Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 12(9)2023 04 26.
Article in English | MEDLINE | ID: mdl-37174660

ABSTRACT

The gut microbiota is now considered as a key player in the development of metabolic dysfunction. Therefore, targeting gut microbiota dysbiosis has emerged as a new therapeutic strategy, notably through the use of live gut microbiota-derived biotherapeutics. We previously highlighted the anti-inflammatory abilities of two Parabacteroides distasonis strains. We herein evaluate their potential anti-obesity abilities and show that the two strains induced the secretion of the incretin glucagon-like peptide 1 in vitro and limited weight gain and adiposity in obese mice. These beneficial effects are associated with reduced inflammation in adipose tissue and the improvement of lipid and bile acid metabolism markers. P. distasonis supplementation also modified the Actinomycetota, Bacillota and Bacteroidota taxa of the mice gut microbiota. These results provide better insight into the capacity of P. distasonis to positively influence host metabolism and to be used as novel source of live biotherapeutics in the treatment and prevention of metabolic-related diseases.


Subject(s)
Gastrointestinal Microbiome , Obesity , Animals , Mice , Obesity/therapy , Obesity/metabolism , Bacteroidetes , Adipose Tissue/metabolism
2.
Sci Rep ; 12(1): 17591, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266398

ABSTRACT

Live biotherapeutic products constitute an emerging therapeutic approach to prevent or treat inflammatory bowel diseases. Lactobacillus acidophilus is a constituent of the human microbiota with probiotic potential, that is illustrated by improvement of intestinal inflammation and antimicrobial activity against several pathogens. In this study, we evaluated the immunomodulatory properties of the L. acidophilus strain BIO5768 at steady state and upon acute inflammation. Supplementation of naïve mice with BIO5768 heightened the transcript level of some IL-17 target genes encoding for protein with microbicidal activity independently of NOD2 signaling. Of these, the BIO5768-induced expression of Angiogenin-4 was blunted in monocolonized mice that are deficient for the receptor of IL-17 (but not for NOD2). Interestingly, priming of bone marrow derived dendritic cells by BIO5768 enhanced their ability to support the secretion of IL-17 by CD4+ T cells. Equally of importance, the production of IL-22 by type 3 innate lymphoid cells is concomitantly heightened in response to BIO5768. When administered alone or in combination with Bifidobacterium animalis spp. lactis BIO5764 and Limosilactobacillus reuteri, BIO5768 was able to alleviate at least partially intestinal inflammation induced by Citrobacter rodentium infection. Furthermore, BIO5768 was also able to improve colitis induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS). In conclusion, we identify a new potential probiotic strain for the management of inflammatory bowel diseases, and provide some insights into its IL-17-dependent and independent mode of action.


Subject(s)
Colitis , Immunity, Innate , Inflammatory Bowel Diseases , Lactobacillus acidophilus , Probiotics , Animals , Mice , Bifidobacterium animalis , Colitis/chemically induced , Colitis/therapy , Colitis/microbiology , Enterobacteriaceae Infections/therapy , Inflammation , Inflammatory Bowel Diseases/therapy , Interleukin-17 , Lymphocytes , Probiotics/pharmacology , Probiotics/therapeutic use , Trinitrobenzenesulfonic Acid/adverse effects
3.
Microorganisms ; 9(3)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803291

ABSTRACT

The role of the gut microbiota in health and disease is well recognized and the microbiota dysbiosis observed in many chronic diseases became a new therapeutic target. The challenge is to get a better insight into the functionality of commensal bacteria and to use this knowledge to select live biotherapeutics as new preventive or therapeutic products. In this study, we set up a screening approach to evaluate the functional capacities of a set of 21 strains isolated from the gut microbiota of neonates and adults. For this purpose, we selected key biological processes involved in the microbiome-host symbiosis and known to impact the host physiology i.e., the production of short-chain fatty acids and the ability to strengthen an epithelial barrier (Caco-2), to induce the release of the anti-inflammatory IL-10 cytokine after co-culture with human immune cells (PBMC) or to increase GLP-1 production from STC-1 endocrine cell line. This strategy highlighted fifteen strains exhibiting beneficial activities among which seven strains combined several of them. Interestingly, this work revealed for the first time a high prevalence of potential health-promoting functions among intestinal commensal strains and identified several appealing novel candidates for the management of chronic diseases, notably obesity and inflammatory bowel diseases.

