1
|
Wiull K, Holmgren E, Svensson S, Eijsink VGH, Grönlund H, Mathiesen G. Surface display of two neoantigens on Lactiplantibacillus plantarum. Sci Rep 2025; 15:7769. [PMID: 40044906 PMCID: PMC11882890 DOI: 10.1038/s41598-025-91180-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
Lactic acid bacteria, such as Lactiplantibacillus plantarum, are becoming increasingly popular hosts for combining production and delivery of therapeutic proteins to immune cells. Soluble antigens are susceptible to rapid proteolysis, hence anchoring of antigens to bacterial cells, which likely protects the antigen, is a preferred delivery strategy that may increase immune responses. In cancer research, personalized immunotherapy has high potential and, in this respect, the so-called neoantigens that accumulate in tumor cells are promising tumor specific targets. Here, we demonstrate that, when using the inducible pSIP expression system, L. plantarum can produce and surface display two neoantigens, NAG1 and ETV6. The antigens could be targeted to both the cell membrane and the cell wall, utilizing four different anchoring methods. The production level and the degree of surface exposure of the antigens varied among the anchors. Flow cytometry analysis showed that antigens anchored to the cell wall were more exposed than those anchored to the cell membrane. To our knowledge these are the first reports of neoantigens being produced and surface-displayed in Lactobacillales.
Collapse
Affiliation(s)
- Kamilla Wiull
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway.
| | - Erik Holmgren
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
| | - Hans Grönlund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- NEOGAP Therapeutics AB, Stockholm, Sweden
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway.
| |
Collapse
|
2
|
Zahirović A, Berlec A. Targeting IL-6 by engineered Lactococcus lactis via surface-displayed affibody. Microb Cell Fact 2022; 21:143. [PMID: 35842694 PMCID: PMC9287920 DOI: 10.1186/s12934-022-01873-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dysregulated production of interleukin (IL)-6 is implicated in the pathology of inflammatory bowel disease (IBD). Neutralization of IL-6 in the gut by safe probiotic bacteria may help alleviate intestinal inflammation. Here, we developed Lactococcus lactis with potent and selective IL-6 binding activity by displaying IL-6-specific affibody on its surface. RESULTS Anti-IL-6 affibody (designated as ZIL) was expressed in fusion with lactococcal secretion peptide Usp45 and anchoring protein AcmA. A high amount of ZIL fusion protein was detected on bacterial surface, and its functionality was validated by confocal microscopy and flow cytometry. Removal of IL-6 from the surrounding medium by the engineered L. lactis was evaluated using enzyme-linked immunosorbent assay. ZIL-displaying L. lactis sequestered recombinant human IL-6 from the solution in a concentration-dependent manner by up to 99% and showed no binding to other pro-inflammatory cytokines, thus proving to be highly specific for IL-6. The removal was equally efficient across different IL-6 concentrations (150-1200 pg/mL) that were found to be clinically relevant in IBD patients. The ability of engineered bacteria to capture IL-6 from cell culture supernatant was assessed using immunostimulated human monocytic cell lines (THP-1 and U-937) differentiated into macrophage-like cells. ZIL-displaying L. lactis reduced the content of IL-6 in the supernatants of both cell lines in a concentration-dependent manner by up to 94%. Dose response analysis showed that bacterial cell concentrations of 107 and 109 CFU/mL (colony forming units per mL) were required for half-maximal removal of recombinant and macrophage-derived IL-6, respectively. CONCLUSION The ability of ZIL-displaying L. lactis to bind pathological concentrations of IL-6 at common bacterial doses suggests physiological significance.
