Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Hum Vaccin Immunother ; 20(1): 2375825, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39101772

ABSTRACT

Multiple research studies have demonstrated the efficacy of lactic acid bacteria in boosting both innate and adaptive immune responses. We have created a Lactococcus lactis variant that produces a modified combination protein with Fms-like tyrosine kinase 3 ligand and co-stimulator O × 40 ligand, known as HuFOLactis. The genetically modified variant was purposely created to activate T cells, NK cells, and DC cells in a laboratory setting. Furthermore, we explored the possibility of using the tumor-penetrating peptide iRGD to deliver HuFOLactis-activated immune cells to hard-to-reach tumor areas. Following brief stimulation with HuFOLactis, immune cell phenotypes and functions were assessed using flow cytometry. Confocal microscopy was employed to demonstrate the infiltrative and cytotoxic capabilities of iRGD-modified HuFOLactis-activated immune cells within tumor spheroids. The efficacy of iRGD modified HuFOLactis-activated immune cells against tumors was assessed in xenograft mouse models. HuFOLactis treatment resulted in notable immune cell activation, demonstrated by elevated levels of CD25, CD69, and CD137. Additionally, these activated immune cells showed heightened cytokine production and enhanced cytotoxicity against MKN45 cell lines. Incorporation of the iRGD modification facilitated the infiltration of HuFOLactis-activated immune cells into multicellular spheroids (MCSs). Additionally, immune cells activated by HuFOLactis and modified with iRGD, in combination with anti-PD-1 treatment, effectively halted tumor growth and prolonged survival in a mouse model of gastric cancer.


Subject(s)
Lactococcus lactis , Animals , Mice , Lactococcus lactis/genetics , Oligopeptides/pharmacology , Humans , Cell Line, Tumor , Female , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays , Dendritic Cells/immunology , Dendritic Cells/drug effects
2.
ACS Biomater Sci Eng ; 10(8): 5210-5225, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39087888

ABSTRACT

Lactococcus lactis (L. lactis), the first genetically modified Generally Recognized As Safe (GRAS) category Lactic Acid producing Bacteria (LAB), is best known for its generalized health-promoting benefits and ability to express heterologous proteins. However, achieving the optimal probiotic effects requires a selective approach that would allow us to study in vivo microbial biodistribution, fate, and immunological consequences. Although the chemical conjugation of fluorophores and chromophores represent the standard procedure to tag microbial cells for various downstream applications, it requires a high-throughput synthesis scheme, which is often time-consuming and expensive. On the contrary, the genetic manipulation of LAB vector, either chromosomally or extra-chromosomally, to express bioluminescent or fluorescent reporter proteins has greatly enhanced our ability to monitor bacterial transit through a complex gut environment. However, with faster passage and quick washing out from the gut due to rhythmic contractions of the digestive tract, real-time tracking of LAB vectors, particularly non-commensal ones, remains problematic. To get a deeper insight into the biodistribution of non-commensal probiotic bacteria in vivo, we bioengineered L. lactis to express fluorescence reporter proteins, mCherry (bright red monomeric fluorescent protein) and mEGFP (monomeric enhanced green fluorescent protein), followed by microencapsulation with a mucoadhesive and biodegradable polymer, chitosan. We show that coating of recombinant Lactococcus lactis (rL. lactis) with chitosan polymer, cross-linked with tripolyphosphate (TPP), retains their ability to express the reporter proteins stably without altering the specificity and sensitivity of fluorescence detection in vitro and in vivo. Further, we provide evidence of enhanced intragastric stability by chitosan-TPP (CS) coating of rL. lactis cells, allowing us to study the spatiotemporal distribution for an extended time in the gut of two unrelated hosts, avian and murine. The present scheme involving genetic modification and chitosan encapsulation of non-commensal LAB vector demonstrates great promise as a non-invasive and intensive tool for active live tracking of gut microbes.