4.
Nutrients ; 13(3)2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33668212

ABSTRACT

Since alterations of the gut microbiota have been shown to play a major role in obesity, probiotics have attracted attention. Our aim was to identify probiotic candidates for the management of obesity using a combination of in vitro and in vivo approaches. We evaluated in vitro the ability of 23 strains to limit lipid accumulation in adipocytes and to enhance the secretion of satiety-promoting gut peptide in enteroendocrine cells. Following the in vitro screening, selected strains were further investigated in vivo, single, or as mixtures, using a murine model of diet-induced obesity. Strain Bifidobacterium longum PI10 administrated alone and the mixture of B. animalis subsp. lactis LA804 and Lactobacillus gasseri LA806 limited body weight gain and reduced obesity-associated metabolic dysfunction and inflammation. These protective effects were associated with changes in the hypothalamic gene expression of leptin and leptin receptor as well as with changes in the composition of gut microbiota and the profile of bile acids. This study provides crucial clues to identify new potential probiotics as effective therapeutic approaches in the management of obesity, while also providing some insights into their mechanisms of action.


Subject(s)
Adipocytes/microbiology , Enteroendocrine Cells/microbiology , Gastrointestinal Microbiome/physiology , Obesity/microbiology , Probiotics/pharmacology , Animals , Bile Acids and Salts/metabolism , Diet/adverse effects , Disease Models, Animal , Gastrointestinal Hormones/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Mice , Obesity/etiology , Obesity Management/methods , Receptors, Leptin/metabolism , Weight Gain/physiology
5.
Probiotics Antimicrob Proteins ; 13(4): 970-981, 2021 08.
Article in English | MEDLINE | ID: mdl-33649898

ABSTRACT

Infectious mastitis is the major cause of early weaning, depriving infants of breastfeeding benefits. It is associated with an inflammatory condition of the breast and lowered resistance to infection. Drug administration during lactation often being contra-indicated, it is therefore important to consider safe therapeutic alternatives to antibiotic and anti-inflammatory therapies, such as probiotics. In this study, we investigated in vitro the probiotic potential of thirteen Lacticaseibacillus (formerly Lactobacillus) rhamnosus strains isolated from the gut microbiota of breastfed healthy infants. Strains were assessed for their ß-hemolytic activity, their resistance to antibiotics, and their antimicrobial activities against strains of Staphylococcus and Streptococcus, most often involved in women mastitis. Their immunomodulating abilities were also studied using in vitro stimulation of human immune cells. None of the strains exhibited ß-hemolytic activity, and all of them were sensitive to ampicillin, penicillin, tetracycline, rifampicin, erythromycin, chloramphenicol, and imipenem but showed resistance to ceftazidime, trimethoprim/sulfamethoxazole, vancomycin, and cefotaxime, reported to be chromosomally encoded and not inducible or transferable. Four L. rhamnosus strains were selected for their large anti-staphylococcal spectrum: L. rhamnosus VR1-5 and L. rhamnosus VR3-1 inhibiting S. aureus, S. epidermis, and S. warneri and L. rhamnosus CB9-2 and L. rhamnosus CB10-5 exerting antagonistic effect against S. aureus and S. epidermis strains. Antimicrobial compounds released in cell-free supernatant showed proteinaceous nature and were thermoresistant. The immune modulatory analysis of the L. rhamnosus strains revealed two strains with significant anti-inflammatory potential, highlighted by strong induction of IL-10 and a weak pro-Th1 cytokine secretion (IL-12 and IFN-γ). L. rhamnosus CB9-2 combined a large anti-staphylococcal activity spectrum and a promising anti-inflammatory profile. This strain, used individually or in a mixture, can be considered as a probiotic candidate for the management of infectious mastitis during lactation.