Collapse
Affiliation(s)
- Abida Zahirović
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia. .,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
3
|
Kazi TA, Acharya A, Mukhopadhyay BC, Mandal S, Arukha AP, Nayak S, Biswas SR. Plasmid-Based Gene Expression Systems for Lactic Acid Bacteria: A Review. Microorganisms 2022; 10:1132. [PMID: 35744650 PMCID: PMC9229153 DOI: 10.3390/microorganisms10061132] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 01/27/2023] Open
Abstract
Lactic acid bacteria (LAB) play a very vital role in food production, preservation, and as probiotic agents. Some of these species can colonize and survive longer in the gastrointestinal tract (GIT), where their presence is crucially helpful to promote human health. LAB has also been used as a safe and efficient incubator to produce proteins of interest. With the advent of genetic engineering, recombinant LAB have been effectively employed as vectors for delivering therapeutic molecules to mucosal tissues of the oral, nasal, and vaginal tracks and for shuttling therapeutics for diabetes, cancer, viral infections, and several gastrointestinal infections. The most important tool needed to develop genetically engineered LABs to produce proteins of interest is a plasmid-based gene expression system. To date, a handful of constitutive and inducible vectors for LAB have been developed, but their limited availability, host specificity, instability, and low carrying capacity have narrowed their spectrum of applications. The current review discusses the plasmid-based vectors that have been developed so far for LAB; their functionality, potency, and constraints; and further highlights the need for a new, more stable, and effective gene expression platform for LAB.
Collapse
Affiliation(s)
- Tawsif Ahmed Kazi
- Department of Botany, Visva-Bharati University, Santiniketan 731235, West Bengal, India; (T.A.K.); (A.A.); (B.C.M.)
| | - Aparupa Acharya
- Department of Botany, Visva-Bharati University, Santiniketan 731235, West Bengal, India; (T.A.K.); (A.A.); (B.C.M.)
| | - Bidhan Chandra Mukhopadhyay
- Department of Botany, Visva-Bharati University, Santiniketan 731235, West Bengal, India; (T.A.K.); (A.A.); (B.C.M.)
| | - Sukhendu Mandal
- Laboratory of Molecular Bacteriology, Department of Microbiology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India;
| | - Ananta Prasad Arukha
- Researcher 5 Department of Neurosurgery, Medical School, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Subhendu Nayak
- Sr. Scientist, Clorox, Better Health VMS, Durham, NC 27701, USA;
| | - Swadesh Ranjan Biswas
- Department of Botany, Visva-Bharati University, Santiniketan 731235, West Bengal, India; (T.A.K.); (A.A.); (B.C.M.)
| |
Collapse
|
4
|
Plasmid Replicons for the Production of Pharmaceutical-Grade pDNA, Proteins and Antigens by Lactococcus lactis Cell Factories. Int J Mol Sci 2021; 22:ijms22031379. [PMID: 33573129 PMCID: PMC7866527 DOI: 10.3390/ijms22031379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
The Lactococcus lactis bacterium found in different natural environments is traditionally associated with the fermented food industry. But recently, its applications have been spreading to the pharmaceutical industry, which has exploited its probiotic characteristics and is moving towards its use as cell factories for the production of added-value recombinant proteins and plasmid DNA (pDNA) for DNA vaccination, as a safer and industrially profitable alternative to the traditional Escherichia coli host. Additionally, due to its food-grade and generally recognized safe status, there have been an increasing number of studies about its use in live mucosal vaccination. In this review, we critically systematize the plasmid replicons available for the production of pharmaceutical-grade pDNA and recombinant proteins by L. lactis. A plasmid vector is an easily customized component when the goal is to engineer bacteria in order to produce a heterologous compound in industrially significant amounts, as an alternative to genomic DNA modifications. The additional burden to the cell depends on plasmid copy number and on the expression level, targeting location and type of protein expressed. For live mucosal vaccination applications, besides the presence of the necessary regulatory sequences, it is imperative that cells produce the antigen of interest in sufficient yields. The cell wall anchored antigens had shown more promising results in live mucosal vaccination studies, when compared with intracellular or secreted antigens. On the other side, engineering L. lactis to express membrane proteins, especially if they have a eukaryotic background, increases the overall cellular burden. The different alternative replicons for live mucosal vaccination, using L. lactis as the DNA vaccine carrier or the antigen producer, are critically reviewed, as a starting platform to choose or engineer the best vector for each application.