Subject(s)
Lactococcus lactis , Luminescent Proteins , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Animals , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Genetic Vectors , Genes, Reporter , Mice , Probiotics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Red Fluorescent Protein
3.
Methods Mol Biol ; 2839: 99-110, 2024.
Article in English | MEDLINE | ID: mdl-39008250

ABSTRACT

Metal ion homeostasis in mitochondria is essential to maintaining proper cellular physiology. However, the ability of metals to bind off target or form complexes with multiple metabolites presents major challenges to understanding the mechanisms that govern this homeostasis. Adding further to the complexity, some of the major mitochondrial transporters have shown substrate promiscuity. In many cases, mitochondrial metals are found in the matrix compartment that is surrounded by the impermeable inner membrane. Four major classes of transporters facilitate the movement of solute across the inner membrane. These are mitochondrial carrier family, ATP-binding cassette transporters, mitochondrial pyruvate carriers, and sideroflexins. For iron, the matrix is the site of iron-sulfur clusters and heme synthesis and therefore transport must occur in a coordinated fashion with the cellular needs for these critical cofactors. Iron could be transported in numerous forms as it has been shown to form complexes with abundant metabolites such as citrate, nucleotides, or glutathione. Here, we describe assays to study iron (or any metal) transport by mitochondrial carrier family proteins expressed in Lactococcus lactis using a nisin-controlled expression system.


Subject(s)
Iron , Lactococcus lactis , Lactococcus lactis/metabolism , Lactococcus lactis/genetics , Iron/metabolism , Metals/metabolism , Mitochondria/metabolism , Biological Transport , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Nisin/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics
4.
Article in English | MEDLINE | ID: mdl-39013608

ABSTRACT

The industrial amino acid production workhorse, Corynebacterium glutamicum naturally produces low levels of 2,3,5,6-tetramethylpyrazine (TMP), a valuable flavor, fragrance, and commodity chemical. Here, we demonstrate TMP production (∼0.8 g L-1) in C. glutamicum type strain ATCC13032 via overexpression of acetolactate synthase and/or α-acetolactate decarboxylase from Lactococcus lactis in CGXII minimal medium supplemented with 40 g L-1 glucose. This engineered strain also demonstrated growth and TMP production when the minimal medium was supplemented with up to 40% (v v-1) hydrolysates derived from ionic liquid-pretreated sorghum biomass. A key objective was to take the fully engineered strain developed in this study and interrogate medium parameters that influence the production of TMP, a critical post-strain engineering optimization. Design of experiments in a high-throughput plate format identified glucose, urea, and their ratio as significant components affecting TMP production. These two components were further optimized using response surface methodology. In the optimized CGXII medium, the engineered strain could produce up to 3.56 g L-1 TMP (4-fold enhancement in titers and 2-fold enhancement in yield, mol mol-1) from 80 g L-1 glucose and 11.9 g L-1 urea in shake flask batch cultivation. ONE-SENTENCE SUMMARY: Corynebacterium glutamicum was metabolically engineered to produce 2,3,5,6-tetramethylpyrazine followed by a design of experiments approach to optimize medium components for high-titer production.


Subject(s)
Corynebacterium glutamicum , Culture Media , Glucose , Metabolic Engineering , Pyrazines , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Pyrazines/metabolism , Metabolic Engineering/methods , Culture Media/chemistry , Glucose/metabolism , Acetolactate Synthase/genetics , Acetolactate Synthase/metabolism , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Lactococcus lactis/enzymology , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Urea/metabolism
5.
Arch Microbiol ; 206(7): 336, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38954047

ABSTRACT

Wild-type Lactococcus lactis strain LAC460 secretes prophage-encoded bacteriocin-like lysin LysL, which kills some Lactococcus strains, but has no lytic effect on the producer. LysL carries two N-terminal enzymatic active domains (EAD), and an unknown C-terminus without homology to known domains. This study aimed to determine whether the C-terminus of LysL carries a cell wall binding domain (CBD) for target specificity of LysL. The C-terminal putative CBD region of LysL was fused with His-tagged green fluorescent protein (HGFPuv). The HGFPuv_CBDlysL gene fusion was ligated into the pASG-IBA4 vector, and introduced into Escherichia coli. The fusion protein was produced and purified with affinity chromatography. To analyse the binding of HGFPuv_CBDLysL to Lactococcus cells, the protein was mixed with LysL-sensitive and LysL-resistant strains, including the LysL-producer LAC460, and the fluorescence of the cells was analysed. As seen in fluorescence microscope, HGFPuv_CBDLysL decorated the cell surface of LysL-sensitive L. cremoris MG1614 with green fluorescence, whereas the resistant L. lactis strains LM0230 and LAC460 remained unfluorescent. The fluorescence plate reader confirmed the microscopy results detecting fluorescence only from four tested LysL-sensitive strains but not from 11 tested LysL-resistant strains. Specific binding of HGFPuv_CBDLysL onto the LysL-sensitive cells but not onto the LysL-resistant strains indicates that the C-terminus of LysL contains specific CBD. In conclusion, this report presents experimental evidence of the presence of a CBD in a lactococcal phage lysin. Moreover, the inability of HGFPuv_CBDLysL to bind to the LysL producer LAC460 may partly explain the host's resistance to its own prophage lysin.