Subject(s)
Anti-Inflammatory Agents , Gastrointestinal Microbiome , Lacticaseibacillus rhamnosus , Mastitis , Probiotics , Female , Humans , Infant , Mastitis/therapy , Staphylococcus aureus , Staphylococcus epidermidis
6.
Cells ; 9(9)2020 09 16.
Article in English | MEDLINE | ID: mdl-32947881

ABSTRACT

Alterations in the gut microbiota composition and diversity seem to play a role in the development of chronic diseases, including inflammatory bowel disease (IBD), leading to gut barrier disruption and induction of proinflammatory immune responses. This opens the door for the use of novel health-promoting bacteria. We selected five Parabacteroides distasonis strains isolated from human adult and neonates gut microbiota. We evaluated in vitro their immunomodulation capacities and their ability to reinforce the gut barrier and characterized in vivo their protective effects in an acute murine model of colitis. The in vitro beneficial activities were highly strain dependent: two strains exhibited a potent anti-inflammatory potential and restored the gut barrier while a third strain reinstated the epithelial barrier. While their survival to in vitro gastric conditions was variable, the levels of P. distasonis DNA were higher in the stools of bacteria-treated animals. The strains that were positively scored in vitro displayed a strong ability to rescue mice from colitis. We further showed that two strains primed dendritic cells to induce regulatory T lymphocytes from naïve CD4+ T cells. This study provides better insights on the functionality of commensal bacteria and crucial clues to design live biotherapeutics able to target inflammatory chronic diseases such as IBD.


Subject(s)
Bacteroidetes/genetics , Bacteroidetes/immunology , Colitis/chemically induced , Colitis/microbiology , Gastrointestinal Microbiome/immunology , Trinitrobenzenesulfonic Acid/adverse effects , Adult , Animals , Bacteroidetes/isolation & purification , Caco-2 Cells , Colitis/immunology , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Disease Models, Animal , Feces/microbiology , Female , Humans , Infant, Newborn , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology
7.
Sci Rep ; 10(1): 5345, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210304

ABSTRACT

Crohn's disease is linked to a decreased diversity in gut microbiota composition as a potential consequence of an impaired anti-microbial response and an altered polarization of T helper cells. Here, we evaluated the immunomodulatory properties of two potential probiotic strains, namely a Bifidobacterium animalis spp. lactis Bl 5764 and a Lactobacillus reuteri Lr 5454 strains. Both strains improved colitis triggered by either 2,4,6-trinitrobenzenesulfonic acid (TNBS) or Citrobacter rodentium infection in mice. Training of dendritic cells (DC) with Lr 5454 efficiently triggered IL-22 secretion and regulatory T cells induction in vitro, while IL-17A production by CD4+ T lymphocytes was stronger when cultured with DCs that were primed with Bl 5764. This strain was sufficient for significantly inducing expression of antimicrobial peptides in vivo through the Crohn's disease predisposing gene encoding for the nucleotide-binding oligomerization domain, containing protein 2 (NOD2). In contrast, NOD2 was dispensable for the impact on antimicrobial peptide expression in mice that were monocolonized with Lr 5454. In conclusion, our work highlights a differential mode of action of two potential probiotic strains that protect mice against colitis, providing the rational for a personalized supportive preventive therapy by probiotics for individuals that are genetically predisposed to Crohn's disease.


