51
|
Exploring the Genome of Fructobacillus tropaeoli CRL 2034, a Fig-Origin Strain that Produces High Levels of Mannitol from Fructose. Curr Microbiol 2020; 77:2215-2225. [PMID: 32601836 DOI: 10.1007/s00284-020-02102-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 06/18/2020] [Indexed: 10/24/2022]
Abstract
We report the draft genome sequence of Fructobacillus tropaeoli CRL 2034, a strain isolated from ripe fig in Tucumán province, Argentina. The interest in studying the genome of this fructophilic lactic acid bacterium strain was motivated by its ability to produce high levels of mannitol from fructose. This polyol has multiple industrial applications; however, it is mainly used as low calorie sugar in the food industry. The assembled genome of this strain consists of a 1.66-Mbp circular chromosome with 1465 coding sequences and a G+C content of 44.6%. The analysis of this genome supports the one step reaction of fructose reduction to mannitol by the mannitol 2-dehydrogenase enzyme, which together with a fructose permease, were identified as involved in mannitol synthesis. In addition, a phylogenetic analysis was performed including other Leuconostocaceae members to which the Fructobacillus genus belongs to; according to the 16S rRNA gene sequences, the strain CRL 2034 was located in the Fructobacillus clade. The present genome sequence could be useful to further elucidate regulatory processes of mannitol and other bioactive metabolites and to highlight the biotechnological potential of this fruit-origin Fructobacillus strain.
Collapse
|
52
|
Buford TW, Sun Y, Roberts LM, Banerjee A, Peramsetty S, Knighton A, Verma A, Morgan D, Torres GE, Li Q, Carter CS. Angiotensin (1-7) delivered orally via probiotic, but not subcutaneously, benefits the gut-brain axis in older rats. GeroScience 2020; 42:1307-1321. [PMID: 32451847 PMCID: PMC7525634 DOI: 10.1007/s11357-020-00196-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/27/2020] [Indexed: 12/21/2022] Open
Abstract
To (1) investigate the efficacy of multiple doses of an orally delivered probiotic bacteria Lactobacillus paracasei (LP) modified to express angiotensin (1-7) (LP-A) in altering physiologic parameters relevant to the gut-brain axis in older rats and to (2) compare this strategy with subcutaneous delivery of synthetic Ang(1-7) peptide on circulating Ang(1-7) concentrations and these gut-brain axis parameters. Male 24-month-old F344BN rats received oral gavage of LP-A, or subcutaneous injection of Ang(1-7) for 0×, 1×, 3×, or 7×/week over 4 weeks. Circulating RAS analytes, inflammatory cytokines, and tryptophan and its downstream metabolites were measured by ELISA, electrochemiluminescence, and LC-MS respectively. Microbiome taxonomic analysis of fecal samples was performed via 16S-based PCR. Inflammatory and tryptophan-related mRNA expression was measured in colon and pre-frontal cortex. All dosing regimens of LP-A induced beneficial changes in fecal microbiome including overall microbiota community structure and α-diversity, while the 3×/week also significantly increased expression of the anti-inflammatory species Akkermansia muciniphila. The 3×/week also increased serum serotonin and the neuroprotective analyte 2-picolinic acid. In the colon, LP-A increased quinolinate phosphoribosyltransferase expression (1×/week) and increased kynurenine aminotransferase II (1× and 3×/week) mRNA expression. LP-A also significantly reduced neuro-inflammatory gene expression in the pre-frontal cortex (3×/week: COX2, IL-1β, and TNFα; 7×/week: COX2 and IL-1β). Subcutaneous delivery of Ang(1-7) increased circulating Ang(1-7) and reduced angiotensin II, but most gut-brain parameters were unchanged in response. Oral-but not subcutaneous-Ang(1-7) altered physiologic parameters related to gut-brain axis, with the most effects observed in 3×/week oral dosing regimen in older rats.
Collapse
Affiliation(s)
- Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL USA
| | - Yi Sun
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL USA
| | - Lisa M. Roberts
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL USA
| | - Anisha Banerjee
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL USA
| | - Sujitha Peramsetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL USA
| | - Anthony Knighton
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL USA
| | - Amrisha Verma
- Department of Ophthalmology, University of Florida, Gainesville, FL USA
| | - Drake Morgan
- Department of Psychiatry, University of Florida, Gainesville, FL USA
| | - Gonzalo E. Torres
- Department of Molecular, Cellular, and Biomedical Sciences, City College of New York, New York, NY USA
| | - Qiuhong Li
- Department of Ophthalmology, University of Florida, Gainesville, FL USA
| | - Christy S. Carter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
53
|
Garza-Morales R, Rendon BE, Malik MT, Garza-Cabrales JE, Aucouturier A, Bermúdez-Humarán LG, McMasters KM, McNally LR, Gomez-Gutierrez JG. Targeting Melanoma Hypoxia with the Food-Grade Lactic Acid Bacterium Lactococcus Lactis. Cancers (Basel) 2020; 12:cancers12020438. [PMID: 32069844 PMCID: PMC7072195 DOI: 10.3390/cancers12020438] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma is the most aggressive form of skin cancer. Hypoxia is a feature of the tumor microenvironment that reduces efficacy of immuno- and chemotherapies, resulting in poor clinical outcomes. Lactococcus lactis is a facultative anaerobic gram-positive lactic acid bacterium (LAB) that is Generally Recognized as Safe (GRAS). Recently, the use of LAB as a delivery vehicle has emerged as an alternative strategy to deliver therapeutic molecules; therefore, we investigated whether L. lactis can target and localize within melanoma hypoxic niches. To simulate hypoxic conditions in vitro, melanoma cells A2058, A375 and MeWo were cultured in a chamber with a gas mixture of 5% CO2, 94% N2 and 1% O2. Among the cell lines tested, MeWo cells displayed greater survival rates when compared to A2058 and A375 cells. Co-cultures of L. lactis expressing GFP or mCherry and MeWo cells revealed that L. lactis efficiently express the transgenes under hypoxic conditions. Moreover, multispectral optoacoustic tomography (MSOT), and near infrared (NIR) imaging of tumor-bearing BALB/c mice revealed that the intravenous injection of either L. lactis expressing β-galactosidase (β-gal) or infrared fluorescent protein (IRFP713) results in the establishment of the recombinant bacteria within tumor hypoxic niches. Overall, our data suggest that L. lactis represents an alternative strategy to target and deliver therapeutic molecules into the tumor hypoxic microenvironment.
Collapse
Affiliation(s)
- Rodolfo Garza-Morales
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (R.G.-M.); (J.E.G.-C.); (K.M.M.)
| | - Beatriz E. Rendon
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA;
| | - Mohammad Tariq Malik
- Department of Microbiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA;
| | - Jeannete E. Garza-Cabrales
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (R.G.-M.); (J.E.G.-C.); (K.M.M.)
| | - Anne Aucouturier
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (A.A.); (L.G.B.-H.)
| | - Luis G. Bermúdez-Humarán
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (A.A.); (L.G.B.-H.)
| | - Kelly M. McMasters
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (R.G.-M.); (J.E.G.-C.); (K.M.M.)
| | - Lacey R. McNally
- Department of Bioengineering, Stephenson Cancer Center, University of Oklahoma, Norman, OK 73019, USA;
| | - Jorge G. Gomez-Gutierrez
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (R.G.-M.); (J.E.G.-C.); (K.M.M.)
- Correspondence: ; Tel.: +1-(502)-852-5745
| |
Collapse
|
54
|
Liu Q, Jiang Y, Yang W, Liu Y, Shi C, Liu J, Gao X, Huang H, Niu T, Yang G, Wang C. Protective effects of a food-grade recombinant Lactobacillus plantarum with surface displayed AMA1 and EtMIC2 proteins of Eimeria tenella in broiler chickens. Microb Cell Fact 2020; 19:28. [PMID: 32046719 PMCID: PMC7014946 DOI: 10.1186/s12934-020-1297-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background Avian coccidiosis posts a severe threat to poultry production. In addition to commercial attenuated vaccines, other strategies to combat coccidiosis are urgently needed. Lactobacillus plantarum has been frequently used for expression of foreign proteins as an oral vaccine delivery system using traditional erythromycin resistance gene (erm). However, antibiotic selection markers were often used during protein expression and they pose a risk of transferring antibiotic resistance genes to the environment, and significantly restricting the application in field production. Therefore, a food-grade recombinant L. plantarum vaccine candidate would dramatically improve its application potential in the poultry industry. Results In this study, we firstly replaced the erythromycin resistance gene (erm) of the pLp_1261Inv-derived expression vector with a non-antibiotic, asd-alr fusion gene, yielding a series of non-antibiotic and reliable, food grade expression vectors. In addition, we designed a dual-expression vector that displayed two foreign proteins on the surface of L. plantarum using the anchoring sequences from either a truncated poly-γ-glutamic acid synthetase A (pgsA′) from Bacillus subtilis or the L. acidophilus surface layer protein (SlpA). EGFP and mCherry were used as marker proteins to evaluate the surface displayed properties of recombinant L. plantarum strains and were inspected by western blot, flow cytometry and fluorescence microscopy. To further determine its application as oral vaccine candidate, the AMA1 and EtMIC2 genes of E. tenella were anchored on the surface of L. plantarum strain. After oral immunization in chickens, the recombinant L. plantarum strain was able to induce antigen specific humoral, mucosal, and T cell-mediated immune responses, providing efficient protection against coccidiosis challenge. Conclusions The novel constructed food grade recombinant L. plantarum strain with double surface displayed antigens provides a potential efficient oral vaccine candidate for coccidiosis.![]()
Collapse
Affiliation(s)
- Qiong Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.,College of Food Engineering, Jilin Engineering Normal University, 3050 KaiXuan Road, Changchun, 130052, Jilin, China
| | - Yanlong Jiang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Wentao Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yongshi Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Chunwei Shi
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Jing Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Xing Gao
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Haibin Huang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Tianming Niu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Guilian Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - Chunfeng Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| |
Collapse
|
55
|
Ju AQ, Yang SB, Zhang HP, Ma X, Zhang DX, Kang YH, Shi QM, Wu TL, Wang GQ, Qian AD, Shan XF, Luan WM. Construction and immune efficacy of recombinant Lactobacillus casei strains expressing Malt from Aeromonas veronii. Microb Pathog 2020; 141:103918. [PMID: 31935441 DOI: 10.1016/j.micpath.2019.103918] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/29/2019] [Accepted: 12/09/2019] [Indexed: 11/16/2022]
Abstract
Aeromonas veronii is an important zoonotic pathogen that causes significant economic losses in the aquaculture industry. The use of probiotics in aquaculture is a practical alternative to antibiotics to promote animal health and aid in disease prevention. In the present study, we aimed to construct a recombinant Lactobacillus casei(surface-displayed or secretory) strain containing Malt from A. veronii TH0426 and assess its potential as an oral vaccine. A 1314-bp Malt gene fragment was successfully amplified and cloned into a prokaryotic protein expression system. Protein expression in resulting recombinant strains Lc-MCS-Malt (surface-displayed) and Lc-pPG-Malt (secretory) was then verified by Western blotting and indirect immunofluorescence. A single band was observed on the Western blots, with the molecular weight of the corresponding protein shown to be 48 kDa. Furthermore, a fluorescent signal for Lc-MCS-Malt was observed by fluorescence microscopy. At 0, 7, 16, 25, and 34 days post-immunization, tissue and blood samples were collected from common carp orally administered with the recombinant L. casei strains for immune-related index analyses. Treatment of common carp with the recombinant vaccine candidate stimulated high serum or skin mucus specific antibody titers and induced a higher lysozyme, ACP, SOD activity, while fish fed with Lc-pPG or PBS had no detectable immobilizing immune responses. Expression of IL-10, IL-1β, TNF-α, and IFN-γ genes in the group immunized with recombinant L. casei were significantly (P < 0.05) up regulated as compared with control groups, indicating that inflammatory response and cell immune response were triggered. Results also showed that recombinant L. casei could stimulate the mucosa through colonization of the intestine, resulting in increased transcription of IL-10, IL-1β, TNF-α, and IFN-γ. Immunity and colonization assays also showed that after 34 days of fasting, recombinant L. casei were still present in the intestines of the immunized fish. Common carp that received Lc-MCS-Malt(53.3%) and Lc-pPG-Malt (46.7%) exhibited higher survival rates than the controls after challenge with the pathogen A. veronii. Our findings suggested that recombinant L. casei can adequately protect fish and improve immunity, providing a theoretical basis for the future development of an oral Lactobacillus vaccine for use in aquaculture.