Subject(s)
Bacteriocins , Cell Wall , Lactococcus lactis , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Cell Wall/metabolism , Bacteriocins/metabolism , Bacteriocins/genetics , Bacteriocins/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Protein Domains , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Protein Binding
6.
BMC Microbiol ; 24(1): 282, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080520

ABSTRACT

BACKGROUND: Gut microbiota have several advantages in influencing the host nutrition, metabolism, immunity and growth. However, the understanding of the gut microbiota in key edible wood-boring beetle larvae remain largely undefined. In the present study, the characteristics of the gut microbiota of two edible wood-boring species (Titocerus jaspideus and Passalus punctiger) from two indigenous forested areas were investigated. RESULTS: Over 50% of Amplicon Sequence Variants (ASVs) constituted of Firmicutes in T. jaspideus. The dominant phyla in both beetle species were Bacteroidota (4.20-19.79%) and Proteobacteria (15.10-23.90%). Lactococcus lactis was the most abundant and core prokaryote in the guts of T. jaspideus. The fungi identified in the gut of both insects belong to the phylum Obazoa (66%) and Ascomycota (> 15%). Scheffersomyeces sp. was the core eukaryote recorded. The diversity of gut microbiota in both insect species did not vary significantly. Most of the prokaryotic genes expressed were predominantly associated with biosynthesis and metabolism. CONCLUSION: Our findings demonstrated that Lactococcus lactis and Scheffersomyeces are core gut microbes of wood boring beetle larvae with desirable probiotic properties and promising use in food product fermentation for improved growth performance, gut barrier health, intestinal flora balance and immune protection for human and animals. Further studies to highlight the latest medical-based applications of L. lactis as live-delivery vector for the administration of therapeutics against both communicable and non-communicable diseases are warranted.


Subject(s)
Coleoptera , Gastrointestinal Microbiome , Lactococcus lactis , Larva , Symbiosis , Animals , Lactococcus lactis/genetics , Coleoptera/microbiology , Larva/microbiology , Wood/microbiology , RNA, Ribosomal, 16S/genetics , Phylogeny , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification
8.
PLoS Genet ; 20(7): e1011340, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38950059

ABSTRACT

Lactococcus lactis is a lactic acid bacterium of major importance for food fermentation and biotechnological applications. The ability to manipulate its genome quickly and easily through competence for DNA transformation would accelerate its general use as a platform for a variety of applications. Natural transformation in this species requires the activation of the master regulator ComX. However, the growth conditions that lead to spontaneous transformation, as well as the regulators that control ComX production, are unknown. Here, we identified the carbon source, nitrogen supply, and pH as key factors controlling competence development in this species. Notably, we showed that these conditions are sensed by three global regulators (i.e., CcpA, CodY, and CovR), which repress comX transcription directly. Furthermore, our systematic inactivation of known signaling systems suggests that classical pheromone-sensing regulators are not involved. Finally, we revealed that the ComX-degrading MecA-ClpCP machinery plays a predominant role based on the identification of a single amino-acid substitution in the adaptor protein MecA of a highly transformable strain. Contrasting with closely-related streptococci, the master competence regulator in L. lactis is regulated both proximally by general sensors and distantly by the Clp degradation machinery. This study not only highlights the diversity of regulatory networks for competence control in Gram-positive bacteria, but it also paves the way for the use of natural transformation as a tool to manipulate this biotechnologically important bacterium.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Lactococcus lactis , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Transformation, Bacterial/genetics , Lactococcus/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , DNA Transformation Competence/genetics
9.
J Appl Microbiol ; 135(7)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38955370