Collapse
|
5
|
Plavec TV, Zahirović A, Zadravec P, Sabotič J, Berlec A. Lectin-Mediated Binding of Engineered Lactococcus lactis to Cancer Cells. Microorganisms 2021; 9:microorganisms9020223. [PMID: 33499141 PMCID: PMC7911926 DOI: 10.3390/microorganisms9020223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/28/2022] Open
Abstract
Lectins have been increasingly utilized as carriers for targeted drug delivery based on their specific binding to glycans located on mammalian cells. This study employed two lectins, B subunit of bacterial Shiga holotoxin (Stx1B) and fungal Clitocybe nebularis lectin (CNL), for surface display on the lactic acid bacterium Lactococcus lactis. The specific adhesion of these engineered, lectin-displaying L. lactis to cancer cells was evaluated. The expression and surface display of both lectins on L. lactis were demonstrated by western blotting and flow cytometry, respectively. MTS assays revealed that recombinant Stx1B had no effect on Caco-2 cell viability at concentrations of ≤25 µg/mL, whereas CNL was non-toxic even at relatively high concentrations of ≤250 µg/mL. Stx1B bound to Caco-2, HT-29 and HeLa cells after 1 h of incubation. CNL bound to Caco-2 cells and recognized several glycoproteins in HT-29 and Caco-2 cell homogenates of which a 70 kDa protein predominated. Confocal microscopy revealed adhesion of Stx1B-displaying L. lactis to HeLa, Caco-2, and, to a lesser extent, HT-29 cells; CNL-displaying L. lactis showed a relatively similar level of adherence to HT-29 and Caco-2 cells. Thus, lectin-displaying L. lactis might serve as a carrier in targeted drug delivery when coupled to a therapeutic moiety.
Collapse
Affiliation(s)
- Tina Vida Plavec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Abida Zahirović
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
| | - Petra Zadravec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
- Lek d.d., Kolodvorska 27, 1234 Mengeš, Slovenia
| | - Jerica Sabotič
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; (T.V.P.); (A.Z.); (P.Z.); (J.S.)
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
6
|
Cell immunocapture microfluidic chip based on high-affinity recombinant protein binders. Biosens Bioelectron 2021; 172:112784. [PMID: 33161292 DOI: 10.1016/j.bios.2020.112784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/30/2020] [Accepted: 10/30/2020] [Indexed: 12/29/2022]
Abstract
Cell immunocapture microfluidic devices represent a rapidly developing field with many potential applications in medical diagnostics. The core of such approach lies in the cell binding to antibody coated surfaces through their surface receptors. Here we show, that the small recombinant protein binders (PBs) can be used for this purpose as well, with the advantage of their constructional flexibility, possibility of fusion with range of tags and cheap mass production. For this purpose, two different PBs derived from Albumin Binding Domain (ABDwt) of streptococcal protein G, so called REX and ARS ligands with proved high affinity and selectivity to the human interleukin-23 (IL-23R) and IL-17 receptor A were used. Four PBs variants recognizing two different epitopes on two different receptors and two PBs variants binding to the same epitope on one receptor but having different peptide spacer with Avitag sequence necessary for their immobilization on sensor surface were tested for cell-capture efficiency. The glass microfluidic Y-system with planar immunocapture channel working in so-called stop-flow dynamic regime was designed. Up to 60-74% immunocapture efficiency of model THP-1 cells on REX/ARS surfaces and practically no cell binding on control ABDwt surfaces was achieved. Moreover, the specific immunocapture of THP-1 cells from mixture with IL-17RA negative DU-145 cells was demonstrated. We discuss the role of the epitope, affinity and immobilization spacer of PBs as well as the influence of stop-flow dynamic regime on the effectivity of THP-1 cell immunocapture. Results can be further exploited in design of microfluidic devices for rare cells immunocapture.
