus/drug effects
18.
J Mol Microbiol Biotechnol ; 14(1-3): 80-9, 2008.
Article in English | MEDLINE | ID: mdl-17957114

ABSTRACT

The noninvasive and food-grade Gram-positive bacterium Lactococcus lactis is well adapted to deliver medical proteins to the mucosal immune system. In the last decade, the potential of live recombinant lactococci to deliver such proteins to the mucosal immune system has been investigated. This approach offers several advantages over the traditional systemic injection, such as easy administration and the ability to elicit both systemic and mucosal immune responses. This paper reviews the current research and advances made with recombinant L. lactis as live vector for the in situ delivery of biologically active interleukin-12, a potent pleiotropic cytokine with adjuvant properties when co-delivered with vaccinal antigens, at mucosal surfaces. Three well-illustrated examples demonstrate the high potential of interleukin-12-secreting lactococci strains for future prophylactic and therapeutic uses.


Subject(s)
Asthma/therapy , Genetic Vectors , Hypersensitivity/therapy , Interleukin-12/immunology , Lactococcus lactis/genetics , Papillomavirus Infections/prevention & control , Uterine Cervical Neoplasms/prevention & control , Vaccines, Synthetic , Administration, Intranasal , Animals , Asthma/prevention & control , Female , Human papillomavirus 16 , Hypersensitivity/etiology , Interleukin-12/genetics , Lactoglobulins/adverse effects , Lactoglobulins/immunology , Mice , Mucous Membrane/microbiology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
19.
Vaccine ; 25(36): 6581-8, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17675182

ABSTRACT

Mucosal immunity plays a major role in the prevention of infectious diseases. Genetically engineered lactic acid bacteria (LAB) have been tested in the last 10 years as safe mucosal delivery vectors. We previously showed that intranasal co-administration of recombinant lactococci displaying human papillomavirus type 16 (HPV-16) E7 antigen at its surface (LL-E7) and secreting biologically active interleukine-12 (LL-IL-12) has therapeutic effects on HPV-16-induced tumors in mice. In this work, to optimize the immunization protocol, a comparison between intragastric and intranasal routes of administration was performed and two different LAB strains (Lactococcus lactis and Lactobacillus plantarum) were tested as delivery vector. E7-specific systemic and mucosal responses as well as potent anti-tumor effects were higher after intranasal immunization with LL-E7 and LL-IL-12 strains than intragastric administration. Comparisons of the immune responses induced by intranasal administration of either LL-E7 or Lb. plantarum anchoring E7 antigen (LP-E7) revealed highest systemic responses with recombinant Lactobacillus. Furthermore, although only a modest mucosal immune response was observed with LP-E7, this strain was able to induce a significant regression of HPV-induced tumors in contrast to LL-E7. Taken together, our results demonstrate the advantage of intranasal over intragastric route of immunization to induce an antigen-specific immune response and suggest that intrinsic immunomodulatory properties of Lb. plantarum play an important role in the immunogenicity of the expressed antigen.


Subject(s)
Immunity, Mucosal/immunology , Immunization/methods , Lactobacillus plantarum/genetics , Lactococcus lactis/genetics , Recombinant Fusion Proteins/immunology , Administration, Intranasal , Animals , Drug Administration Routes , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors/genetics , Immunoblotting , Interleukin-12/genetics , Interleukin-12/immunology , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neoplasms/etiology , Neoplasms/immunology , Neoplasms/prevention & control , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins , Papillomavirus Infections/complications , Papillomavirus Infections/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics
20.
Clin Vaccine Immunol ; 14(3): 226-33, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17202306

ABSTRACT

The Th1/Th2 balance deregulation toward a Th2 immune response plays a central role in allergy. We previously demonstrated that administration of recombinant Lactococcus lactis strains expressing bovine beta-lactoglobulin (BLG), a major cow's milk allergen, partially prevents mice from sensitization. In the present study, we aimed to improve this preventive effect by coadministration of L. lactis BLG and a second recombinant L. lactis strain producing biologically active interleukin-12 (IL-12). This L. lactis strain producing IL-12 was previously used to enhance the Th1 immune response in a tumoral murine model (L. G. Bermúdez-Humarán et al., J. Immunol. 175:7297-7302, 2005). A comparison of the administration of either BLG alone or BLG in the presence of IL-12 was conducted. A BLG-specific primary Th1 immune response was observed only after intranasal coadministration of both L. lactis BLG and IL-12-producing L. lactis, as demonstrated by the induction of serum-specific immunoglobulin G2a (IgG2a) concomitant with gamma interferon secretion by splenocytes, confirming the adjuvanticity of IL-12-producing L. lactis. Immunized mice were further sensitized by intraperitoneal administration of purified BLG, and the allergic reaction was elicited by intranasal challenge with purified BLG. Mice pretreated with BLG in either the presence or the absence of IL-12 were rendered completely tolerant to further allergic sensitization and elicitation. Pretreatment with either L. lactis BLG or L. lactis BLG and IL-12-producing L. lactis induces specific anti-BLG IgG2a production in serum and bronchoalveolar lavage (BAL) fluid. Although specific serum IgE was not affected by these pretreatments, the levels of eosinophilia and IL-5 secretion in BAL fluid were significantly reduced after BLG challenge in the groups pretreated with L. lactis BLG and L. lactis BLG-IL-12-producing L. lactis, demonstrating a decreased allergic reaction. Our data demonstrate for the first time (i) the induction of a protective Th1 response by the association of L. lactis BLG and IL-12-producing L. lactis which inhibits the elicitation of the allergic reaction to BLG in mice and (ii) the efficiency of intranasal administration of BLG for the induction of tolerance.


Subject(s)
Interleukin-12/biosynthesis , Lactococcus lactis/immunology , Lactoglobulins/immunology , Milk Hypersensitivity/prevention & control , Vaccines, Synthetic/immunology , Administration, Intranasal , Animals , Cattle , Female , Immunoglobulin G/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...