101
|
Michalek J, Hezova R, Turanek-Knötigova P, Gabkova J, Strioga M, Lubitz W, Kudela P. Oncolysate-loaded Escherichia coli bacterial ghosts enhance the stimulatory capacity of human dendritic cells. Cancer Immunol Immunother 2017; 66:149-159. [PMID: 27864613 PMCID: PMC11029152 DOI: 10.1007/s00262-016-1932-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 11/14/2016] [Indexed: 12/15/2022]
Abstract
The natural adjuvant properties of bacterial ghosts (BGs) lie within the presence of intact pathogen-associated molecular patterns on their surface. BGs can improve the direct delivery, natural processing and presentation of target antigens within dendritic cells (DCs). Moreover, sensitization of human DCs by cancer cell lysate (oncolysate)-loaded BGs in the presence of IFN-α and GM-CSF enhanced DC maturation as indicated by an increased expression of maturation markers and co-stimulatory molecules, higher production of IL-12p70 and stimulation of significantly increased proliferation of both autologous CD4+ and CD8+ T cells compared to DCs matured in the presence of purified lipopolysaccharide. The induced T cells efficiently recognized oncolysate-derived tumor-associated antigens expressed by cancer cells used for the production of oncolysate. Our optimized one-step simultaneous antigen delivery and DC maturation-inducing method emerges as a promising tool for the development and implementation of next-generation cellular cancer immunotherapies.
Collapse
Affiliation(s)
- Jaroslav Michalek
- Cellthera, s.r.o., Brno, Czech Republic
- Advanced Cell Immunotherapy Unit, Department of Pharmacology, Medical Faculty, Masaryk University, Brno, Czech Republic
- Department of Pediatrics, The University Hospital Brno, Brno, Czech Republic
| | - Renata Hezova
- Advanced Cell Immunotherapy Unit, Department of Pharmacology, Medical Faculty, Masaryk University, Brno, Czech Republic
| | - Pavlina Turanek-Knötigova
- Advanced Cell Immunotherapy Unit, Department of Pharmacology, Medical Faculty, Masaryk University, Brno, Czech Republic
- Department of Pharmacology and Immunotherapy, Veterinary Research Institute, Brno, Czech Republic
| | - Jana Gabkova
- Institute of Mathematics and Physics, Faculty of Mechanical Engineering, Slovak University of Technology, Bratislava, Slovak Republic
| | - Marius Strioga
- Department of Immunology, National Cancer Institute, Vilnius, Lithuania
| | - Werner Lubitz
- BIRD-C GmbH, Dr. Bohrgasse 2-8, 1030, Vienna, Austria
| | - Pavol Kudela
- BIRD-C GmbH, Dr. Bohrgasse 2-8, 1030, Vienna, Austria.
| |
Collapse
|
102
|
Kassmannhuber J, Rauscher M, Schöner L, Witte A, Lubitz W. Functional display of ice nucleation protein InaZ on the surface of bacterial ghosts. Bioengineered 2017; 8:488-500. [PMID: 28121482 DOI: 10.1080/21655979.2017.1284712] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
In a concept study the ability to induce heterogeneous ice formation by Bacterial Ghosts (BGs) from Escherichia coli carrying ice nucleation protein InaZ from Pseudomonas syringae in their outer membrane was investigated by a droplet-freezing assay of ultra-pure water. As determined by the median freezing temperature and cumulative ice nucleation spectra it could be demonstrated that both the living recombinant E. coli and their corresponding BGs functionally display InaZ on their surface. Under the production conditions chosen both samples belong to type II ice-nucleation particles inducing ice formation at a temperature range of between -5.6 °C and -6.7 °C, respectively. One advantage for the application of such BGs over their living recombinant mother bacteria is that they are non-living native cell envelopes retaining the biophysical properties of ice nucleation and do no longer represent genetically modified organisms (GMOs).