ABSTRACT

AIMS: This study aims to evaluate the storage stability of the freeze-dried recombinant Lactococcus lactis NZ3900-fermented milk powder expressing K-ras (Kristen rat sarcoma viral oncogene homolog) mimotopes targeting colorectal cancer in vacuum packaging. METHODS AND RESULTS: The freeze-dried L. lactis-fermented milk powder stored in 4-ply retortable polypropylene (RCPP)-polyamide (PA)-aluminium (AL)-polyethylene terephthalate (PET) and aluminium polyethylene (ALPE) was evaluated throughout 49 days of accelerated storage (38°C and 90% relative humidity). The fermented milk powder stored in 4-ply packaging remained above 6 log10 CFU g-1 viability, displayed lower moisture content (6.1%), higher flowability (43° angle of repose), water solubility (62%), and survivability of L. lactis after simulated gastric and intestinal digestion (>82%) than ALPE packaging after 42 days of accelerated storage. K-ras mimotope expression was detected intracellularly and extracellularly in the freeze-dried L. lactis-fermented milk powder upon storage. CONCLUSIONS: This suggests that fermented milk powder is a suitable food carrier for this live oral vaccine.


Subject(s)
Food Packaging , Freeze Drying , Lactococcus lactis , Lactococcus lactis/metabolism , Lactococcus lactis/genetics , Food Packaging/methods , Animals , Vacuum , Powders , Cultured Milk Products/microbiology , Fermentation , Milk/chemistry , Genes, ras/genetics , Food Storage
10.
BMC Microbiol ; 24(1): 197, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849732

ABSTRACT

BACKGROUND: Emerging evidence emphasized the role of oral microbiome in oral lichen planus (OLP). To date, no dominant pathogenic bacteria have been identified consistently. It is noteworthy that a decreased abundance of Streptococcus, a member of lactic acid bacteria (LAB) in OLP patients has been commonly reported, indicating its possible effect on OLP. This study aims to investigate the composition of LAB genera in OLP patients by high-throughput sequencing, and to explore the possible relationship between them. METHODS: We collected saliva samples from patients with OLP (n = 21) and healthy controls (n = 22) and performed 16 S rRNA gene high-throughput sequencing. In addition, the abundance of LAB genera was comprehensively analyzed and compared between OLP and HC group. To verify the expression of Lactococcus lactis, real time PCR was conducted in buccal mucosa swab from another 14 patients with OLP and 10 HC. Furthermore, the correlation was conducted between clinical severity of OLP and LAB. RESULTS: OLP and HC groups showed similar community richness and diversity. The members of LAB, Lactococcus and Lactococcus lactis significantly decreased in saliva of OLP cases and negatively associated with OLP severity. In addition, Lactococcus and Lactococcus lactis showed negative relationship with Fusobacterium and Aggregatibacter, which were considered as potential pathogens of OLP. Similarly, compared with healthy controls, the amount of Lactococcus lactis in mucosa lesion of OLP patients was significantly decreased. CONCLUSIONS: A lower amount of Lactococcus at genus level, Lactococcus lactis at species level was observed in OLP cases and associated with disease severity. Further studies to verify the relationship between LAB and OLP, as well as to explore the precise mechanism is needed.


Subject(s)
Lactobacillales , Lichen Planus, Oral , Microbiota , RNA, Ribosomal, 16S , Saliva , Humans , Saliva/microbiology , Female , Male , Lichen Planus, Oral/microbiology , Middle Aged , Lactobacillales/genetics , Lactobacillales/isolation & purification , Lactobacillales/classification , RNA, Ribosomal, 16S/genetics , Adult , High-Throughput Nucleotide Sequencing , Aged , Mouth Mucosa/microbiology , Case-Control Studies , DNA, Bacterial/genetics , Lactococcus lactis/genetics , Lactococcus lactis/isolation & purification
11.
Appl Microbiol Biotechnol ; 108(1): 397, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38922350