Subject(s)
Bifidobacterium animalis , Colitis/microbiology , Colitis/therapy , Dendritic Cells/physiology , Limosilactobacillus reuteri , Probiotics/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Citrobacter rodentium/pathogenicity , Colitis/chemically induced , Colitis/pathology , Disease Models, Animal , Enterobacteriaceae Infections/microbiology , Female , Gastrointestinal Microbiome , Germ-Free Life , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis-Associated Proteins/genetics , T-Lymphocytes, Helper-Inducer/physiology , T-Lymphocytes, Regulatory/physiology , Trinitrobenzenesulfonic Acid/toxicity
8.
Cell Physiol Biochem ; 53(5): 774-793, 2019.
Article in English | MEDLINE | ID: mdl-31647207

ABSTRACT

BACKGROUND/AIMS: Deregulation of the complex interaction among host genetics, gut microbiota and environmental factors on one hand and aberrant immune responses on the other hand, are known to be associated with the development of inflammatory bowel disease. Recent studies provided strong evidence that autophagy plays a key role in the etiology of Crohn's disease (CD). Probiotics may exhibit many therapeutic properties, including anti-inflammatory abilities. While successful results have been obtained in ulcerative colitis patients, probiotics remain inefficient in CD for unknown reason. It remains therefore important to better understand their molecular mechanisms of action. METHODS: The activation of autophagy was examined by stimulating bone marrow-derived dendritic cells by the bacteria, followed by confocal microscopy and western blot analysis. The impact of blocking in vitro autophagy was performed in peripheral blood mononuclear cells using 3-methyl adenine or bafilomycin followed by cytokine secretion measurement by ELISA. The role of autophagy in the anti-inflammatory capacities of the bacterial strains was evaluated in vivo using an acute trinitrobenzene sulfonic acid-induced murine model of colitis. The impact of BMDC was evaluated by adoptive transfer, notably using bone marrow cells derived from autophagy-related 16-like 1-deficient mice. RESULTS: We showed that selected lactobacilli and bifidobacteria are able to induce autophagy activation in BMDCs. Blocking in vitro autophagy abolished the capacity of the strains to induce the release of the anti-inflammatory cytokine interleukin-10, while it exacerbated the secretion of the pro-inflammatory cytokine interleukin-1ß. We confirmed in the TNBS-induced mouse model of colitis that autophagy is involved in the protective capacity of these selected strains, and showed that dendritic cells are involved in this process. CONCLUSION: We propose autophagy as a novel mechanism involved in the regulatory capacities of probiotics.


Subject(s)
Autophagy , Bifidobacterium/physiology , Lactobacillus/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy-Related Proteins , Bone Marrow Cells/cytology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chemokines/genetics , Chemokines/metabolism , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Female , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Macrolides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout
9.
Front Immunol ; 10: 1830, 2019.
Article in English | MEDLINE | ID: mdl-31428104

ABSTRACT

Yersinioses caused by Yersinia pestis, Yersinia pseudotuberculosis, and Yersinia enterocolitica are significant concerns in human and veterinary health. The link between virulence and the potent LcrV antigen has prompted the latter's selection as a major component of anti-Yersinia vaccines. Here, we report that (i) the group of Yersinia species encompassing Y. pestis and Y. pseudotuberculosis produces at least five different clades of LcrV and (ii) vaccination of mice with an LcrV-secreting Lactococcus lactis only protected against Yersinia strains producing the same LcrV clade as that of used for vaccination. By vaccinating with engineered LcrVs and challenging mice with strains producing either type of LcrV or a LcrV mutated for regions of interest, we highlight key polymorphic residues responsible for the absence of cross-protection. Our results show that an anti-LcrV-based vaccine should contain multiple LcrV clades if protection against the widest possible array of Yersinia strains is sought.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Lactococcus lactis/immunology , Pore Forming Cytotoxic Proteins/immunology , Yersinia pestis/immunology , Yersinia pseudotuberculosis/immunology , Animals , Antibodies, Bacterial/immunology , Cross Protection/immunology , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Vaccination/methods , Virulence/immunology , Yersinia Infections/immunology
10.
Sci Rep ; 7: 43211, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28233848