Collapse
Affiliation(s)
- Johannes Kassmannhuber
- a BIRD-C GmbH ; Vienna , Austria.,b Centre of Molecular Biology ; University of Vienna ; Vienna , Austria
| | | | | | - Angela Witte
- c Department of Microbiology , Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna , Vienna , Austria
| | | |
Collapse
|
103
|
Kraśko JA, Žilionytė K, Darinskas A, Strioga M, Rjabceva S, Zalutsky I, Derevyanko M, Kulchitsky V, Lubitz W, Kudela P, Miseikyte-Kaubriene E, Karaman O, Didenko H, Potebnya H, Chekhun V, Pašukonienė V. Bacterial ghosts as adjuvants in syngeneic tumour cell lysate-based anticancer vaccination in a murine lung carcinoma model. Oncol Rep 2016; 37:171-178. [PMID: 27878261 DOI: 10.3892/or.2016.5252] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/22/2016] [Indexed: 11/06/2022] Open
Abstract
Instead of relying on external anticancer factors for treatment, immunotherapy utilizes the host's own immune system and directs it against given tumour antigens. This study demonstrated that it is possible to overcome the documented immunosuppressive properties of tumour cell lysate by supplementing it with appropriate adjuvant. Lewis lung carcinoma (LLC)‑challenged C57BL/6 mice were treated with LLC cryo‑lysate mixed with either bacterial ghosts (BGs) generated from E. coli Nissle 1917 or B. subtilis 70 kDa protein as adjuvants. Median and overall survival, the size of metastatic foci in lung tissue and levels of circulating CD8a+ T cells were evaluated and compared to the untreated control mice or mice treated with LLC lysate alone. After primary tumour removal, a course of three subcutaneous vaccinations with LLC lysate supplemented with BGs led to a significant increase in overall survival (80% after 84 days of follow‑up vs. 40% in untreated control mice), a significant increase in circulating CD8a+ T cells (16.57 vs. 12.6% in untreated control mice) and a significant decrease in metastasis foci area and incidence. LLC lysate supplemented with B. subtilis protein also improved the inspected parameters in the treated mice, when compared against the untreated control mice, but not to a significant degree. Therefore, whole cell lysate supplemented with BGs emerges as an immunostimulatory construct with potential clinical applications in cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Iosif Zalutsky
- Institute of Physiology, BY-220072 Minsk, Republic of Belarus
| | | | | | | | | | | | - Olha Karaman
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - Hennadii Didenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - Hryhorii Potebnya
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - Vasyl Chekhun
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | | |
Collapse
|
104
|
Bioengineered and biohybrid bacteria-based systems for drug delivery. Adv Drug Deliv Rev 2016; 106:27-44. [PMID: 27641944 DOI: 10.1016/j.addr.2016.09.007] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
The use of bacterial cells as agents of medical therapy has a long history. Research that was ignited over a century ago with the accidental infection of cancer patients has matured into a platform technology that offers the promise of opening up new potential frontiers in medical treatment. Bacterial cells exhibit unique characteristics that make them well-suited as smart drug delivery agents. Our ability to genetically manipulate the molecular machinery of these cells enables the customization of their therapeutic action as well as its precise tuning and spatio-temporal control, allowing for the design of unique, complex therapeutic functions, unmatched by current drug delivery systems. Early results have been promising, but there are still many important challenges that must be addressed. We present a review of promises and challenges of employing bioengineered bacteria in drug delivery systems and introduce the biohybrid design concept as a new additional paradigm in bacteria-based drug delivery.