ABSTRACT

Functional M cells are differentiated by receptor activator of NF-κB ligand (RANKL) and capture of luminal antigens to initiate immune responses. We aimed to use postbiotic-based recombinant chicken RANKL (cRANKL) to promote M cell differentiation and test the efficacy of oral vaccines. Chicks were divided into three groups that were administered phosphate-buffered saline (PBS), cell extracts of wild-type Lactococcus lactis subsp. lactis IL1403 (WT_CE), or cell extracts of recombinant L. lactis expressing cRANKL (cRANKL_CE). The expression of the M cell marker was measured, and the gut microbiome was profiled. The efficiency of the infectious bursal disease (IBD) vaccine was tested after 12 consecutive days of administering cRANKL_CE. The chickens that were administered cRANKL_CE (p = 0.038) had significantly higher Annexin A5 (ANXA5) mRNA expression levels than those in the PBS group (PBS vs. WT_CE, p = 0.657). In the gut microbiome analysis, no significant changes were observed. However, the relative abundance of Escherichia-Shigella was negatively correlated (r = - 0.43, p = 0.019) with ANXA5 mRNA expression in Peyer's patches. cRANKL_CE/IBD (p = 0.018) had significantly higher IBD-specific faecal IgA levels than PBS/IBD (PBS/IBD vs. WT_CE/IBD, p = 0.217). Postbiotic-based recombinant cRANKL effectively improved the expression of M cell markers and the efficiency of oral vaccines. No significant changes were observed in the gut microbiome after administration of postbiotic-based recombinant cRANKL. This strategy can be used for the development of feed additives and adjuvants. KEY POINTS: • Postbiotic-based recombinant cRANKL enhanced the expression of ANXA5 in chicken. • The relative abundance of Escherichia-Shigella was negatively correlated with ANXA5 expression. • Postbiotic-based recombinant cRANKL effectively improved the efficiency of oral vaccine.


Subject(s)
Chickens , Gastrointestinal Microbiome , Lactococcus lactis , RANK Ligand , Recombinant Proteins , Animals , Chickens/immunology , Administration, Oral , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Lactococcus lactis/immunology , RANK Ligand/immunology , RANK Ligand/genetics , RANK Ligand/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/administration & dosage , Birnaviridae Infections/prevention & control , Birnaviridae Infections/immunology , Birnaviridae Infections/veterinary , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Poultry Diseases/microbiology , Infectious bursal disease virus/immunology , Infectious bursal disease virus/genetics , Cell Differentiation , Peyer's Patches/immunology
12.
Food Microbiol ; 122: 104555, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38839234

ABSTRACT

Fermentation contributes to the taste and odor of plant cheeses. The selection of functional cultures for the fermentation of plant cheeses, however, is in its infancy. This study aimed to select lactic acid bacteria for ripening of soy and lupin cheese analogues. Bacillus velezensis and B. amyloliquefaciens were used for germination of seeds to produce proteolytic enzymes; Lactococcus lactis and Lactiplantibacillus plantarum served as primary acidifying cultures. Levilactobacillus hammesii, Furfurilactobacillus milii, or Lentilactobacillus buchneri were assessed as adjunct cultures for the ripening of plant cheese. Growth of bacilli was inhibited at low pH. Both Lc. lactis and Lp. plantarum were inactived during plant cheese ripening. Cell counts of Lv. hammesii remained stable over 45 d of ripening while Ff. milii and Lt. buchneri grew slowly. Sequencing of full length 16S rRNA genes confirmed that the inocula the plant cheeses accounted for more than 98% of the bacterial communities. HPLC analysis revealed that Lt. buchneri metabolized lactate to acetate and 1,2-propanediol during ripening. Bacilli enhanced proteolysis as measured by quantification of free amino nitrogen, and the release of glutamate. LC-MS/MS analysis quantified kokumi-active dipeptides. The concentrations of γ-Glu-Leu, γ-Glu-Ile, and γ-Glu-Ala, γ-Glu-Cys in unripened cheeses were increased by seed germination but γ-Glu-Phe was degraded. Lt. buchneri but not Lv. hammesii or Ff. milii accumulated γ-Glu-Val, γ-Glu-Ile or γ-Glu-Leu during ripening, indicating strain-specific differences. In conclusion, a consortium of bacilli, acidification cultures and adjunct cultures accumulates taste- and kokumi-active compounds during ripening of plant cheeses.


Subject(s)
Cheese , Fermentation , Food Microbiology , Cheese/microbiology , Cheese/analysis , Lupinus/microbiology , Lupinus/growth & development , Glycine max/microbiology , Glycine max/growth & development , Taste , Bacillus/metabolism , Bacillus/genetics , Bacillus/growth & development , Hydrogen-Ion Concentration , Lactobacillales/metabolism , Lactobacillales/genetics , Lactobacillales/growth & development , Lactococcus lactis/metabolism , Lactococcus lactis/growth & development , Lactococcus lactis/genetics , RNA, Ribosomal, 16S/genetics
13.
Lett Appl Microbiol ; 77(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38816215