Collapse
Affiliation(s)
- An-Qi Ju
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Shu-Bao Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Hai-Peng Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Ma
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Dong-Xing Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yuan-Huan Kang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qiu-Mei Shi
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Tong-Lei Wu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Gui-Qin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Ai-Dong Qian
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xiao-Feng Shan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| | - Wei-Min Luan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
56
|
Quaranta G, Sanguinetti M, Masucci L. Fecal Microbiota Transplantation: A Potential Tool for Treatment of Human Female Reproductive Tract Diseases. Front Immunol 2019; 10:2653. [PMID: 31827467 PMCID: PMC6890827 DOI: 10.3389/fimmu.2019.02653] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/28/2019] [Indexed: 12/16/2022] Open
Abstract
The gastro-intestinal tract is an extensive organ involved in several activities, with a crucial role in immunity. Billions of commensal and transient microorganisms, known as the gut microbiota, and potential pathogens, which are constantly stimulating intestinal immunity, colonize the intestinal epithelial surface. The gut microbiota may be regarded as analogous to a solid organ with multiple different functions. In the last decade, many studies have demonstrated that intestinal bacteria can be a decisive factor in the health-disease balance of the intestine, and they can also be responsible for illnesses in other locations. For this reason, fecal microbiota transplantation (FMT) represents an important therapeutic option for Clostridium difficile infections and hold promise for different clinical conditions, such as multiple sclerosis, autism, obesity, and other systemic diseases. FMT consists of the infusion of a fecal suspension from a healthy donor to a recipient in order to restore gut flora alterations. Similar to the gut, the female reproductive tract is an example of a very complex biological ecosystem. Recent studies indicate a possible relationship between the gut and female tract microbiota, associating specific intestinal bacteria patterns with genital female diseases, such as polycystic ovary syndrome (PCOS), endometriosis and bacterial vaginosis (BV). FMT could represent a potential innovative treatment option in this field.
Collapse
Affiliation(s)
- Gianluca Quaranta
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maurizio Sanguinetti
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy.,Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Masucci
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy.,Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
57
|
van Tilburg AY, Cao H, van der Meulen SB, Solopova A, Kuipers OP. Metabolic engineering and synthetic biology employing Lactococcus lactis and Bacillus subtilis cell factories. Curr Opin Biotechnol 2019; 59:1-7. [DOI: 10.1016/j.copbio.2019.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/05/2018] [Accepted: 01/16/2019] [Indexed: 12/11/2022]
|
58
|
Zuo F, Zeng Z, Hammarström L, Marcotte H. Inducible Plasmid Self-Destruction (IPSD) Assisted Genome Engineering in Lactobacilli and Bifidobacteria. ACS Synth Biol 2019; 8:1723-1729. [PMID: 31277549 DOI: 10.1021/acssynbio.9b00114] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Genome engineering is essential for application of synthetic biology in probiotics including lactobacilli and bifidobacteria. Several homologous recombination system-based mutagenesis tools have been developed for these bacteria, but still have many limitations in different species or strains. Here we developed a genome engineering method based on an inducible self-destruction plasmid delivering homologous DNA into bacteria. Excision of the replicon by induced recombinase facilitates selection of homologous recombination events. This new genome editing tool called inducible plasmid self-destruction (IPSD) was successfully used to perform gene knockout and knock-in in lactobacilli and bifidobacteria. Due to its simplicity and universality, the IPSD strategy may provide a general approach for genetic engineering of various bacterial species.
Collapse
Affiliation(s)
- Fanglei Zuo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| | - Zhu Zeng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| | - Lennart Hammarström
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| | - Harold Marcotte
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| |
Collapse
|
59
|
Yue Z, Zhang L, Li C, Chen Y, Tai Y, Shen Y, Sun Z. Advances and potential of gene therapy for type 2 diabetes mellitus. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1643783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- Zonghao Yue
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
- Department of Food Science, Institute of Food and Drug Inspection, Zhoukou Normal University, Zhoukou, PR China
| | - Lijuan Zhang
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Chunyan Li
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Yanjuan Chen
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Yaping Tai
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Yihao Shen
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Zhongke Sun
- Department of Bioengineering, College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, PR China
- Department of Food Science, Institute of Food and Drug Inspection, Zhoukou Normal University, Zhoukou, PR China
- Department of Molecular Engineering, College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
60
|
Verma A, Xu K, Du T, Zhu P, Liang Z, Liao S, Zhang J, Raizada MK, Grant MB, Li Q. Expression of Human ACE2 in Lactobacillus and Beneficial Effects in Diabetic Retinopathy in Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:161-170. [PMID: 31380462 PMCID: PMC6661465 DOI: 10.1016/j.omtm.2019.06.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/25/2019] [Indexed: 02/06/2023]
Abstract
The angiotensin converting enzyme 2 (ACE2) catalyzes the degradation of Angiotensin II (Ang II) to generate Angiotensin-(1-7), which reduces inflammation and oxidative stress stimulated by Ang II. ACE2 has been shown to be protective in cardiovascular and metabolic diseases including diabetes and its complications. However, the challenge for its clinical application is large-scale production of high-quality ACE2 with sufficient target tissue bioavailability. We developed an expression and delivery system based on the use of probiotic species Lactobacillus paracasei (LP) to serve as a live vector for oral delivery of human ACE2. We show that codon-optimized ACE2 can be efficiently expressed in LP. Mice treated with the recombinant LP expressing the secreted ACE2 in fusion with the non-toxic subunit B of cholera toxin, which acts as a carrier to facilitate transmucosal transport, showed increased ACE2 activities in serum and tissues. ACE2-LP administration reduced the number of acellular capillaries, blocked retinal ganglion cell loss, and decreased retinal inflammatory cytokine expression in two mouse models of diabetic retinopathy. These results provide proof of concept for feasibility of using engineered probiotic species as live vector for delivery of human ACE2 with enhanced tissue bioavailability for treating diabetic retinopathy, as well as other diabetic complications.
Collapse
Affiliation(s)
- Amrisha Verma
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL 32610-0284, USA
| | - Kang Xu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL 32610-0284, USA
| | - Tao Du
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL 32610-0284, USA
| | - Ping Zhu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL 32610-0284, USA
| | - Zhibing Liang
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL 32610-0284, USA
| | - Shengquan Liao
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL 32610-0284, USA
| | - Juantao Zhang
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL 32610-0284, USA
| | - Mohan K Raizada
- Department of Physiology & Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Maria B Grant
- Department of Ophthalmology & Visual Sciences, University of Alabama, Birmingham, AL 35294, USA
| | - Qiuhong Li
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL 32610-0284, USA
| |
Collapse
|
61
|
Dendritic Cell Targeting of Bovine Viral Diarrhea Virus E2 Protein Expressed by Lactobacillus casei Effectively Induces Antigen-Specific Immune Responses via Oral Vaccination. Viruses 2019; 11:v11060575. [PMID: 31242608 PMCID: PMC6630502 DOI: 10.3390/v11060575] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 01/01/2023] Open
Abstract
Bovine viral diarrhea caused by bovine viral diarrhea virus (BVDV) is an important disease in cattle, resulting in significant economic losses to the cattle industry worldwide. In order to develop an effective vaccine against BVDV infection, we constructed a dendritic cell (DC)-targeting oral probiotic vaccine (pPG-E2-DCpep/LC W56) using Lactobacillus casei as antigen delivery carrier to express BVDV glycoprotein E2 fused with DC-targeting peptide, and the immunogenicity of orally administered probiotic vaccine was evaluated in mice model. Our results showed that after immunization with the probiotic vaccine, significantly levels of antigen-specific sera IgG and mucosal sIgA antibodies (p < 0.05) with BVDV-neutralizing activity were induced in vivo. Challenge experiment showed that pPG-E2-DCpep/LC W56 can provide effective immune protection against BVDV, and BVDV could be effectively cleared from the intestine of immunized mice post-challenge. Moreover, the pPG-E2-DCpep/LC W56 could efficiently activate DCs in the intestinal Peyer’s patches, and significantly levels of lymphoproliferative responses, Th1-associated IFN-γ, and Th2-associated IL-4 were observed in mice immunized with pPG-E2-DCpep/LC W56 (p < 0.01). Our results clearly demonstrate that the probiotic vaccine could efficiently induce anti-BVDV mucosal, humoral, and cellular immune responses via oral immunization, indicating a promising strategy for the development of oral vaccine against BVDV.