Collapse
|
7
|
Mühlen S, Dersch P. Treatment Strategies for Infections With Shiga Toxin-Producing Escherichia coli. Front Cell Infect Microbiol 2020; 10:169. [PMID: 32435624 PMCID: PMC7218068 DOI: 10.3389/fcimb.2020.00169] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/31/2020] [Indexed: 01/07/2023] Open
Abstract
Infections with Shiga toxin-producing Escherichia coli (STEC) cause outbreaks of severe diarrheal disease in children and the elderly around the world. The severe complications associated with toxin production and release range from bloody diarrhea and hemorrhagic colitis to hemolytic-uremic syndrome, kidney failure, and neurological issues. As the use of antibiotics for treatment of the infection has long been controversial due to reports that antibiotics may increase the production of Shiga toxin, the recommended therapy today is mainly supportive. In recent years, a variety of alternative treatment approaches such as monoclonal antibodies or antisera directed against Shiga toxin, toxin receptor analogs, and several vaccination strategies have been developed and evaluated in vitro and in animal models. A few strategies have progressed to the clinical trial phase. Here, we review the current understanding of and the progress made in the development of treatment options against STEC infections and discuss their potential.
Collapse
Affiliation(s)
- Sabrina Mühlen
- Institute for Infectiology, University of Münster, Münster, Germany.,German Center for Infection Research (DZIF), Associated Site University of Münster, Münster, Germany
| | - Petra Dersch
- Institute for Infectiology, University of Münster, Münster, Germany.,German Center for Infection Research (DZIF), Associated Site University of Münster, Münster, Germany
| |
Collapse
|
8
|
Cho SW, Yim J, Seo SW. Engineering Tools for the Development of Recombinant Lactic Acid Bacteria. Biotechnol J 2020; 15:e1900344. [DOI: 10.1002/biot.201900344] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/27/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Sung Won Cho
- School of Chemical and Biological EngineeringInstitute of Chemical ProcessesSeoul National University 1 Gwanak‐ro Gwanak‐gu Seoul 08826 Republic of Korea
| | - Jaewoo Yim
- School of Chemical and Biological EngineeringInstitute of Chemical ProcessesSeoul National University 1 Gwanak‐ro Gwanak‐gu Seoul 08826 Republic of Korea
| | - Sang Woo Seo
- School of Chemical and Biological EngineeringInstitute of Chemical ProcessesSeoul National University 1 Gwanak‐ro Gwanak‐gu Seoul 08826 Republic of Korea
| |
Collapse
|
9
|
Petroková H, Mašek J, Kuchař M, Vítečková Wünschová A, Štikarová J, Bartheldyová E, Kulich P, Hubatka F, Kotouček J, Turánek Knotigová P, Vohlídalová E, Héžová R, Mašková E, Macaulay S, Dyr JE, Raška M, Mikulík R, Malý P, Turánek J. Targeting Human Thrombus by Liposomes Modified with Anti-Fibrin Protein Binders. Pharmaceutics 2019; 11:pharmaceutics11120642. [PMID: 31810280 PMCID: PMC6955937 DOI: 10.3390/pharmaceutics11120642] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 12/22/2022] Open
Abstract
Development of tools for direct thrombus imaging represents a key step for diagnosis and treatment of stroke. Nanoliposomal carriers of contrast agents and thrombolytics can be functionalized to target blood thrombi by small protein binders with selectivity for fibrin domains uniquely formed on insoluble fibrin. We employed a highly complex combinatorial library derived from scaffold of 46 amino acid albumin-binding domain (ABD) of streptococcal protein G, and ribosome display, to identify variants recognizing fibrin cloth in human thrombus. We constructed a recombinant target as a stretch of three identical fibrin fragments of 16 amino acid peptide of the Bβ chain fused to TolA protein. Ribosome display selection followed by large-scale Enzyme-Linked ImmunoSorbent Assay (ELISA) screening provided four protein variants preferentially binding to insoluble form of human fibrin. The most specific binder variant D7 was further modified by C-terminal FLAG/His-Tag or double His-tag for the attachment onto the surface of nanoliposomes via metallochelating bond. D7-His-nanoliposomes were tested using in vitro flow model of coronary artery and their binding to fibrin fibers was demonstrated by confocal and electron microscopy. Thus, we present here the concept of fibrin-targeted binders as a platform for functionalization of nanoliposomes in the development of advanced imaging tools and future theranostics.