ABSTRACT

γ-Aminobutyric acid (GABA) is an inhibitory neurotransmitter of the central nervous system that impacts physical and mental health. Low GABA levels have been documented in several diseases, including multiple sclerosis and depression, and studies suggest that GABA could improve disease outcomes in those conditions. Probiotic bacteria naturally produce GABA and have been engineered to enhance its synthesis. Strains engineered thus far use inducible expression systems that require the addition of exogenous molecules, which complicates their development as therapeutics. This study aimed to overcome this challenge by engineering Lactococcus lactis with a constitutive GABA synthesis gene cassette. GABA synthesizing and transport genes (gadB and gadC) were cloned onto plasmids downstream of constitutive L. lactis promoters [P2, P5, shortened P8 (P8s)] of different strengths and transformed into L. lactis. Fold increase in gadCB expression conferred by these promoters (P2, P5, and P8s) was 322, 422, and 627, respectively, compared to the unmodified strain (P = 0.0325, P8s). GABA synthesis in the highest gadCB expressing strain, L. lactis-P8s-glutamic acid decarboxylase (GAD), was dependent on media supplementation with glutamic acid and significantly higher than the unmodified strain (P < 0.0001, 125 mM, 200 mM glutamic acid). Lactococcus lactis-P8s-GAD is poised for therapeutic testing in animal models of low-GABA-associated disease.


Subject(s)
Glutamate Decarboxylase , Lactococcus lactis , Promoter Regions, Genetic , gamma-Aminobutyric Acid , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/biosynthesis , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Genetic Engineering , Plasmids/genetics , Glutamic Acid/metabolism , Metabolic Engineering , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
14.
Microb Biotechnol ; 17(5): e14421, 2024 May.
Article in English | MEDLINE | ID: mdl-38752994

ABSTRACT

The distinct conjugation machineries encoded by plasmids pNP40 and pUC11B represent the most prevalent plasmid transfer systems among lactococcal strains. In the current study, we identified genetic determinants that underpin pNP40- and pUC11B-mediated, high-frequency mobilisation of other, non-conjugative plasmids. The mobilisation frequencies of the smaller, non-conjugative plasmids and the minimal sequences required for their mobilisation were determined, owing to the determination of the oriT sequences of both pNP40 and pUC11B, which allowed the identification of similar sequences in some of the non-conjugative plasmids that were shown to promote their mobilisation. Furthermore, the auxiliary gene mobC, two distinct functional homologues of which are present in several plasmids harboured by the pNP40- and pUC11B-carrying host strains, was observed to confer a high-frequency mobilisation phenotype. These findings provide mechanistic insights into how lactococcal conjugative plasmids achieve conjugation and promote mobilisation of non-conjugative plasmids. Ultimately, these insights would be harnessed to optimise conjugation and mobilisation strategies for the rapid and predictable development of robust and technologically improved strains.


Subject(s)
Conjugation, Genetic , Gene Transfer, Horizontal , Plasmids , Plasmids/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Lactococcus lactis/genetics
15.
Int Immunopharmacol ; 134: 112204, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38703567

ABSTRACT

Brucella infections typically occur in mucosal membranes, emphasizing the need for mucosal vaccinations. This study evaluated the effectiveness of orally administering Lactococcus lactis (L. lactis) for producing the Brucella abortus multi-epitope OMPs peptide. A multi-epitope plasmid was generated through a reverse vaccinology method, and mice were administered the genetically modified L. lactis orally as a vaccine. The plasmid underwent digestion, synthesizing a 39 kDa-sized protein known as OMPs by the target group. The sera of mice that were administered the pNZ8124-OMPs-L. lactis vaccine exhibited a notable presence of IgG1 antibodies specific to outer membrane proteins (OMPs), heightened levels of interferon (IFN-λ) and tumor necrosis factor alpha (TNF-α), and enhanced transcription rates of interleukin 4 (IL-4) and interleukin 10 (IL-10). The spleen sections from the pNZ8124-OMPs-L. lactis and IRIBA group had less morphological damage associated with inflammation, infiltration of lymphocytes, and lesions to the spleen. The findings present a novel approach to utilizing the food-grade, non-pathogenic L. lactis as a protein cell factory to synthesize innovative immunological candidate OMPs. This approach offers a distinctive way to evaluate experimental medicinal items' practicality, safety, affordability, and long-term sustainability.