Collapse
|
62
|
Gong L, Ren C, Xu Y. Deciphering the crucial roles of transcriptional regulator GadR on gamma-aminobutyric acid production and acid resistance in Lactobacillus brevis. Microb Cell Fact 2019; 18:108. [PMID: 31196094 PMCID: PMC6567505 DOI: 10.1186/s12934-019-1157-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/07/2019] [Indexed: 11/26/2022] Open
Abstract
Background In lactic acid bacteria (LAB), acid stress leads to decreases of cell vitality and fermentation yield. Glutamate decarboxylase (GAD) system is regarded as one of the essential acid-resistance mechanisms in LAB. However, the regulation of GAD system is not well identified in the genus Lactobacillus. Although potential transcriptional regulator gene located upstream of GAD system genes was found in several Lactobacillus species, such as Lactobacillus (L.) brevis, the contribution of the regulator to acid resistance of the genus Lactobacillus has not been experimentally determined. Results The potential transcriptional regulator gene gadR was disrupted by homologous recombination in L. brevis ATCC 367, leading to the decreased expression of gadC and gadB. The inactivation of GadR completely eliminated γ-aminobutyric acid (GABA) production and decreased the glutamate-dependent acid resistance. Moreover, expression of gadC and gadB in the presence of glutamate was increased and glutamate also stimulated the expression of gadR. In addition, L. brevis D17, a strain screened from acidic fermented grains of Chinese liquor production, had much higher expression level of gadR than the typical strain L. brevis ATCC 367. Under the pH-controlled and mixed-feed fermentation, L. brevis D17 achieved a titer of 177.74 g/L and a productivity of 4.94 g/L/h of GABA within 36 h. However, the L. brevis ATCC 367 only achieved a titer of 6.44 g/L and 0.18 g/L/h of GABA although the same fermentation control approach was employed. Conclusions GadR is a positive transcriptional regulator controlling GABA conversion and acid resistance in L. brevis. L. brevis strains with hyper-expressing of gadR are excellent candidates for GABA production in industrial scale. Electronic supplementary material The online version of this article (10.1186/s12934-019-1157-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Luchan Gong
- Key Laboratory of Industrial Biotechnology of Ministry of Education, State Key Laboratory of Food Science and Technology, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Cong Ren
- Key Laboratory of Industrial Biotechnology of Ministry of Education, State Key Laboratory of Food Science and Technology, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Yan Xu
- Key Laboratory of Industrial Biotechnology of Ministry of Education, State Key Laboratory of Food Science and Technology, School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
63
|
Untersmayr E, Bax HJ, Bergmann C, Bianchini R, Cozen W, Gould HJ, Hartmann K, Josephs DH, Levi‐Schaffer F, Penichet ML, O'Mahony L, Poli A, Redegeld FA, Roth‐Walter F, Turner MC, Vangelista L, Karagiannis SN, Jensen‐Jarolim E. AllergoOncology: Microbiota in allergy and cancer-A European Academy for Allergy and Clinical Immunology position paper. Allergy 2019; 74:1037-1051. [PMID: 30636005 PMCID: PMC6563061 DOI: 10.1111/all.13718] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 12/18/2022]
Abstract
The microbiota can play important roles in the development of human immunity and the establishment of immune homeostasis. Lifestyle factors including diet, hygiene, and exposure to viruses or bacteria, and medical interventions with antibiotics or anti-ulcer medications, regulate phylogenetic variability and the quality of cross talk between innate and adaptive immune cells via mucosal and skin epithelia. More recently, microbiota and their composition have been linked to protective effects for health. Imbalance, however, has been linked to immune-related diseases such as allergy and cancer, characterized by impaired, or exaggerated immune tolerance, respectively. In this AllergoOncology position paper, we focus on the increasing evidence defining the microbiota composition as a key determinant of immunity and immune tolerance, linked to the risk for the development of allergic and malignant diseases. We discuss novel insights into the role of microbiota in disease and patient responses to treatments in cancer and in allergy. These may highlight opportunities to improve patient outcomes with medical interventions supported through a restored microbiome.
Collapse
Affiliation(s)
- Eva Untersmayr
- Institute of Pathophysiology and Allergy ResearchCenter of Pathophysiology, Infectiology and ImmunologyMedical University ViennaViennaAustria
| | - Heather J. Bax
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonGuy's HospitalLondonUK
| | | | - Rodolfo Bianchini
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| | - Wendy Cozen
- Center for Genetic EpidemiologyDepartment of Preventive MedicineKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of PathologyKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
- Norris Comprehensive Cancer CenterKeck School of Medicine of Los AngelesLos AngelesCaliforniaUSA
| | - Hannah J. Gould
- Randall Centre for Cell and Molecular BiophysicsSchool of Basic & Medical BiosciencesKing's College LondonNew Hunt's HouseLondonUK
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Karin Hartmann
- Department of DermatologyUniversity of LuebeckLuebeckGermany
| | - Debra H. Josephs
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonGuy's HospitalLondonUK
| | - Francesca Levi‐Schaffer
- Pharmacology and Experimental Therapeutics UnitSchool of PharmacyFaculty of MedicineThe Institute for Drug ResearchThe Hebrew University of JerusalemJerusalemIsrael
| | - Manuel L. Penichet
- Division of Surgical OncologyDepartment of SurgeryDavid Geffen School of MedicineUniversity of California, Los AngelesCaliforniaUSA
- Department of Microbiology, Immunology and Molecular GeneticsDavid Geffen School of MedicineUniversity of California, Los AngelesCaliforniaUSA
- Jonsson Comprehensive Cancer CenterUniversity of CaliforniaLos AngelesCaliforniaUSA
- The Molecular Biology InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
- UCLA AIDS InstituteLos AngelesCaliforniaUSA
| | - Liam O'Mahony
- Departments of Medicine and MicrobiologyAPC Microbiome IrelandNational University of IrelandCorkIreland
| | - Aurelie Poli
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Frank A. Redegeld
- Division of PharmacologyFaculty of ScienceUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Franziska Roth‐Walter
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| | - Michelle C. Turner
- Barcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
- McLaughlin Centre for Population Health Risk AssessmentUniversity of OttawaOttawaOntarioCanada
| | - Luca Vangelista
- Department of Biomedical SciencesNazarbayev University School of MedicineAstanaKazakhstan
| | - Sophia N. Karagiannis
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
| | - Erika Jensen‐Jarolim
- Institute of Pathophysiology and Allergy ResearchCenter of Pathophysiology, Infectiology and ImmunologyMedical University ViennaViennaAustria
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| |
Collapse
|
64
|
Recombinant lactococcus lactis secreting viral protein 1 of enterovirus 71 and its immunogenicity in mice. Biotechnol Lett 2019; 41:867-872. [PMID: 31119429 DOI: 10.1007/s10529-019-02695-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/19/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To construct recombinant Lactococcus lactis (L. lactis) expressing viral protein 1 (VP1) of enterovirus 71 (EV71) and evaluate its immunogenicity to be used as an oral vaccine in BALB/c mice. RESULTS Recombinant L. lactis competent in secreting VP1 (~ 30 kDa) into the extracellular environment with the aid of the signal peptide Usp45 was produced. Enzyme-linked immunosorbent assay showed that significant VP1-specific antibody response including the production of both serum IgG and fecal IgA (p < 0.05) was elicited in BALB/c mice upon oral immunization with recombinant L. lactis. Moreover, in contrast to negative control, recombinant L. lactis induced adequate neutralizing antibodies in mouse sera (p < 0.05) as demonstrated in virus neutralization assay, whereas the presence of neutralizing antibodies in fecal samples was obvious but not significant (p > 0.05). CONCLUSIONS Recombinant L. lactis expressing VP1 of EV71 has the potential to be used as an oral vaccine candidate. The findings may provide some preliminary evidences for further development of effective and needle-free EV71 vaccines.
Collapse
|
65
|
Probiotic Bacteria: A Promising Tool in Cancer Prevention and Therapy. Curr Microbiol 2019; 76:939-949. [PMID: 30949803 PMCID: PMC6586914 DOI: 10.1007/s00284-019-01679-8] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022]
Abstract
Gut microbiota is widely considered to be one of the most important components to maintain balanced homeostasis. Looking forward, probiotic bacteria have been shown to play a significant role in immunomodulation and display antitumour properties. Bacterial strains could be responsible for detection and degradation of potential carcinogens and production of short-chain fatty acids, which affect cell death and proliferation and are known as signaling molecules in the immune system. Lactic acid bacteria present in the gut has been shown to have a role in regression of carcinogenesis due to their influence on immunomodulation, which can stand as a proof of interaction between bacterial metabolites and immune and epithelial cells. Probiotic bacteria have the ability to both increase and decrease the production of anti-inflammatory cytokines which play an important role in prevention of carcinogenesis. They are also capable of activating phagocytes in order to eliminate early-stage cancer cells. Application of heat-killed probiotic bacteria coupled with radiation had a positive influence on enhancing immunological recognition of cancer cells. In the absence of active microbiota, murine immunity to carcinogens has been decreased. There are numerous cohort studies showing the correlation between ingestion of dairy products and the risk of colon and colorectal cancer. An idea of using probiotic bacteria as vectors to administer drugs has emerged lately as several papers presenting successful results have been revealed. Within the next few years, probiotic bacteria as well as gut microbiota are likely to become an important component in cancer prevention and treatment.
Collapse
|
66
|
Clarke G, Sandhu KV, Griffin BT, Dinan TG, Cryan JF, Hyland NP. Gut Reactions: Breaking Down Xenobiotic–Microbiome Interactions. Pharmacol Rev 2019; 71:198-224. [DOI: 10.1124/pr.118.015768] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
67
|
Wang W, Song Y, Liu L, Zhang Y, Wang T, Zhang W, Li K, Qi X, Gao Y, Gao L, Liu C, Zhang Y, Wang Y, Pan Q, He G, Wang X, Cui H. Neutralizing-antibody-mediated protection of chickens against infectious bursal disease via one-time vaccination with inactivated recombinant Lactococcus lactis expressing a fusion protein constructed from the RCK protein of Salmonella enterica and VP2 of infectious bursal disease virus. Microb Cell Fact 2019; 18:21. [PMID: 30704494 PMCID: PMC6357496 DOI: 10.1186/s12934-019-1061-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/11/2019] [Indexed: 02/07/2023] Open
Abstract
Background Infectious bursal disease (IBD) is an acute contagious immunosuppressive disease which lead to acute bursal injury and immune dysfunction in poultry. It has caused heavy economic losses in the commercial poultry industry for many years in worldwide. Attenuated live vaccine has widely used in poultry showing some promising signs against IBDV infection. But it has defects such as generating enhanced virulence and immunosuppression prohibits. Therefore, the development of mucosal vaccines using the food-grade lactic acid bacterium is necessary. Here, we construct a recombinant Lactococcus co-expressing the major IBDV antigens VP2 and RCK protein of Salmonella enterica to prevent IBD. Results The recombinant fusion protein VP2-RCK was expressed in a soluble and stable form in the cytoplasm of the recombinant Lactococcus lactis. Animal experiments showed that: (1) the survival rates of the injected immunization inactivated recombinant LAB group and oral immunization live recombinant LAB group were 100% and 80%, respectively; (2) ELISA titers of all serum samples from all experimental groups were negative, but high amounts of specific neutralizing antibodies were detected (1:210 to 1:212); and (3) the bursas of the injected immunization inactivated recombinant LAB group did not suffer damage, as confirmed by clinical observation and bursal histopathological examination. Our results indicate that r-L. lactis-OptiVP2-RCK induces a specific neutralizing-antibody-mediated immune response that confers full protection against very-virulent IBDV (vvIBDV) challenge. Conclusion Lactococcus lactis NZ3900 strain and its matching plasmid pNZ8149 could express the recombinant fusion protein VP2-RCK in a soluble form in the cytoplasm. The protective efficacy of r-L. lactis-OptiVP2-RCK (100%) was better than r-L. lactis-OptiVP2 (0%) which prove RCK protein played its unique role. The neutralizing antibodies titers against infectious bursal disease virus via one-time vaccination with inactivated r-L. lactis-OptiVP2-RCK could reach 1:210 to 1:212, but ELISA titers of all serum samples were negative. For this phenomenon, perhaps because of the change of delivery pathway or the spatial structure of fusion protein. We need further study to test these hypotheses.