ABSTRACT

Gut microbiota dysbiosis plays a central role in the development and perpetuation of chronic inflammation in inflammatory bowel disease (IBD) and therefore is key target for interventions with high quality and functional probiotics. The local production of stable probiotic formulations at limited cost is considered an advantage as it reduces transportation cost and time, thereby increasing the effective period at the consumer side. In the present study, we compared the anti-inflammatory capacities of the Bifidobacterium animalis subsp. lactis (B. lactis) INL1, a probiotic strain isolated in Argentina from human breast milk, with the commercial strain B. animalis subsp. lactis BB12. The impact of spray-drying, a low-cost alternative of bacterial dehydration, on the functionality of both bifidobacteria was also investigated. We showed for both bacteria that the spray-drying process did not impact on bacterial survival nor on their protective capacities against acute and chronic colitis in mice, opening future perspectives for the use of strain INL1 in populations with IBD.


Subject(s)
Bifidobacterium animalis/isolation & purification , Colitis/prevention & control , Desiccation/methods , Milk, Human/microbiology , Probiotics/administration & dosage , Probiotics/isolation & purification , Technology, Pharmaceutical/methods , Animals , Argentina , Bacteriological Techniques/methods , Bifidobacterium animalis/physiology , Disease Models, Animal , Humans , Mice , Microbial Viability
11.
Vaccine ; 34(47): 5762-5767, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27742220

ABSTRACT

BACKGROUND: Pseudotuberculosis is an infection caused by the bacterial enteropathogen Yersinia pseudotuberculosis and is considered to be a significant problem in veterinary medicine. We previously found that intranasal administration of a recombinant Lactococcus lactis strain that secretes the low-calcium response V (LcrV) antigen from Y. pseudotuberculosis (Ll-LcrV) confers protection against a lethal Y. pseudotuberculosis infection. Here, we aimed at characterizing the immunological basis of this LcrV-elicited protective response and at determining the duration of vaccine-induced immunity. METHODS: Splenocytes from BALB/c mice intranasally immunized with Ll-LcrV or Ll as control were immunostained then analyzed by flow cytometry. Protection against a lethal intravenous injection of Y. pseudotuberculosis was also determined (i) in immunized BALB/c mice depleted or not of CD4+, CD8+ or CD25+ cells and (ii) in naïve BALB/c mice receiving serum from immunized mice by counting the number of bacteria in liver and spleen. Lastly, survival rate of immunized BALB/c mice following a lethal intravenous injection of Y. pseudotuberculosis was followed up to 9-months. RESULTS: We found that T and B lymphocytes but not non-conventional lymphoid cells were affected by Ll-LcrV immunization. We also observed that depletion of CD4+ and CD25+ but not CD8+ cells in immunized mice eradicated protection against a lethal systemic Y. pseudotuberculosis infection, suggesting that activated CD4+ T lymphocytes are required for vaccine-induced protection. Adoptive transfer of LcrV-specific antibodies from Ll-LcrV-immunized animals significantly reduced the bacterial counts in the liver compared to non-vaccinated mice. Lastly, the protective immunity conferred by Ll-LcrV decreased slightly over time; nevertheless almost 60% of the mice survived a lethal bacterial challenge at 9months post-vaccination. CONCLUSION: Mucosal vaccination of mice with Ll-LcrV induced cell- and antibody-mediated protective immunity against Y. pseudotuberculosis infection in the mouse and the protection is long-lasting.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Immunity, Active/immunology , Lactococcus lactis/immunology , Pore Forming Cytotoxic Proteins/immunology , Yersinia pseudotuberculosis Infections/prevention & control , Yersinia pseudotuberculosis/immunology , Administration, Intranasal , Animals , Antigens, Bacterial/genetics , Bacterial Load , CD4 Antigens/immunology , CD8 Antigens/immunology , Female , Humans , Injections, Intravenous , Interleukin-2 Receptor alpha Subunit/immunology , Lactococcus lactis/genetics , Mice , Mice, Inbred BALB C , Pore Forming Cytotoxic Proteins/genetics , Primary Cell Culture , Spleen/immunology , Spleen/microbiology , Statistics, Nonparametric , Time Factors , Vaccination , Vaccines, Synthetic/immunology , Yersinia pseudotuberculosis/genetics
12.
Appl Environ Microbiol ; 81(16): 5344-9, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26025906