Collapse
Affiliation(s)
- Hana Petroková
- Laboratory of Ligand Engineering, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic; (H.P.); (M.K.)
| | - Josef Mašek
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Milan Kuchař
- Laboratory of Ligand Engineering, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic; (H.P.); (M.K.)
| | - Andrea Vítečková Wünschová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Jana Štikarová
- Department of Biochemistry, Institute of Hematology and Blood Transfusion, U nemocnice 2094/1, 128 20 Praha 2, Czech Republic; (J.Š.); (J.E.D.)
| | - Eliška Bartheldyová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Pavel Kulich
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - František Hubatka
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Jan Kotouček
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Pavlína Turánek Knotigová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Eva Vohlídalová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Renata Héžová
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Eliška Mašková
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
| | - Stuart Macaulay
- Malvern Instruments Ltd., Enigma Business Park, Grove Lane, Malvern WR14 1XZ, UK;
| | - Jan Evangelista Dyr
- Department of Biochemistry, Institute of Hematology and Blood Transfusion, U nemocnice 2094/1, 128 20 Praha 2, Czech Republic; (J.Š.); (J.E.D.)
| | - Milan Raška
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hněvotínská 3, 775 15 Olomouc, Czech Republic
| | - Robert Mikulík
- The International Clinical Research Center ICRC and Neurology Department of St. Anne’s University Hospital in Brno, Pekařská 53, 656 91 Brno, Czech Republic;
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology, Czech Academy of Sciences, v.v.i., BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic; (H.P.); (M.K.)
- Correspondence: (P.M.); (J.T.); Tel.: +420-325-873-763 (P.M.); +420-732-813-577 (J.T.)
| | - Jaroslav Turánek
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, v.v.i., Hudcova 70, 621 00 Brno, Czech Republic; (J.M.); (A.V.W.); (E.B.); (P.K.); (F.H.); (J.K.); (P.T.K.); (E.V.); (R.H.); (E.M.)
- Correspondence: (P.M.); (J.T.); Tel.: +420-325-873-763 (P.M.); +420-732-813-577 (J.T.)
| |
Collapse
|
10
|
Sreerohini S, Balakrishna K, Parida M. Oral immunization of mice with Lactococcus lactis expressing Shiga toxin truncate confers enhanced protection against Shiga toxins of Escherichia coli O157:H7 and Shigella dysenteriae. APMIS 2019; 127:671-680. [PMID: 31344276 DOI: 10.1111/apm.12983] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/11/2019] [Indexed: 12/25/2022]
Abstract
Regardless of the communal impact of Shiga toxins, till today neither a specific treatment nor licensed vaccine is available. Lactococcus lactis (L. lactis), generally regarded as safe organism, is well known to provide a valuable approach regarding the oral delivery of vaccines. This study was undertaken to evaluate the protective efficacy of Stx2a1 expressed in nisin-inducible L. lactis, against Shiga toxins (Stx1, Stx2) in mouse model. Oral immunization of BALB/c mice with LL-Stx2a1 elicited significant serum antibody titer with elevated fecal and serum IgA, along with minimized intestinal and kidney damage resulting in survival of immunized animals at 84% and 100% when challenged with 10 × LD50 of Escherichia coli O157 and Shigella dysenteriae toxins, respectively. HeLa cells incubated with immune sera and toxin mixture revealed high neutralizing capacity with 90% cell survivability against both the toxins. Mice immunized passively with both toxins and antibody mixture survived the observation period of 15 days, and the controls administered with sham sera and toxins were succumbed to death within 3 days. Our results revealed protective efficacy and toxin neutralization ability of LL-Stx2a1, proposing it as an oral vaccine candidate against Shiga toxicity mediated by E. coli O157 and S. dysenteriae.