Collapse
Affiliation(s)
- Wenqian Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yuxin Song
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.,College of Animal Science and Technology, Shihezi University, Shihezi, 832003, Xinjiang, China
| | - Linlin Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yuan Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.,College of Animal Science and Technology, Shihezi University, Shihezi, 832003, Xinjiang, China
| | - Tingting Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Wang Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.,College of Animal Science and Technology, Shihezi University, Shihezi, 832003, Xinjiang, China
| | - Kai Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Xiaole Qi
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yulong Gao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Li Gao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Changjun Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yanping Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yongqiang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Qing Pan
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Gaoming He
- College of Animal Science and Technology, Shihezi University, Shihezi, 832003, Xinjiang, China
| | - Xiaomei Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| | - Hongyu Cui
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| |
Collapse
|
68
|
Engineering of lactic acid bacteria for delivery of therapeutic proteins and peptides. Appl Microbiol Biotechnol 2019; 103:2053-2066. [DOI: 10.1007/s00253-019-09628-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/11/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023]
|
69
|
del Rio B, Redruello B, Fernandez M, Martin MC, Ladero V, Alvarez MA. Lactic Acid Bacteria as a Live Delivery System for the in situ Production of Nanobodies in the Human Gastrointestinal Tract. Front Microbiol 2019. [PMCID: PMC6346216 DOI: 10.3389/fmicb.2018.03179] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
70
|
E. coli Nissle 1917 is a safe mucosal delivery vector for a birch-grass pollen chimera to prevent allergic poly-sensitization. Mucosal Immunol 2019; 12:132-144. [PMID: 30242254 DOI: 10.1038/s41385-018-0084-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/27/2018] [Accepted: 08/25/2018] [Indexed: 02/04/2023]
Abstract
Allergic poly-sensitization affects a large number of allergic patients and poses a great challenge for their treatment. In this study we evaluated the effects of the probiotic Escherichia coli Nissle 1917 (EcN) expressing a birch and grass pollen allergen chimera 'Bet v 1, Phl p 1 and Phl p 5' (EcN-Chim) on allergy prevention after oral or intranasal application in poly-sensitized mice. In contrast to oral application, intranasal pretreatment with EcN-Chim prior to poly-sensitization led to a significant reduction of lung inflammation (eosinophils, IL-5, and IL-13 in bronchoalveolar lavage) along with suppressed levels of allergen-specific serum IgE. The suppression was associated with increased levels of allergen-specific IgA in lungs and serum IgG2a along with increased Foxp3, TGF-β, and IL-10 mRNA in bronchial lymph nodes. In vitro EcN induced high levels of IL-10 and IL-6 in both lung and intestinal epithelial cells. Importantly, using in vivo imaging techniques we demonstrated that intranasally applied EcN do not permanently colonize nose, lung, and gut and this strain might therefore be a safe delivery vector against allergy in humans. In conclusion, our data show that intranasal application of recombinant EcN expressing a multiallergen chimera presents a novel and promising treatment strategy for prevention of allergic poly-sensitization.
Collapse
|
71
|
Maddaloni M, Kochetkova I, Hoffman C, Pascual DW. Delivery of IL-35 by Lactococcus lactis Ameliorates Collagen-Induced Arthritis in Mice. Front Immunol 2018; 9:2691. [PMID: 30515168 PMCID: PMC6255909 DOI: 10.3389/fimmu.2018.02691] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
IL-35, a relatively newly discovered cytokine belonging to the larger IL-12 family, shows unique anti-inflammatory properties, believed to be associated with dedicated receptors and signaling pathways. IL-35 plays a pivotal role in the development and the function of both regulatory B (Bregs) and T cells (Tregs). In order to further its therapeutic potential, a dairy Lactococcus lactis strain was engineered to express murine IL-35 (LL-IL35), and this recombinant strain was applied to suppress collagen-induced arthritis (CIA). Oral administration of LL-IL35 effectively reduced the incidence and disease severity of CIA. When administered therapeutically, LL-IL35 abruptly halted CIA progression with no increase in disease severity by reducing neutrophil influx into the joints. LL-IL35 treatment reduced IFN-γ and IL-17 3.7- and 8.5-fold, respectively, and increased IL-10 production compared to diseased mice. Foxp3+ and Foxp3- CD39+ CD4+ T cells were previously shown to be the Tregs responsible for conferring protection against CIA. Inquiry into their induction revealed that both CCR6+ and CCR6- Foxp3+or- CD39+ CD4+ T cells act as the source of the IL-10 induced by LL-IL35. Thus, this study demonstrates the feasibility and benefits of engineered probiotics for treating autoimmune diseases.
Collapse
Affiliation(s)
- Massimo Maddaloni
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, United States
| | - Irina Kochetkova
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Carol Hoffman
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, United States
| | - David W. Pascual
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
72
|
Pedrolli DB, Ribeiro NV, Squizato PN, de Jesus VN, Cozetto DA. Engineering Microbial Living Therapeutics: The Synthetic Biology Toolbox. Trends Biotechnol 2018; 37:100-115. [PMID: 30318171 DOI: 10.1016/j.tibtech.2018.09.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 12/31/2022]
Abstract
Microbes can be engineered to act like living therapeutics designed to perform specific actions in the human body. From fighting and preventing infections to eliminating tumors and treating metabolic disorders, engineered living systems are the next generation of therapeutics. In recent years, synthetic biologists have greatly expanded the genetic toolbox for microbial living therapeutics, adding sensors, regulators, memory circuits, delivery devices, and kill switches. These advances have paved the way for successful engineering of fully functional living therapeutics, with sensing, production, and biocontainment devices. However, some important tools are still missing from the box. In this review, we cover the most recent biological parts and approaches developed and describe the missing tools needed to build robust living therapeutics.
Collapse
Affiliation(s)
- Danielle B Pedrolli
- Universidade Estadual Paulista (UNESP), School of Pharmaceutical Sciences, Department of Bioprocess and Biotechnology, 14800-903 Araraquara, Brazil; Members of Team AQA Unesp at iGEM 2017.
| | - Nathan V Ribeiro
- Universidade Estadual Paulista (UNESP), School of Pharmaceutical Sciences, Department of Bioprocess and Biotechnology, 14800-903 Araraquara, Brazil; Members of Team AQA Unesp at iGEM 2017
| | - Patrick N Squizato
- Universidade Estadual Paulista (UNESP), School of Pharmaceutical Sciences, Department of Bioprocess and Biotechnology, 14800-903 Araraquara, Brazil; Members of Team AQA Unesp at iGEM 2017
| | - Victor N de Jesus
- Universidade Estadual Paulista (UNESP), School of Pharmaceutical Sciences, Department of Bioprocess and Biotechnology, 14800-903 Araraquara, Brazil; Members of Team AQA Unesp at iGEM 2017
| | - Daniel A Cozetto
- Universidade Estadual Paulista (UNESP), School of Pharmaceutical Sciences, Department of Bioprocess and Biotechnology, 14800-903 Araraquara, Brazil; Members of Team AQA Unesp at iGEM 2017
| | | |
Collapse
|
73
|
Namai F, Yamamoto Y, Sato T, Ogita T, Shimosato T. Recombinant mouse calcitonin gene-related peptide secreted by Lactococcus lactis inhibits lipopolysaccharide-induced inflammatory response in macrophages. Anim Sci J 2018; 89:1707-1711. [PMID: 30294861 DOI: 10.1111/asj.13115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/10/2018] [Accepted: 09/10/2018] [Indexed: 12/30/2022]
Abstract
We describe the development of a genetically modified strain of lactic acid bacteria (gmLAB) capable of producing a recombinant mouse calcitonin gene-related peptide (rCGRP). This strain (NZ-CGRP) was generated by introducing a CGRP secretion plasmid into Lactococcus lactis NZ9000. Western blotting confirmed the secretion of rCGRP in the presence of the inducer nisin. Highly purified rCGRP was obtained from the culture supernatants of NZ-CGRP. We demonstrated that prophylactic exposure of a culture of mouse peritoneal macrophages to rCGRP inhibited lipopolysaccharide (LPS) induction of tumor necrosis factor-α (TNF-α). The rCGRP-secreting gmLAB strain holds promise for development as a new anti-inflammatory prophylactic.
Collapse
Affiliation(s)
- Fu Namai
- Department of Agricultural and Life Science, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Yoshinari Yamamoto
- Department of Biofunctional Science and Technology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Takashi Sato
- Department of Pulmonology, Graduate School of Medicine, Yokohama City University, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Tasuku Ogita
- Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takeshi Shimosato
- Institute for Biomedical Sciences, Shinshu University, Nagano, Japan.,Research Center for Fungal and Microbial Dynamism, Shinshu University, Nagano, Japan
| |
Collapse
|
74
|
Balakrishnan B, Taneja V. Microbial modulation of the gut microbiome for treating autoimmune diseases. Expert Rev Gastroenterol Hepatol 2018; 12:985-996. [PMID: 30146910 DOI: 10.1080/17474124.2018.1517044] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Many studies have shown the relationship between autoimmune diseases and the gut microbiome in humans: those with autoimmune conditions display gut microbiome dysbiosis. The big question that needs to be addressed is if restoring eubiosis of the gut microbiota can help suppress the autoimmune condition by activating various immune regulatory mechanisms. Inducing these self-healing mechanisms should prolong good health in affected individuals. Area covered: Here, we review the available clinical and preclinical studies that have used selective bacteria for modulating gut microbiota for treating autoimmune diseases. The potential bacterial candidates and their mechanism of action in treating autoimmune diseases will be discussed. We searched for genetically modified and potential probiotics for diseases and discuss the most likely candidates. Expert commentary: To achieve eubiosis, manipulation of the gut microbiota must occur in some form. Several approaches for modulating gut microbiota include prebiotic diets, antimicrobial interventions, fecal microbiota transplants, and selective probiotics. One novel approach showing promising results is the use of selective bacterial candidates to modulate microbial composition. Use of single microbe for treatment has an advantage as compared to multi-species as microbes grow at different rates and if needed, a single microbe is easy to target.
Collapse
Affiliation(s)
| | - Veena Taneja
- a Department of Immunology , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
75
|
Gifre-Renom L, Cano-Garrido O, Fàbregas F, Roca-Pinilla R, Seras-Franzoso J, Ferrer-Miralles N, Villaverde A, Bach À, Devant M, Arís A, Garcia-Fruitós E. A new approach to obtain pure and active proteins from Lactococcus lactis protein aggregates. Sci Rep 2018; 8:13917. [PMID: 30224788 PMCID: PMC6141594 DOI: 10.1038/s41598-018-32213-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/15/2018] [Indexed: 11/25/2022] Open
Abstract
The production of pure and soluble proteins is a complex, protein-dependent and time-consuming process, in particular for those prone-to-aggregate and/or difficult-to-purify. Although Escherichia coli is widely used for protein production, recombinant products must be co-purified through costly processes to remove lipopolysaccharide (LPS) and minimize adverse effects in the target organism. Interestingly, Lactococcus lactis, which does not contain LPS, could be a promising alternative for the production of relevant proteins. However, to date, there is no universal strategy to produce and purify any recombinant protein, being still a protein-specific process. In this context and considering that L. lactis is also able to form functional protein aggregates under overproduction conditions, we explored the use of these aggregates as an alternative source of soluble proteins. In this study, we developed a widely applicable and economically affordable protocol to extract functional proteins from these nanoclusters. For that, two model proteins were used: mammary serum amyloid A3 (M-SAA3) and metalloproteinase 9 (MMP-9), a difficult-to-purify and a prone-to-aggregate protein, respectively. The results show that it is possible to obtain highly pure, soluble, LPS-free and active recombinant proteins from L. lactis aggregates through a cost-effective and simple protocol with special relevance for difficult-to-purify or highly aggregated proteins.