Subject(s)
Brucella Vaccine , Brucella abortus , Brucellosis , Lactococcus lactis , Mice, Inbred BALB C , Animals , Brucella abortus/immunology , Brucellosis/prevention & control , Brucellosis/immunology , Lactococcus lactis/genetics , Lactococcus lactis/immunology , Brucella Vaccine/immunology , Brucella Vaccine/administration & dosage , Brucella Vaccine/genetics , Mice , Female , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Epitopes/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Spleen/immunology , Genetic Vectors , Immunoglobulin G/blood , Immunoglobulin G/immunology , Cytokines/metabolism
16.
Nat Commun ; 15(1): 3955, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729929

ABSTRACT

Widespread manganese-sensing transcriptional riboswitches effect the dependable gene regulation needed for bacterial manganese homeostasis in changing environments. Riboswitches - like most structured RNAs - are believed to fold co-transcriptionally, subject to both ligand binding and transcription events; yet how these processes are orchestrated for robust regulation is poorly understood. Through a combination of single-molecule and bulk approaches, we discover how a single Mn2+ ion and the transcribing RNA polymerase (RNAP), paused immediately downstream by a DNA template sequence, are coordinated by the bridging switch helix P1.1 in the representative Lactococcus lactis riboswitch. This coordination achieves a heretofore-overlooked semi-docked global conformation of the nascent RNA, P1.1 base pair stabilization, transcription factor NusA ejection, and RNAP pause extension, thereby enforcing transcription readthrough. Our work demonstrates how a central, adaptable RNA helix functions analogous to a molecular fulcrum of a first-class lever system to integrate disparate signals for finely balanced gene expression control.


Subject(s)
DNA-Directed RNA Polymerases , Gene Expression Regulation, Bacterial , Lactococcus lactis , Nucleic Acid Conformation , RNA, Bacterial , Riboswitch , Transcription, Genetic , Riboswitch/genetics , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/chemistry , Manganese/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Single Molecule Imaging
17.
J Dairy Sci ; 107(9): 6576-6591, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38762103

ABSTRACT

Lactococcus lactis, widely used in the manufacture of dairy products, encounters various environmental stresses both in natural habitats and during industrial processes. It has evolved intricate machinery of stress sensing and defense to survive harsh stress conditions. Here, we identified a novel TetR/AcrR family transcription regulator, designated AcrR1, to be a repressor for acid and antibiotic tolerance that was derepressed in the presence of vancomycin or under acid stress. The survival rates of acrR1 deletion strain ΔAcrR1 under acid and vancomycin stresses were about 28.7-fold (pH 3.0, HCl), 8.57-fold (pH 4.0, lactic acid) and 2.73-fold (300 ng/mL vancomycin) greater than that of original strain F44. We also demonstrated that ΔAcrR1 was better able to maintain intracellular pH homeostasis and had a lower affinity to vancomycin. No evident effects of AcrR1 deletion on the growth and morphology of strain F44 were observed. Subsequently, we characterized that the transcription level of genes associated with amino acids biosynthesis, carbohydrate transport and metabolism, multidrug resistance, and DNA repair proteins significantly upregulated in ΔAcrR1 using transcriptome analysis and quantitative reverse transcription-PCR assays. Additionally, AcrR1 could repress the transcription of the nisin post-translational modification gene, nisC, leading to a 16.3% increase in nisin yield after AcrR1 deletion. Our results not only refined the knowledge of the regulatory mechanism of TetR/AcrR family regulator in L. lactis, but presented a potential strategy to enhance industrial production of nisin.


Subject(s)
Anti-Bacterial Agents , Lactococcus lactis , Nisin , Lactococcus lactis/metabolism , Lactococcus lactis/genetics , Nisin/biosynthesis , Nisin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/biosynthesis , Drug Resistance, Microbial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial
18.
World J Microbiol Biotechnol ; 40(7): 199, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727988