Collapse
Affiliation(s)
- Sagi Sreerohini
- Division of Food Microbiology, Defence Food Research Laboratory, Mysore, India
| | - Konduru Balakrishna
- Division of Food Microbiology, Defence Food Research Laboratory, Mysore, India
| | - Manmohan Parida
- Division of Food Microbiology, Defence Food Research Laboratory, Mysore, India
| |
Collapse
|
11
|
Engineered Lactococcus lactis Secreting IL-23 Receptor-Targeted REX Protein Blockers for Modulation of IL-23/Th17-Mediated Inflammation. Microorganisms 2019; 7:microorganisms7050152. [PMID: 31137908 PMCID: PMC6560508 DOI: 10.3390/microorganisms7050152] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/09/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
Lactococcus lactis, a probiotic bacterium of food origin, has recently been demonstrated as a suitable strain for the production and in vivo delivery of therapeutically important proteins into the gut. We aimed to engineer recombinant L. lactis cells producing/secreting REX binding proteins that have been described as IL-23 receptor (IL-23R) blockers and IL-23R antagonists suppressing the secretion of cytokine IL-17A, a pivotal step in the T-helper Th17-mediated pro-inflammatory cascade, as well as in the development of autoimmune diseases, including inflammatory bowel disease (IBD). To reach this goal, we introduced cDNA sequences coding for REX009, REX115, and REX125 proteins into plasmid vectors carrying a Usp45 secretion signal, a FLAG tag sequence consensus, and a LysM-containing cA surface anchor (AcmA), thus allowing cell-surface peptidoglycan anchoring. These plasmids, or their non-FLAG/non-AcmA versions, were introduced into L. lactis host cells, thus generating unique recombinant L. lactis-REX strains. We demonstrate that all three REX proteins are expressed in L. lactis cells and are efficiently displayed on the bacterial surface, as tested by flow cytometry using an anti-FLAG antibody conjugate. Upon 10-fold concentration of the conditioned media, a REX125 secretory variant can be detected by Western blotting. To confirm that the FLAG/non-FLAG REX proteins displayed by L. lactis retain their binding specificity, cell-surface interactions of REX proteins with an IL-23R-IgG chimera were demonstrated by flow cytometry. In addition, statistically significant binding of secreted REX009 and REX115 proteins to bacterially produced, soluble human IL-23R was confirmed by ELISA. We conclude that REX-secreting L. lactis strains were engineered that might serve as IL-23/IL-23R blockers in an experimentally induced mouse model of colitis.
Collapse
|
12
|
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: 6.2] [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]
|
13
|
Li R, Kang G, Hu M, Huang H. Ribosome Display: A Potent Display Technology used for Selecting and Evolving Specific Binders with Desired Properties. Mol Biotechnol 2018; 61:60-71. [DOI: 10.1007/s12033-018-0133-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
14
|
Ronzetti M, Baljinnyam B, Yasgar A, Simeonov A. Testing for drug-human serum albumin binding using fluorescent probes and other methods. Expert Opin Drug Discov 2018; 13:1005-1014. [PMID: 30320522 PMCID: PMC11369766 DOI: 10.1080/17460441.2018.1534824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Drug plasma protein binding remains highly relevant to research and drug development, making the assessment and profiling of compound affinity to plasma proteins essential to drug discovery efforts. Although there are a number of fully-characterized methods, they lack the throughput to handle large numbers of compounds. As the evaluation of adsorption, distribution, metabolism, and excretion is addressed earlier in the drug development timeline, the need for higher-throughput methods has grown. Areas Covered: This review will highlight recent developments on methods for profiling drug plasma binding, with an emphasis on fluorescent probes and emerging high-throughput methodologies. Expert Opinion: There have been a number of high-throughput assays developed in recent years to meet the scaled up demands for compound profiling. Ultimately, the selection of assay technology relies on a number of factors, such as capabilities of the laboratory and the breadth and amount of data required. Fluorescent probe displacement assays are highly flexible and amenable to high-throughput screening, easily scaling up to handle large compound libraries. Recent developments in fluorescence technologies, such as homogenous time-resolved fluorescence and probes utilizing the aggregation-induced emission effect, have improved the sensitivity of these assays. Other technologies, such as microscale thermophoresis and quantitative structure-activity relationship modeling, are gaining popularity as alternative techniques for drug plasma protein binding characterization.