Collapse
Affiliation(s)
- L Gifre-Renom
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140, Caldes de Montbui, Spain
| | - O Cano-Garrido
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193, Cerdanyola del Vallès, Spain
| | - F Fàbregas
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140, Caldes de Montbui, Spain
| | - R Roca-Pinilla
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140, Caldes de Montbui, Spain
| | - J Seras-Franzoso
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193, Cerdanyola del Vallès, Spain.,Cibbim-Nanomedicine, Hospital Vall d'Hebron, Institut de Recerca de la Vall d'Hebron (VHIR), 08035, Barcelona, Spain
| | - N Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193, Cerdanyola del Vallès, Spain
| | - A Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, 08193, Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193, Cerdanyola del Vallès, Spain
| | - À Bach
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140, Caldes de Montbui, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - M Devant
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140, Caldes de Montbui, Spain
| | - A Arís
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140, Caldes de Montbui, Spain.
| | - E Garcia-Fruitós
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140, Caldes de Montbui, Spain.
| |
Collapse
|
76
|
Quintana I, Espariz M, Villar SR, González FB, Pacini MF, Cabrera G, Bontempi I, Prochetto E, Stülke J, Perez AR, Marcipar I, Blancato V, Magni C. Genetic Engineering of Lactococcus lactis Co-producing Antigen and the Mucosal Adjuvant 3' 5'- cyclic di Adenosine Monophosphate (c-di-AMP) as a Design Strategy to Develop a Mucosal Vaccine Prototype. Front Microbiol 2018; 9:2100. [PMID: 30258417 PMCID: PMC6143824 DOI: 10.3389/fmicb.2018.02100] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/16/2018] [Indexed: 12/20/2022] Open
Abstract
Lactococcus lactis is a promising candidate for the development of mucosal vaccines. More than 20 years of experimental research supports this immunization approach. In addition, 3′ 5′- cyclic di-adenosine monophosphate (c-di-AMP) is a bacterial second messenger that plays a key role in the regulation of diverse physiological functions (potassium and cellular wall homeostasis, among others). Moreover, recent studies showed that c-di-AMP has a strong mucosal adjuvant activity that promotes both humoral and cellular immune responses. In this study, we report the development of a novel mucosal vaccine prototype based on a genetically engineered L. lactis strain. First, we demonstrate that homologous expression of cdaA gen in L. lactis is able to increase c-di-AMP levels. Thus, we hypothesized that in vivo synthesis of the adjuvant can be combined with production of an antigen of interest in a separate form or jointly in the same strain. Therefore, a specifically designed fragment of the trans-sialidase (TScf) enzyme from the Trypanosoma cruzi parasite, the etiological agent of Chagas disease, was selected to evaluate as proof of concept the immune response triggered by our vaccine prototypes. Consequently, we found that oral administration of a L. lactis strain expressing antigenic TScf combined with another L. lactis strain producing the adjuvant c-di-AMP could elicit a TS-specific immune response. Also, an additional L. lactis strain containing a single plasmid with both cdaA and tscf genes under the Pcit and Pnis promoters, respectively, was also able to elicit a specific immune response. Thus, the current report is the first one to describe an engineered L. lactis strain that simultaneously synthesizes the adjuvant c-di-AMP as well as a heterologous antigen in order to develop a simple and economical system for the formulation of vaccine prototypes using a food grade lactic acid bacterium.
Collapse
Affiliation(s)
- Ingrid Quintana
- Laboratorio de Fisiología y Genética de Bacterias Lácticas, Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET UNR), Universidad Nacional de Rosario, Rosario, Argentina.,Department of General Microbiology, GZMB, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Martín Espariz
- Laboratorio de Fisiología y Genética de Bacterias Lácticas, Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET UNR), Universidad Nacional de Rosario, Rosario, Argentina.,Laboratorio de Biotecnología e Inocuidad de los Alimentos, Facultad de Ciencias Bioquímicas y Farmacéuticas - Municipalidad de Granadero Baigorria (UNR), Rosario, Argentina
| | - Silvina R Villar
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET UNR), Rosario, Argentina.,Facultad de Ciencias Médicas, Centro de Investigación y Producción de Reactivos Biológicos, Universidad Nacional de Rosario, Rosario, Argentina
| | - Florencia B González
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET UNR), Rosario, Argentina
| | - Maria F Pacini
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET UNR), Rosario, Argentina
| | - Gabriel Cabrera
- Laboratorio de Tecnología Inmunológica, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Iván Bontempi
- Laboratorio de Tecnología Inmunológica, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Estefanía Prochetto
- Laboratorio de Tecnología Inmunológica, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Jörg Stülke
- Department of General Microbiology, GZMB, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Ana R Perez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET UNR), Rosario, Argentina.,Facultad de Ciencias Médicas, Centro de Investigación y Producción de Reactivos Biológicos, Universidad Nacional de Rosario, Rosario, Argentina
| | - Iván Marcipar
- Laboratorio de Tecnología Inmunológica, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Victor Blancato
- Laboratorio de Fisiología y Genética de Bacterias Lácticas, Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET UNR), Universidad Nacional de Rosario, Rosario, Argentina.,Laboratorio de Biotecnología e Inocuidad de los Alimentos, Facultad de Ciencias Bioquímicas y Farmacéuticas - Municipalidad de Granadero Baigorria (UNR), Rosario, Argentina
| | - Christian Magni
- Laboratorio de Fisiología y Genética de Bacterias Lácticas, Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET UNR), Universidad Nacional de Rosario, Rosario, Argentina.,Laboratorio de Biotecnología e Inocuidad de los Alimentos, Facultad de Ciencias Bioquímicas y Farmacéuticas - Municipalidad de Granadero Baigorria (UNR), Rosario, Argentina
| |
Collapse
|
77
|
Synthetic biology in probiotic lactic acid bacteria: At the frontier of living therapeutics. Curr Opin Biotechnol 2018; 53:224-231. [PMID: 29550614 DOI: 10.1016/j.copbio.2018.01.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/22/2017] [Accepted: 01/24/2018] [Indexed: 12/19/2022]
Abstract
The trillions of microbes hosted by humans can dictate health or illness depending on a multitude of genetic, environmental, and lifestyle factors that help define the human ecosystem. As the human microbiota is characterized, so can the interconnectivity of microbe-host-disease be realized and manipulated. Designing microbes as therapeutic agents can not only enable targeted drug delivery but also restore homeostasis within a perturbed microbial community. Used for centuries in fermentation and preservation of food, lactic acid bacteria (LAB) have a long history of safe, and occasionally health promoting, interactions with the human gut, making them ideal candidates for engineered functionality. This review outlines available genetic tools, recent developments in biomedical applications, as well as potential future applications of synthetic biology to program LAB-based therapeutic systems.
Collapse
|
78
|
Pidutti P, Federici F, Brandi J, Manna L, Rizzi E, Marini U, Cecconi D. Purification and characterization of ribosomal proteins L27 and L30 having antimicrobial activity produced by the Lactobacillus salivarius SGL 03. J Appl Microbiol 2018; 124:398-407. [PMID: 29159916 DOI: 10.1111/jam.13646] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/31/2017] [Accepted: 11/13/2017] [Indexed: 01/31/2023]
Abstract
AIMS The aim of this study was to investigate the antimicrobial potential of proteins secreted by a new strain of Lactobacillus salivarius. METHODS AND RESULTS The secretome of L. salivarius SGL 03 strain was analysed by gel-assisted fractionation and MS/MS to identify low-molecular-mass proteins. This strategy allowed us to identify 10 secreted proteins. Then, a combination of heterologous expression and agar well diffusion was used to characterize them as to their antimicrobial activity, mechanisms of action and stability. Our findings indicate that L27 and L30 proteins of the 50S ribosomal subunit have antimicrobial activity against Streptococcus pyogenes, Streptococcus uberis and Enterococcus faecium. In addition, both proteins are bactericidal against S. pyogenes and maintain their antimicrobial activity after different protease treatments, at acidic pH, after heat treatment, and if stored in a refrigerated ambient at least at 4°C. CONCLUSIONS The overall results demonstrated that the L27 and L30 ribosomal proteins are of interest as new antimicrobial molecules to prevent the growth of S. pyogenes, S. uberis and E. faecium. SIGNIFICANCE AND IMPACT OF THE STUDY Our results provide the first insight into the extra-ribosomal activity of L27 and L30 secreted proteins of L. salivarius. This study demonstrated the capacity of L. salivarius SGL 03 to produce antimicrobial molecules and suggested this strain as a promising probiotic candidate.
Collapse
Affiliation(s)
- P Pidutti
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona, Italy
| | | | - J Brandi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona, Italy
| | - L Manna
- Sintal Dietetics s.r.l., Teramo, Italy
| | - E Rizzi
- Sintal Dietetics s.r.l., Teramo, Italy
| | - U Marini
- Sintal Dietetics s.r.l., Teramo, Italy
| | - D Cecconi
- Department of Biotechnology, Proteomics and Mass Spectrometry Laboratory, University of Verona, Verona, Italy
| |
Collapse
|
79
|
Zhang DX, Kang YH, Chen L, Siddiqui SA, Wang CF, Qian AD, Shan XF. Oral immunization with recombinant Lactobacillus casei expressing OmpAI confers protection against Aeromonas veronii challenge in common carp, Cyprinus carpio. FISH & SHELLFISH IMMUNOLOGY 2018; 72:552-563. [PMID: 29155272 DOI: 10.1016/j.fsi.2017.10.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/10/2017] [Accepted: 10/24/2017] [Indexed: 06/07/2023]
Abstract
Aeromonas veronii is a gram-negative pathogen capable of infecting both fish and mammals, including humans, and natural infection in fish results in irreparable damage to the aquaculture industry. Lactic acid bacteria (LAB) have a number of properties that make them attractive candidates as delivery vehicles for presentation to the mucosa sites of compounds with pharmaceutical interest, in particular vaccines. In this study, we generated two recombinant Lactobacillus casei (surface-displayed or secretory) expressing the OmpAI of A.veronii and evaluated the effect on immune responses in fish model. A 1022 bp gene fragment of the 42 kDa OmpAI antigen of A.veronii was cloned into pPG-1 (surface-displayed) and pPG-2 (secretory) and electrotransformed into Lactobacillus casei CC16. The recombinant plasmid in L.casei could be stably inherited over 50 generations, and production of OmpAI protein had slight limited effects on cells growth. Treatment of common carp with the recombinant vaccine candidate stimulated high serum or skin mucus specific antibody titers and induced a higher lysozyme, ACP, SOD activity, while fish fed with Lc-pPG or PBS had no detectable immobilizing immune responses. Expression of IL-10, IL-β, IFN-γ, TNF-α genes in the group immunized with recombinant L.casei were significantly (P < 0.05) up regulated as compared with control groups, indicating that inflammatory response and cell immune response were triggered. Further, viable recombinant L.casei strains were directly delivered and survive throughout the intestinal tract, the recombinant OmpAI was also detected in intestine mucosal. The results showed that common carp received Lc-pPG1-OmpAI (66.7%) and Lc-pPG2-OmpAI (50.0%) had higher survival rates compared with the controls after challenge with A.veronii, indicating that Lc-pPG1-OmpAI and Lc-pPG2-OmpAI had beneficial effects on immune response and enhanced disease resistance of common carp against A.veronii infection. Our study here demonstrates, for the first time, the ability of recombinant L.casei as oral vaccine against A.veronii infection in carps. The combination of OmpAI delivery and LAB approach may be a promising mucosal therapeutic agent for treating and controlling A.veronii.