ABSTRACT

Glucagon-like peptide-1(GLP-1) is an incretin hormone secreted primarily from the intestinal L-cells in response to meals. GLP-1 is a key regulator of energy metabolism and food intake. It has been proven that P9 protein from A. muciniphila could increase GLP-1 release and improve glucose homeostasis in HFD-induced mice. To obtain an engineered Lactococcus lactis which produced P9 protein, mature polypeptide chain of P9 was codon-optimized, fused with N-terminal signal peptide Usp45, and expressed in L. lactis NZ9000. Heterologous secretion of P9 by recombinant L. lactis NZP9 were successfully detected by SDS-PAGE and western blotting. Notably, the supernatant of L. lactis NZP9 stimulated GLP-1 production of NCI-H716 cells. The relative expression level of GLP-1 biosynthesis gene GCG and PCSK1 were upregulated by 1.63 and 1.53 folds, respectively. To our knowledge, this is the first report on the secretory expression of carboxyl-terminal processing protease P9 from A. muciniphila in L. lactis. Our results suggest that genetically engineered L. lactis which expressed P9 may have therapeutic potential for the treatment of diabetes, obesity and other metabolic disorders.


Subject(s)
Akkermansia , Glucagon-Like Peptide 1 , Lactococcus lactis , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/genetics , Akkermansia/genetics , Akkermansia/metabolism , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Humans , L Cells , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Animals , Mice , Cell Line , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
19.
PLoS One ; 19(4): e0298680, 2024.
Article in English | MEDLINE | ID: mdl-38557757

ABSTRACT

In the dairy industry bacteriophage (phage) contamination significantly impairs the production and quality of products like yogurt and cheese. To combat this issue, the strains of bacteria used as starter cultures possess mechanisms that make them resistant to phage infection, such as envelope resistance, or processes that render them immune to phage infection, such as restriction-modification and CRISPR-Cas. Lactococcus lactis, used to manufacture cheese and other dairy products, can also block the reproduction of infecting phages by abortive infection (Abi), a process in which phage-infected cells die before the phage replicate. We employ mathematical-computer simulation models and experiments with two Lactococcus lactis strains and two lytic phages to investigate the conditions under which Abi can limit the proliferation of phages in L. lactis populations and prevent the extinction of their populations by these viruses. According to our model, if Abi is almost perfect and there are no other populations of bacteria capable of supporting the replication of the L. lactis phages, Abi can protect bacterial populations from succumbing to infections with these viruses. This prediction is supported by the results of our experiment, which indicate that Abi can help protect L. lactis populations from extinction by lytic phage infections. However, our results also predict abortive infection is only one element of L. lactis defenses against phage infection. Mutant phages that can circumvent the Abi systems of these bacteria emerge. The survival of L. lactis populations then depends on the evolution of envelope mutants that are resistant to the evolved host-range phage.


Subject(s)
Bacteriophages , Lactococcus lactis , Bacteriophages/genetics , Lactococcus lactis/genetics , Computer Simulation , Bacterial Proteins , Bacteria
20.
BMC Genomics ; 25(1): 324, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561675

ABSTRACT

Lactococcus lactis is widely applied by the dairy industry for the fermentation of milk into products such as cheese. Adaptation of L. lactis to the dairy environment often depends on functions encoded by mobile genetic elements (MGEs) such as plasmids. Other L. lactis MGEs that contribute to industrially relevant traits like antimicrobial production and carbohydrate utilization capacities belong to the integrative conjugative elements (ICE). Here we investigate the prevalence of ICEs in L. lactis using an automated search engine that detects colocalized, ICE-associated core-functions (involved in conjugation or mobilization) in lactococcal genomes. This approach enabled the detection of 36 candidate-ICEs in 69 L. lactis genomes. By phylogenetic analysis of conserved protein functions encoded in all lactococcal ICEs, these 36 ICEs could be classified in three main ICE-families that encompass 7 distinguishable ICE-integrases and are characterized by apparent modular-exchangeability and plasticity. Finally, we demonstrate that phylogenetic analysis of the conjugation-associated VirB4 ATPase function differentiates ICE- and plasmid-derived conjugation systems, indicating that conjugal transfer of lactococcal ICEs and plasmids involves genetically distinct machineries. Our genomic analysis and sequence-based classification of lactococcal ICEs creates a comprehensive overview of the conserved functional repertoires encoded by this family of MGEs in L. lactis, which can facilitate the future exploitation of the functional traits they encode by ICE mobilization to appropriate starter culture strains.


Subject(s)
Lactococcus lactis , Lactococcus lactis/genetics , Phylogeny , Plasmids/genetics , Proteins/metabolism , Genome , Conjugation, Genetic , DNA Transposable Elements
SELECTION OF CITATIONS
SEARCH DETAIL