Collapse
Affiliation(s)
- Michael Ronzetti
- a National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland , USA
| | - Bolormaa Baljinnyam
- a National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland , USA
| | - Adam Yasgar
- a National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland , USA
| | - Anton Simeonov
- a National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland , USA
| |
Collapse
|
15
|
ABD-Derived Protein Blockers of Human IL-17 Receptor A as Non-IgG Alternatives for Modulation of IL-17-Dependent Pro-Inflammatory Axis. Int J Mol Sci 2018; 19:ijms19103089. [PMID: 30304852 PMCID: PMC6213189 DOI: 10.3390/ijms19103089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/04/2018] [Accepted: 10/06/2018] [Indexed: 12/14/2022] Open
Abstract
Interleukin 17 (IL-17) and its cognate receptor A (IL-17RA) play a crucial role in Th17 cells-mediated pro-inflammatory pathway and pathogenesis of several autoimmune disorders including psoriasis. IL-17 is mainly produced by activated Th-17 helper cells upon stimulation by IL-23 and, via binding to its receptors, mediates IL-17-driven cell signaling in keratinocytes. Hyper-proliferation of keratinocytes belongs to major clinical manifestations in psoriasis. To modulate IL-17-mediated inflammatory cascade, we generated a unique collection of IL-17RA-targeting protein binders that prevent from binding of human IL-17A cytokine to its cell-surface receptor. To this goal, we used a highly complex combinatorial library derived from scaffold of albumin-binding domain (ABD) of streptococcal protein G, and ribosome display selection, to yield a collection of ABD-derived high-affinity ligands of human IL-17RA, called ARS binders. From 67 analyzed ABD variants, 7 different sequence families were identified. Representatives of these groups competed with human IL-17A for binding to recombinant IL-17RA receptor as well as to IL-17RA-Immunoglobulin G chimera, as tested in enzyme-linked immunosorbent assay (ELISA). Five ARS variants bound to IL-17RA-expressing THP-1 cells and blocked binding of human IL-17 cytokine to the cell surface, as tested by flow cytometry. Three variants exhibited high-affinity binding with a nanomolar Kd value to human keratinocyte HaCaT cells, as measured using Ligand Tracer Green Line. Upon IL-17-stimulated activation, ARS variants inhibited secretion of Gro-α (CXCL1) by normal human skin fibroblasts in vitro. Thus, we identified a novel class of inhibitory ligands that might serve as immunosuppressive IL-17RA-targeted non-IgG protein antagonists.
Collapse
|
16
|
Duncan SA, Baganizi DR, Sahu R, Singh SR, Dennis VA. SOCS Proteins as Regulators of Inflammatory Responses Induced by Bacterial Infections: A Review. Front Microbiol 2017; 8:2431. [PMID: 29312162 PMCID: PMC5733031 DOI: 10.3389/fmicb.2017.02431] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 11/23/2017] [Indexed: 12/31/2022] Open
Abstract
Severe bacterial infections can lead to both acute and chronic inflammatory conditions. Innate immunity is the first defense mechanism employed against invading bacterial pathogens through the recognition of conserved molecular patterns on bacteria by pattern recognition receptors (PRRs), especially the toll-like receptors (TLRs). TLRs recognize distinct pathogen-associated molecular patterns (PAMPs) that play a critical role in innate immune responses by inducing the expression of several inflammatory genes. Thus, activation of immune cells is regulated by cytokines that use the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway and microbial recognition by TLRs. This system is tightly controlled by various endogenous molecules to allow for an appropriately regulated and safe host immune response to infections. Suppressor of cytokine signaling (SOCS) family of proteins is one of the central regulators of microbial pathogen-induced signaling of cytokines, principally through the inhibition of the activation of JAK/STAT signaling cascades. This review provides recent knowledge regarding the role of SOCS proteins during bacterial infections, with an emphasis on the mechanisms involved in their induction and regulation of antibacterial immune responses. Furthermore, the implication of SOCS proteins in diverse processes of bacteria to escape host defenses and in the outcome of bacterial infections are discussed, as well as the possibilities offered by these proteins for future targeted antimicrobial therapies.