Collapse
Affiliation(s)
- Dong-Xing Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Yuan-Huan Kang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Long Chen
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Shahrood Ahmed Siddiqui
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Chun-Feng Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Ai-Dong Qian
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| | - Xiao-Feng Shan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
80
|
Abstract
ABSTRACT
The
Lactobacillus
genus is a diverse group of microorganisms, many of which are of industrial and medical relevance. Several
Lactobacillus
species have been used as probiotics, organisms that when present in sufficient quantities confer a health benefit to the host. A significant limitation to the mechanistic understanding of how these microbes provide health benefits to their hosts and how they can be used as therapeutic delivery systems has been the lack of genetic strategies to efficiently manipulate their genomes. This article will review the development and employment of traditional genetic tools in lactobacilli and highlight the latest methodologies that are allowing for precision genome engineering of these probiotic organisms. The application of these tools will be key in providing mechanistic insights into probiotics as well as maximizing the value of lactobacilli as either a traditional probiotic or as a platform for the delivery of therapeutic proteins. Finally, we will discuss concepts that we consider relevant for the delivery of engineered therapeutics to the human gut.
Collapse
|
81
|
Golomb BL, Yu AO, Coates LC, Marco ML. The Lactococcus lactis KF147 nonribosomal peptide synthetase/polyketide synthase system confers resistance to oxidative stress during growth on plant leaf tissue lysate. Microbiologyopen 2017; 7. [PMID: 28921941 PMCID: PMC5822349 DOI: 10.1002/mbo3.531] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 07/24/2017] [Accepted: 08/01/2017] [Indexed: 01/07/2023] Open
Abstract
Strains of Lactococcus lactis isolated from plant tissues possess adaptations that support their survival and growth in plant‐associated microbial habitats. We previously demonstrated that genes coding for a hybrid nonribosomal peptide synthetase/polyketide synthase (NRPS/PKS) system involved in production of an uncharacterized secondary metabolite are specifically induced in L. lactis KF147 during growth on plant tissues. Notably, this NRPS/PKS has only been identified in plant‐isolated strains of L. lactis. Here, we show that the L. lactis KF147 NRPS/PKS genes have homologs in certain Streptococcus mutans isolates and the genetic organization of the NRPS/PKS locus is conserved among L. lactis strains. Using an L. lactis KF147 mutant deficient in synthesis of NrpC, a 4′‐phosphopantetheinyl transferase, we found that the NRPS/PKS system improves L. lactis during growth under oxidative conditions in Arapidopsis thaliana leaf lysate. The NRPS/PKS system also improves tolerance of L. lactis to reactive oxygen species and specifically H2O2 and superoxide radicals in culture medium. These findings indicate that this secondary metabolite provides a novel mechanism for reactive oxygen species detoxification not previously known for this species.
Collapse
Affiliation(s)
- Benjamin L Golomb
- Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Annabelle O Yu
- Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Laurynne C Coates
- Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, CA, USA
| |
Collapse
|
82
|
Recombinant Mouse Osteocalcin Secreted by Lactococcus lactis Promotes Glucagon-Like Peptide-1 Induction in STC-1 Cells. Curr Microbiol 2017; 75:92-98. [DOI: 10.1007/s00284-017-1354-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/06/2017] [Indexed: 12/23/2022]
|
83
|
Shigemori S, Namai F, Yamamoto Y, Nigar S, Sato T, Ogita T, Shimosato T. Genetically modified Lactococcus lactis producing a green fluorescent protein–bovine lactoferrin fusion protein suppresses proinflammatory cytokine expression in lipopolysaccharide-stimulated RAW 264.7 cells. J Dairy Sci 2017; 100:7007-7015. [DOI: 10.3168/jds.2017-12872] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/12/2017] [Indexed: 01/02/2023]
|
84
|
Recombinant Lactococcus lactis expressing bioactive exendin-4 to promote insulin secretion and beta-cell proliferation in vitro. Appl Microbiol Biotechnol 2017; 101:7177-7186. [DOI: 10.1007/s00253-017-8410-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/24/2017] [Accepted: 06/26/2017] [Indexed: 12/14/2022]
|
85
|
Oloketuyi SF, Khan F. Inhibition strategies of Listeria monocytogenes biofilms-current knowledge and future outlooks. J Basic Microbiol 2017; 57:728-743. [PMID: 28594071 DOI: 10.1002/jobm.201700071] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/12/2017] [Accepted: 05/12/2017] [Indexed: 12/30/2022]
Abstract
There is an increasing trend in the food industry on the Listeria monocytogenes biofilm formation and inhibition. This is attributed to its easy survival on contact surfaces, resistance to disinfectants or antibiotics and growth under the stringent condition used for food processing and preservation thereby leading to food contamination products by direct or indirect exposure. Though, there is a lack of conclusive evidences about the mechanism of biofilm formation, in this review, the concept of biofilm formation and various chemical, physical, and green technology approaches to prevent or control the biofilm formed is discussed. State-of-the-art approaches ranging from the application of natural to synthetic molecules with high effectiveness and non-toxicity targeted at the different steps of biofilm formation could positively influence the biofilm inhibition in the future.
Collapse
Affiliation(s)
- Sandra F Oloketuyi
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, U.P., India
| | - Fazlurrahman Khan
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, U.P., India
| |
Collapse
|
86
|
Isolation of two lactobacilli, producers of two new bacteriocin-like substances (BLS) for potential food-preservative use. Eur Food Res Technol 2017. [DOI: 10.1007/s00217-017-2913-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
87
|
Use of genetically modified bacteria for drug delivery in humans: Revisiting the safety aspect. Sci Rep 2017; 7:2294. [PMID: 28536456 PMCID: PMC5442108 DOI: 10.1038/s41598-017-02591-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/12/2017] [Indexed: 01/05/2023] Open
Abstract
The use of live, genetically modified bacteria as delivery vehicles for biologics is of considerable interest scientifically and has attracted significant commercial investment. We have pioneered the use of the commensal gut bacterium Bacteroides ovatus for the oral delivery of therapeutics to the gastrointestinal tract. Here we report on our investigations of the biological safety of engineered B. ovatus bacteria that includes the use of thymineless death as a containment strategy and the potential for the spread of transgenes in vivo in the mammalian gastrointestinal tract. We demonstrate the ability of GM-strains of Bacteroides to survive thymine starvation and overcome it through the exchange of genetic material. We also provide evidence for horizontal gene transfer in the mammalian gastrointestinal tract resulting in transgene-carrying wild type bacteria. These findings sound a strong note of caution on the employment of live genetically modified bacteria for the delivery of biologics.
Collapse
|
88
|
Tan TS, Syed Hassan S, Yap WB. Expression of surface-bound nonstructural 1 (NS1) protein of influenza virus A H5N1 on Lactobacillus casei strain C1. Lett Appl Microbiol 2017; 64:446-451. [PMID: 28370088 DOI: 10.1111/lam.12738] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/28/2017] [Indexed: 12/26/2022]
Abstract
The study aimed to construct a recombinant Lactobacillus casei expressing the nonstructural (NS) 1 protein of influenza A virus H5N1 on its cell wall. The NS1 gene was first amplified and fused to the pSGANC332 expression plasmid. The NS1 protein expression was carried out by Lact. casei strain C1. PCR screening and DNA sequencing confirmed the presence of recombinant pSG-NS1-ANC332 plasmid in Lact. casei. The plasmid was stably maintained (98·94 ± 1·65%) by the bacterium within the first 20 generations without selective pressure. The NS1 was expressed as a 49-kDa protein in association with the anchoring peptide. The yield was 1·325 ± 0·065 μg mg-1 of bacterial cells. Lactobacillus casei expressing the NS1 on its cell wall was red-fluorescently stained, but the staining was not observed on Lact. casei carrying the empty pSGANC332. The results implied that Lact. casei strain C1 is a promising host for the expression of surface-bound NS1 protein using the pSGANC332 expression plasmid. SIGNIFICANCE AND IMPACT OF THE STUDY The study has demonstrated, for the first time, the expression of nonstructural 1 (NS1) protein of influenza A virus H5N1 on the cell wall of Lactobacillus casei using the pSGANC332 expression plasmid. Display of NS1 protein on the bacterial cell wall was evident under an immunofluorescence microscopic observation. Lactobacillus casei carrying the NS1 protein could be developed into a universal oral influenza vaccine since the NS1 is highly conserved among influenza viruses.
Collapse
Affiliation(s)
- T S Tan
- Biomedical Science Programme, School of Diagnostic and Applied Health Sciences, Faculty of Health Sciences, National University of Malaysia, Kuala Lumpur, Malaysia
| | - S Syed Hassan
- School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway Selangor, Malaysia
| | - W B Yap
- Biomedical Science Programme, School of Diagnostic and Applied Health Sciences, Faculty of Health Sciences, National University of Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
89
|
Yang WC, Hsu TC, Cheng KC, Liu JR. Expression of the Clonostachys rosea lactonohydrolase gene by Lactobacillus reuteri to increase its zearalenone-removing ability. Microb Cell Fact 2017; 16:69. [PMID: 28438205 PMCID: PMC5404306 DOI: 10.1186/s12934-017-0687-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/21/2017] [Indexed: 01/08/2023] Open
Abstract
Background Mycotoxins are secondary metabolites produced by filamentous fungi that can contaminate agricultural crops in the field as well as during harvest, transportation, processing, or storage. Zearalenone (ZEN), a non-steroidal estrogenic mycotoxin, produced by Fusarium species, has been shown to be associated with reproductive disorders in farm animals and to a lesser extent in hyperoestrogenic syndromes in humans. Thus, the decontamination of ZEN in foods and feeds is an important issue. Results In this study, the gene encoding ZHD101, a ZEN-degrading enzyme produced by Clonostachys rosea IFO 7063, was cloned into an Escherichia coli–Lactobacillus shuttle vector, pNZ3004, and the resultant plasmid pNZ-zhd101 was then introduced via electroporation into Lactobacillus reuteri Pg4, a probiotic strain isolated from the gastrointestinal tract of broilers. The transformed strain L. reuteri pNZ-zhd101 acquired the capacity to degrade ZEN. In addition, the production of recombinant ZHD101 did not affect cell growth, acid and bile salt tolerance, and had only a minor effect on the adhesion ability of L. reuteri pNZ-zhd101. Conclusions To the best of our knowledge, this is the first report of successful expression of a ZEN-degrading enzyme by intestinal lactobacilli.