ABSTRACT

Lactic acid bacteria are found in the gastrointestinal tract of mammals and have received tremendous attention due to their health-promoting properties. We report the development of two dual-color luciferase-producing Lactobacillus (Lb.) plantarum and Lactococcus (Lc.) lactis strains for noninvasive simultaneous tracking in the mouse gastrointestinal tract. We previously described the functional expression of the red luciferase mutant (CBRluc) from Pyrophorus plagiophthalamus in Lb. plantarum NCIMB8826 and Lc. lactis MG1363 (C. Daniel, S. Poiret, V. Dennin, D. Boutillier, and B. Pot, Appl Environ Microbiol 79:1086-1094, 2013, http://dx.doi.org/10.1128/AEM.03221-12). In this study, we determined that CBRluc is a better-performing luciferase for in vivo localization of both lactic acid bacteria after oral administration than the green click beetle luciferase mutant construct developed in this study. We further established the possibility to simultaneously detect red- and green-emitting lactic acid bacteria by dual-wavelength bioluminescence imaging in combination with spectral unmixing. The difference in spectra of light emission by the red and green click beetle luciferase mutants and dual bioluminescence detection allowed in vitro and in vivo quantification of the red and green emitted signals; thus, it allowed us to monitor the dynamics and fate of the two bacterial populations simultaneously. Persistence and viability of both strains simultaneously administered to mice in different ratios was studied in vivo in anesthetized mice and ex vivo in mouse feces. The application of dual-luciferase-labeled bacteria has considerable potential to simultaneously study the interactions and potential competitions of different targeted bacteria and their hosts.


Subject(s)
Color , Gastrointestinal Tract/microbiology , Lactobacillus plantarum/physiology , Lactococcus lactis/physiology , Luciferases/analysis , Luminescent Measurements/methods , Animals , Genes, Reporter , Lactobacillus plantarum/enzymology , Lactobacillus plantarum/genetics , Lactobacillus plantarum/metabolism , Lactococcus lactis/enzymology , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Luciferases/genetics , Mice , Microbial Viability , Staining and Labeling
13.
Appl Environ Microbiol ; 79(4): 1086-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23204409

ABSTRACT

Lactic acid bacteria, especially lactobacilli, are common inhabitants of the gastrointestinal tract of mammals, for which they have received considerable attention due to their putative health-promoting properties. In this study, we describe the development and application of luciferase-expressing Lactobacillus plantarum and Lactococcus lactis strains for noninvasive in vivo monitoring in the digestive tract of mice. We report for the first time the functional in vitro expression in Lactobacillus plantarum NCIMB8826 and in Lactococcus lactis MG1363 of the click beetle luciferase (CBluc), as well as Gaussia and bacterial luciferases, using a combination of vectors, promoters, and codon-optimized genes. We demonstrate that a CBluc construction is the best-performing luciferase system for the noninvasive in vivo detection of lactic acid bacteria after oral administration. The persistence and viability of both strains was studied by bioluminescence imaging in anesthetized mice and in mouse feces. In vivo bioluminescence imaging confirmed that after a single or multiple oral administrations, L. lactis has shorter survival times in the mouse gastrointestinal tract than L. plantarum, and it also revealed the precise gut compartments where both strains persisted. The application of luciferase-labeled bacteria has significant potential to allow the in vivo and ex vivo study of the interactions of lactic acid bacteria with their mammalian host.


Subject(s)
Gastrointestinal Tract/microbiology , Lactobacillus plantarum/growth & development , Lactococcus lactis/growth & development , Whole Body Imaging , Animals , Genes, Reporter , Luciferases/analysis , Luciferases/genetics , Luminescent Measurements , Mice , Microbial Viability , Staining and Labeling/methods , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...