Collapse
Affiliation(s)
- Skyla A Duncan
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Dieudonné R Baganizi
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Rajnish Sahu
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Shree R Singh
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Vida A Dennis
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| |
Collapse
|
17
|
Škrlec K, Pucer Janež A, Rogelj B, Štrukelj B, Berlec A. Evasin-displaying lactic acid bacteria bind different chemokines and neutralize CXCL8 production in Caco-2 cells. Microb Biotechnol 2017; 10:1732-1743. [PMID: 28736998 PMCID: PMC5658612 DOI: 10.1111/1751-7915.12781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/30/2017] [Accepted: 06/05/2017] [Indexed: 01/26/2023] Open
Abstract
Chemokines are key signals in the immune system and play an important role as proinflammatory mediators in the pathology of inflammatory bowel disease and colorectal cancer, making them an important target for therapy. Recombinant lactic acid bacteria (LAB) were engineered to bind CC and CXC chemokines by displaying chemokine‐binding proteins evasin‐1, evasin‐3 and evasin‐4 on their surface. Evasin genes were cloned into lactococcal surface display vector and overexpressed in L. lactis NZ9000 and NZ9000ΔhtrA in fusion with secretion signal and surface anchor. Evasin‐displaying bacteria removed from 15% to 90% of 11 different chemokines from the solution as determined with ELISA and Luminex multiplexing assays, whereby L. lactis NZ9000ΔhtrA proved more efficient. Lactobacillus salivarius ATCC 11741 was coated with L. . lactis‐expressed evasin fusion protein, and its ability to bind chemokines was also confirmed. Evasin‐3‐displaying L. lactis removed 76.0% of IL‐1β‐induced CXCL8 from the supernatant of Caco‐2 epithelial cells. It also prevented secretion of CXCL8 from Caco‐2 cells in a time‐dependent manner when added before induction with IL‐1β. Evasin‐displaying LAB have the ability to bind multiple chemokines simultaneously and exert synergistic activity. This innovative treatment approach therefore has the potential for mucosal therapy of inflammatory bowel disease or colorectal cancer.
Collapse
Affiliation(s)
- Katja Škrlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia.,Graduate School of Biomedicine, Faculty of Medicine, University of Ljubljana, SI-1000, Ljubljana, Slovenia
| | - Anja Pucer Janež
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Rogelj
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia.,Biomedical Research Institute (BRIS), Puhova 10, SI-1000, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000, Ljubljana, Slovenia
| | - Borut Štrukelj
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
18
|
Chua KJ, Kwok WC, Aggarwal N, Sun T, Chang MW. Designer probiotics for the prevention and treatment of human diseases. Curr Opin Chem Biol 2017; 40:8-16. [PMID: 28478369 DOI: 10.1016/j.cbpa.2017.04.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 04/04/2017] [Accepted: 04/13/2017] [Indexed: 12/24/2022]
Abstract
Various studies have shown the beneficial effects of probiotics in humans. The use of synthetic biology to engineer programmable probiotics that specifically targets cancer, infectious agents, or other metabolic diseases has gained much interest since the last decade. Developments made in synthetic probiotics as therapeutics within the last three years will be discussed in this review.
Collapse
Affiliation(s)
- Koon Jiew Chua
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore
| | - Wee Chiew Kwok
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore
| | - Nikhil Aggarwal
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore
| | - Tao Sun
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore
| | - Matthew Wook Chang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore.
| |
Collapse
|