Collapse
Affiliation(s)
- Wen-Chun Yang
- Graduate Institute of Food Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Tsui-Chun Hsu
- Department of Animal Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Kuan-Chen Cheng
- Graduate Institute of Food Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan. .,Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402, Taiwan.
| | - Je-Ruei Liu
- Department of Animal Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan. .,Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan. .,Agricultural Biotechnology Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.
| |
Collapse
|
90
|
Bosma EF, Forster J, Nielsen AT. Lactobacilli and pediococci as versatile cell factories - Evaluation of strain properties and genetic tools. Biotechnol Adv 2017; 35:419-442. [PMID: 28396124 DOI: 10.1016/j.biotechadv.2017.04.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/29/2017] [Accepted: 04/03/2017] [Indexed: 12/14/2022]
Abstract
This review discusses opportunities and bottlenecks for cell factory development of Lactic Acid Bacteria (LAB), with an emphasis on lactobacilli and pediococci, their metabolism and genetic tools. In order to enable economically feasible bio-based production of chemicals and fuels in a biorefinery, the choice of product, substrate and production organism is important. Currently, the most frequently used production hosts include Escherichia coli and Saccharomyces cerevisiae, but promising examples are available of alternative hosts such as LAB. Particularly lactobacilli and pediococci can offer benefits such as thermotolerance, an extended substrate range and increased tolerance to stresses such as low pH or high alcohol concentrations. This review will evaluate the properties and metabolism of these organisms, and provide an overview of their current biotechnological applications and metabolic engineering. We substantiate the review by including experimental results from screening various lactobacilli and pediococci for transformability, growth temperature range and ability to grow under biotechnologically relevant stress conditions. Since availability of efficient genetic engineering tools is a crucial prerequisite for industrial strain development, genetic tool development is extensively discussed. A range of genetic tools exist for Lactococcus lactis, but for other species of LAB like lactobacilli and pediococci such tools are less well developed. Whereas lactobacilli and pediococci have a long history of use in food and beverage fermentation, their use as platform organisms for production purposes is rather new. By harnessing their properties such as thermotolerance and stress resistance, and by using emerging high-throughput genetic tools, these organisms are very promising as versatile cell factories for biorefinery applications.
Collapse
Affiliation(s)
- Elleke F Bosma
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet B220, 2800 Kgs. Lyngby, Denmark
| | - Jochen Forster
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet B220, 2800 Kgs. Lyngby, Denmark
| | - Alex Toftgaard Nielsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet B220, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
91
|
|
92
|
Gifre L, Arís A, Bach À, Garcia-Fruitós E. Trends in recombinant protein use in animal production. Microb Cell Fact 2017; 16:40. [PMID: 28259156 PMCID: PMC5336677 DOI: 10.1186/s12934-017-0654-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 02/26/2017] [Indexed: 02/06/2023] Open
Abstract
Recombinant technologies have made possible the production of a broad catalogue of proteins of interest, including those used for animal production. The most widely studied proteins for the animal sector are those with an important role in reproduction, feed efficiency, and health. Nowadays, mammalian cells and fungi are the preferred choice for recombinant production of hormones for reproductive purposes and fibrolytic enzymes to enhance animal performance, respectively. However, the development of low-cost products is a priority, particularly in livestock. The study of cell factories such as yeast and bacteria has notably increased in the last decades to make the new developed reproductive hormones and fibrolytic enzymes a real alternative to the marketed ones. Important efforts have also been invested to developing new recombinant strategies for prevention and therapy, including passive immunization and modulation of the immune system. This offers the possibility to reduce the use of antibiotics by controlling physiological processes and improve the efficacy of preventing infections. Thus, nowadays different recombinant fibrolytic enzymes, hormones, and therapeutic molecules with optimized properties have been successfully produced through cost-effective processes using microbial cell factories. However, despite the important achievements for reducing protein production expenses, alternative strategies to further reduce these costs are still required. In this context, it is necessary to make a giant leap towards the use of novel strategies, such as nanotechnology, that combined with recombinant technology would make recombinant molecules affordable for animal industry.
Collapse
Affiliation(s)
- Laia Gifre
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140 Caldes de Montbui, Spain
| | - Anna Arís
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140 Caldes de Montbui, Spain
| | - Àlex Bach
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140 Caldes de Montbui, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Elena Garcia-Fruitós
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140 Caldes de Montbui, Spain
| |
Collapse
|
93
|
Shigemori S, Shimosato T. Applications of Genetically Modified Immunobiotics with High Immunoregulatory Capacity for Treatment of Inflammatory Bowel Diseases. Front Immunol 2017; 8:22. [PMID: 28179904 PMCID: PMC5263139 DOI: 10.3389/fimmu.2017.00022] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), including ulcerative colitis and Crohn’s disease, are chronic inflammatory diseases characterized by dysregulated immune responses of the gastrointestinal tract. In recent years, the incidence of IBDs has increased in developed nations, but their prophylaxis/treatment is not yet established. Site-directed delivery of molecules showing anti-inflammatory properties using genetically modified (gm)-probiotics shows promise as a new strategy for the prevention and treatment of IBD. Advantages of gm-probiotics include (1) the ability to use bacteria as a delivery vehicle, enabling safe and long-term use by humans, (2) decreased risks of side effects, and (3) reduced costs. The intestinal delivery of anti-inflammatory proteins such as cytokines and enzymes using Lactococcus lactis has been shown to regulate host intestinal homeostasis depending on the delivered protein-specific machinery. Additionally, clinical experience using interleukin 10-secreting Lc. lactis has been shown to be safe and to facilitate biological containment in IBD therapy. On the other hand, some preclinical studies have demonstrated that gm-strains of immunobiotics (probiotic strains able to beneficially regulate the mucosal immunity) provide beneficial effects on intestinal inflammation as a result of the synergy between the immunoregulatory effects of the bacterium itself and the anti-inflammatory effects of the delivered recombinant proteins. In this review, we discuss the rapid progression in the development of strategies for the prophylaxis and treatment of IBD using gm-probiotics that exhibit immune regulation effects (gm-immunobiotics). In particular, we discuss the type of strains used as delivery agents.
Collapse
Affiliation(s)
- Suguru Shigemori
- Department of Bioscience and Food Production Science, Interdisciplinary Graduate School of Science and Technology, Shinshu University, Nagano, Japan; Japan Society for the Promotion of Science, Tokyo, Japan
| | - Takeshi Shimosato
- Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan; Supramolecular Complexes Unit, Research Center for Fungal and Microbial Dynamism, Shinshu University, Nagano, Japan
| |
Collapse
|
94
|
Zeng Z, Yu R, Zuo F, Zhang B, Peng D, Ma H, Chen S. Heterologous Expression and Delivery of Biologically Active Exendin-4 by Lactobacillus paracasei L14. PLoS One 2016; 11:e0165130. [PMID: 27764251 PMCID: PMC5072737 DOI: 10.1371/journal.pone.0165130] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/06/2016] [Indexed: 01/31/2023] Open
Abstract
Exendin-4, a glucagon-like protein-1 (GLP-1) receptor agonist, is an excellent therapeutic peptide drug for type 2 diabetes due to longer lasting biological activity compared to GLP-1. This study explored the feasibility of using probiotic Lactobacillus paracasei as an oral vector for recombinant exendin-4 peptide delivery, an alternative to costly chemical synthesis and inconvenient administration by injection. L. paracasei transformed with a plasmid encoding the exendin-4 gene (L. paracasei L14/pMG76e-exendin-4) with a constitutive promotor was successfully constructed and showed efficient secretion of exendin-4. The secreted exendin-4 significantly enhanced insulin secretion of INS-1 β-cells, along with an increment in their proliferation and inhibition of their apoptosis, corresponding to the effect of GLP-1 on these cells. The transcription level of the pancreatic duodenal homeobox-1 gene (PDX-1), a key transcription factor for cellular insulin synthesis and secretion, was upregulated by the treatment with secreted exendin-4, paralleling the upregulation of insulin gene expression. Caco-2 cell monolayer permeability assay showed a 34-fold increase in the transport of exendin-4 delivered by L. paracasei vs. that of free exendin-4 (control), suggesting effective facilitation of exendin-4 transport across the intestinal barrier by this delivery system. This study demonstrates that the probiotic Lactobacillus can be engineered to secrete bioactive exendin-4 and facilitate its transport through the intestinal barrier, providing a novel strategy for oral exendin-4 delivery using this lactic acid bacterium.
Collapse
Affiliation(s)
- Zhu Zeng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
| | - Rui Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
| | - Fanglei Zuo
- Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
| | - Bo Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
| | - Deju Peng
- Yangling Zhongyang Joint Ranch Co. Ltd., Beiyang Breeding Area, Yangling Street Agency, Yangling District, Xi'an, P. R. China
| | - Huiqin Ma
- College of Agriculture and Biotechnology, China Agricultural University, Beijing, P. R. China
| | - Shangwu Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, P. R. China
- * E-mail:
| |
Collapse
|
95
|
pMPES: A Modular Peptide Expression System for the Delivery of Antimicrobial Peptides to the Site of Gastrointestinal Infections Using Probiotics. Pharmaceuticals (Basel) 2016; 9:ph9040060. [PMID: 27782051 PMCID: PMC5198035 DOI: 10.3390/ph9040060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/14/2016] [Indexed: 12/18/2022] Open
Abstract
Antimicrobial peptides are a promising alternative to traditional antibiotics, but their utility is limited by high production costs and poor bioavailability profiles. Bacterial production and delivery of antimicrobial peptides (AMPs) directly at the site of infection may offer a path for effective therapeutic application. In this study, we have developed a vector that can be used for the production and secretion of seven antimicrobial peptides from both Escherichia coli MC1061 F' and probiotic E.coli Nissle 1917. The vector pMPES (Modular Peptide Expression System) employs the Microcin V (MccV) secretion system and a powerful synthetic promoter to drive AMP production. Herein, we demonstrate the capacity of pMPES to produce inhibitory levels of MccV, Microcin L (MccL), Microcin N (McnN), Enterocin A (EntA), Enterocin P (EntP), Hiracin JM79 (HirJM79) and Enterocin B (EntB). To our knowledge, this is the first demonstration of such a broadly-applicable secretion system for AMP production. This type of modular expression system could expedite the development of sorely needed antimicrobial technologies.
Collapse
|
96
|
Cano-Garrido O, Sánchez-Chardi A, Parés S, Giró I, Tatkiewicz WI, Ferrer-Miralles N, Ratera I, Natalello A, Cubarsi R, Veciana J, Bach À, Villaverde A, Arís A, Garcia-Fruitós E. Functional protein-based nanomaterial produced in microorganisms recognized as safe: A new platform for biotechnology. Acta Biomater 2016; 43:230-239. [PMID: 27452157 DOI: 10.1016/j.actbio.2016.07.038] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/12/2016] [Accepted: 07/20/2016] [Indexed: 12/18/2022]
Abstract
UNLABELLED Inclusion bodies (IBs) are protein-based nanoparticles formed in Escherichia coli through stereospecific aggregation processes during the overexpression of recombinant proteins. In the last years, it has been shown that IBs can be used as nanostructured biomaterials to stimulate mammalian cell attachment, proliferation, and differentiation. In addition, these nanoparticles have also been explored as natural delivery systems for protein replacement therapies. Although the production of these protein-based nanomaterials in E. coli is economically viable, important safety concerns related to the presence of endotoxins in the products derived from this microorganism need to be addressed. Lactic acid bacteria (LAB) are a group of food-grade microorganisms that have been classified as safe by biologically regulatory agencies. In this context, we have demonstrated herein, for the first time, the production of fully functional, IB-like protein nanoparticles in LAB. These nanoparticles have been fully characterized using a wide range of techniques, including field emission scanning electron microscopy (FESEM), transmission electron microscopy (TEM), dynamic light scattering (DLS), Fourier transform infrared (FTIR) spectroscopy, zymography, cytometry, confocal microscopy, and wettability and cell coverage measurements. Our results allow us to conclude that these materials share the main physico-chemical characteristics with IBs from E. coli and moreover are devoid of any harmful endotoxin contaminant. These findings reveal a new platform for the production of protein-based safe products with high pharmaceutical interest. STATEMENT OF SIGNIFICANCE The development of both natural and synthetic biomaterials for biomedical applications is a field in constant development. In this context, E. coli is a bacteria that has been widely studied for its ability to naturally produce functional biomaterials with broad biomedical uses. Despite being effective, products derived from this species contain membrane residues able to trigger a non-desired immunogenic responses. Accordingly, exploring alternative bacteria able to synthesize such biomaterials in a safe molecular environment is becoming a challenge. Thus, the present study describes a new type of functional protein-based nanomaterial free of toxic contaminants with a wide range of applications in both human and veterinary medicine.
Collapse
|
97
|
Cano-Garrido O, Céspedes MV, Unzueta U, Saccardo P, Roldán M, Sánchez-Chardi A, Cubarsi R, Vázquez E, Mangues R, García-Fruitós E, Villaverde A. CXCR4(+)-targeted protein nanoparticles produced in the food-grade bacterium Lactococcus lactis. Nanomedicine (Lond) 2016; 11:2387-98. [PMID: 27529439 DOI: 10.2217/nnm-2016-0200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIM Lactococcus lactis is a Gram-positive (endotoxin-free) food-grade bacteria exploited as alternative to Escherichia coli for recombinant protein production. We have explored here for the first time the ability of this platform as producer of complex, self-assembling protein materials. MATERIALS & METHODS Biophysical properties, cell penetrability and in vivo biodistribution upon systemic administration of tumor-targeted protein nanoparticles produced in L. lactis have been compared with the equivalent material produced in E. coli. RESULTS Protein nanoparticles have been efficiently produced in L. lactis, showing the desired size, internalization properties and biodistribution. CONCLUSION In vitro and in vivo data confirm the potential and robustness of the production platform, pointing out L. lactis as a fascinating cell factory for the biofabrication of protein materials intended for therapeutic applications.
Collapse
Affiliation(s)
- Olivia Cano-Garrido
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Cerdanyola del Vallès, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - María Virtudes Céspedes
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193 Cerdanyola del Vallès, Spain.,Oncogenesis & Antitumor Drug Group, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu I Sant Pau, 08025 Barcelona, Spain
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193 Cerdanyola del Vallès, Spain.,Oncogenesis & Antitumor Drug Group, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu I Sant Pau, 08025 Barcelona, Spain
| | - Paolo Saccardo
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - Mònica Roldán
- Servei de Microscòpia, Universitat Autònoma de Barcelona, Bellaterra 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Alejandro Sánchez-Chardi
- Servei de Microscòpia, Universitat Autònoma de Barcelona, Bellaterra 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Rafael Cubarsi
- Departament de Matemàtica Aplicada IV. Universitat Politècnica de Catalunya. Jordi Girona 1-3. 08034 Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Cerdanyola del Vallès, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193 Cerdanyola del Vallès, Spain
| | - Ramon Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193 Cerdanyola del Vallès, Spain.,Oncogenesis & Antitumor Drug Group, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu I Sant Pau, 08025 Barcelona, Spain
| | - Elena García-Fruitós
- Department of Ruminant Production, Institut de Recerca i Tecnologia Agroalimentàries (IRTA), 08140 Caldes de Montbui, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Cerdanyola del Vallès, Spain.,Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Cerdanyola del Vallès, Spain.,CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
98
|
KHODER GHALIA, AL-MENHALI ASMAA, AL-YASSIR FARAH, KARAM SHERIFM. Potential role of probiotics in the management of gastric ulcer. Exp Ther Med 2016; 12:3-17. [PMID: 27347010 PMCID: PMC4906699 DOI: 10.3892/etm.2016.3293] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 03/03/2016] [Indexed: 02/07/2023] Open
Abstract
Gastric ulcer is one of the most common chronic gastrointestinal diseases characterized by a significant defect in the mucosal barrier. Helicobacter pylori (H. pylori) infection and the frequent long-term use of non-steroidal anti-inflammatory drugs are major factors involved in gastric ulcer development. Acid inhibitors and antibiotics are commonly used to treat gastric ulcer. However, in the last few decades, the accumulating evidence for resistance to antibiotics and the side effects of antibiotics and acid inhibitors have drawn attention to the possible use of probiotics in the prevention and treatment of gastric ulcer. Probiotics are live microorganisms that when administered in adequate amounts confer health benefits on the host. Currently, the available experimental and clinical studies indicate that probiotics are promising for future applications in the management of gastric ulcers. This review aims to provide an overview of the general health benefits of probiotics on various systemic and gastrointestinal disorders with a special focus on gastric ulcer and the involved cellular and molecular mechanisms: i) Protection of gastric mucosal barrier; ii) upregulation of prostaglandins, mucus, growth factors and anti-inflammatory cytokines; iii) increased cell proliferation to apoptosis ratio; and iv) induction of angiogenesis. Finally, some of the available data on the possible use of probiotics in H. pylori eradication are discussed.
Collapse
Affiliation(s)
- GHALIA KHODER
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - ASMA A. AL-MENHALI
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 17666, United Arab Emirates
| | - FARAH AL-YASSIR
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates
| | - SHERIF M. KARAM
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates
| |
Collapse
|
99
|
Zhang B, Li A, Zuo F, Yu R, Zeng Z, Ma H, Chen S. Recombinant Lactococcus lactis NZ9000 secretes a bioactive kisspeptin that inhibits proliferation and migration of human colon carcinoma HT-29 cells. Microb Cell Fact 2016; 15:102. [PMID: 27287327 PMCID: PMC4901401 DOI: 10.1186/s12934-016-0506-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/01/2016] [Indexed: 01/14/2023] Open
Abstract
Background Proteinaceous bioactive substances and pharmaceuticals are most conveniently administered orally. However, the facing problems are the side effects of proteolytic degradation and denaturation in the gastrointestinal tract. In recent years, lactic acid bacteria (LAB) have been verified to be a promising delivery vector for susceptible functional proteins and drugs. KiSS-1 peptide, a cancer suppressor, plays a critical role in inhibiting cancer metastasis and its activity has been confirmed by direct administration. However, whether this peptide can be functionally expressed in LAB and exert activity on cancer cells, thus providing a potential alternative administration manner in the future, has not been demonstrated. Results A recombinant Lactococcus lactis strain NZ9000-401-kiss1 harboring a plasmid containing the gene of the tumor metastasis-inhibiting peptide KiSS1 was constructed. After optimization of the nisin induction conditions, the recombinant strain efficiently secreted KiSS1 with a maximum detectable amount of 27.9 μg/ml in Dulbecco’s Modified Eagle medium. The 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide and would healing assays, respectively, indicated that the secreted KiSS1 peptide remarkably inhibited HT-29 cell proliferation and migration. Furthermore, the expressed KiSS1 was shown to induce HT-29 cell morphological changes, apoptosis and reduce the expression of matrix metalloproteinase 9 (MMP-9) at both mRNA and protein levels. Conclusions A recombinant L. lactis NZ9000-401-kiss1 successfully expressing the human kiss1 was constructed. The secreted KiSS1 peptide inhibited human HT-29 cells’ proliferation and migration probably by invoking, or mediating the cell-apoptosis pathway and by down regulating MMP-9 expression, respectively. Our results suggest that L. lactis is an ideal cell factory for secretory expression of tumor metastasis-inhibiting peptide KiSS1, and the KiSS1-producing L. lactis strain may serve as a new tool for cancer therapy in the future.
Collapse
Affiliation(s)
- Bo Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing, 100083, People's Republic of China.,Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
| | - Angdi Li
- Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
| | - Fanglei Zuo
- Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
| | - Rui Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing, 100083, People's Republic of China.,Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
| | - Zhu Zeng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing, 100083, People's Republic of China.,Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
| | - Huiqin Ma
- College of Horticulture, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Shangwu Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing, 100083, People's Republic of China. .,Key Laboratory of Functional Dairy, Department of Food Science and Engineering, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China.
| |
Collapse
|
100
|
The effects of maternal and post-weaning diet interaction on glucose metabolism and gut microbiota in male mice offspring. Biosci Rep 2016; 36:BSR20160103. [PMID: 27129301 PMCID: PMC5293570 DOI: 10.1042/bsr20160103] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/27/2016] [Indexed: 01/21/2023] Open
Abstract
Substantial studies demonstrated that maternal nutrition can significantly determine the susceptibility to developing some metabolic diseases in offspring. However, investigations into the later-life effects of these diets on gut microbiota in the offspring are limited. Our objective was to explore the effects of maternal and post-weaning diet interaction on offspring's gut microbiota and glucose metabolism in later life. The male offspring of dams fed on either a high-fat (HF) diet or control (C) diet and then weaned to either a HF or C diet, generating four groups: C-C, HF-C, C-HF and HF-HF (n=8 in each group). The C-C offspring had lower body weight than C-HF group at 16 weeks of age (P<0.01) and both C-HF and HF-HF offspring had higher body weight than C-C group at 24 and 32 weeks of age (P<0.001 respectively). The blood glucose (BG) levels of the male offspring from the C and HF dams weaned HF diet were significantly higher at 30 min, 60 min and 120 min (P<0.001) after intraperitoneal glucose administration compared with those of the C-C group. The C-HF group had higher BG at 30 min than HF-HF group (P<0.01). Furthermore, area under the curve (AUC) in C-HF and HF-HF groups was also significantly larger than C-C group (P<0.001). Fasting BG and homoeostasis model assessment of insulin resistance (HOMA-IR) of the offspring were significantly higher in C-HF and HF-HF groups than C-C group at 32 weeks of age (P<0.05). Operational taxonomic unit (OTU), Chao and Shannon indexes showed a significantly lower diversity in C-HF offspring compared with HF-C and C-C groups (P<0.05). The dominant phyla of all the groups were Bacteroidetes, Firmicutes and Proteobacteria, which also showed significantly different percentages in the group (P<0.05). Furthermore, it is indicated that Lactobacillus and Bacteroides were significantly associated with glucose response to a glucose load (P<0.05). In conclusion, maternal and post-weaning diet interaction predisposes the offspring to aberrant glucose metabolism and alterations of gut microbiota in later life. Our study is novel in focusing on the effects of maternal and post-weaning diet interaction on offspring gut microbiota and glucose metabolism in later life.
Collapse
|