Collapse
|
105
|
Characterization of Chemically-Induced Bacterial Ghosts (BGs) Using Sodium Hydroxide-Induced Vibrio parahaemolyticus Ghosts (VPGs). Int J Mol Sci 2016; 17:ijms17111904. [PMID: 27854308 PMCID: PMC5133902 DOI: 10.3390/ijms17111904] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/01/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022] Open
Abstract
Acellular bacterial ghosts (BGs) are empty non-living bacterial cell envelopes, commonly generated by controlled expression of the cloned lysis gene E of bacteriophage PhiX174. In this study, Vibrio parahaemolyticus ghosts (VPGs) were generated by chemically-induced lysis and the method is based on minimum inhibitory concentration (MIC) of sodium hydroxide (NaOH), acetic acid, boric acid, citric acid, maleic acid, hydrochloric acid, and sulfuric acid. The MIC values of the respective chemicals were 3.125, 6.25, <50.0, 25.0, 6.25, 1.56, and 0.781 mg/mL. Except for boric acid, the lysis efficiency reached more than 99.99% at 5 min after treatment of all chemicals. Among those chemicals, NaOH-induced VPGs appeared completely DNA-free, which was confirmed by quantitative real-time PCR. Besides, lipopolysaccharides (LPS) extracted from the NaOH-induced VPGs showed no distinctive band on SDS-PAGE gel after silver staining. On the other hand, LPS extracted from wild-type bacterial cells, as well as the organic acids-induced VPGs showed triple major bands and LPS extracted from the inorganic acids-induced VPGs showed double bands. It suggests that some surface structures in LPS of the NaOH-induced VPGs may be lost, weakened, or modified by the MIC of NaOH. Nevertheless, Limulus amoebocyte lysate assay revealed that there is no significant difference in endotoxic activity between the NaOH-induced VPGs and wild-type bacterial cells. Macrophages exposed to the NaOH-induced VPGs at 0.5 × 106 CFU/mL showed cell viability of 97.9%, however, the MIC of NaOH did not reduce the cytotoxic effect of wild-type bacterial cells. Like Escherichia coli LPS, the NaOH-induced VPGs are an excellent activator of pro-inflammatory cytokines (IL-1β and iNOS), anti-inflammatory cytokine (IL-10), and dual activities (IL-6) in the stimulated macrophage cells. On the other hand, the induction of TNF-α mRNA was remarkable in the macrophages exposed with wild-type cells. Scanning electron microscopy showed the formation of trans-membrane lysis tunnel structures in the NaOH-induced VPGs. SDS-PAGE and agarose gel electrophoresis also confirmed that cytoplasmic proteins and genomic DNA released from the VPGs to culture medium through the lysis tunnel structures. Taken together, all these data indicate that the NaOH-induced VPGs show the potency of a safe, economical, and effective inactivated bacterial vaccine candidate.
Collapse
|
106
|
Kwon AJ, Moon JY, Kim WK, Kim S, Hur J. Protection efficacy of the Brucella abortus ghost vaccine candidate lysed by the N-terminal 24-amino acid fragment (GI24) of the 36-amino acid peptide PMAP-36 (porcine myeloid antimicrobial peptide 36) in murine models. J Vet Med Sci 2016; 78:1541-1548. [PMID: 27349900 PMCID: PMC5095622 DOI: 10.1292/jvms.16-0036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Brucella abortus cells were lysed by the N-terminal 24-amino acid fragment (GI24) of the 36-amino acid peptide PMAP-36 (porcine myeloid
antimicrobial peptide 36). Next, the protection efficacy of the lysed fragment as a vaccine candidate was evaluated. Group A mice were immunized with sterile
PBS, group B mice were intraperitoneally (ip) immunized with 3 × 108 colony-forming units (CFUs) of B. abortus strain RB51, group C
mice were immunized ip with 3 × 108 cells of the B. abortus vaccine candidate, and group D mice were orally immunized with 3 ×
109 cells of the B. abortus vaccine candidate. Brucella lipopolysaccharide (LPS)-specific serum IgG titers were
considerably higher in groups C and D than in group A. The levels of interleukin (IL)-4, IL-10, tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ)
were significantly higher in groups B–D than in group A. After an ip challenge with B. abortus 544, only group C mice showed a significant
level of protection as compared to group A. Overall, these results show that ip immunization with a vaccine candidate lysed by GI24 can effectively protect mice
from systemic infection with virulent B. abortus.
Collapse
Affiliation(s)
- Ae Jeong Kwon
- Veterinary Public Health, College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | | | | | | | | |
Collapse
|
107
|
Bobbala S, Hook S. Is There an Optimal Formulation and Delivery Strategy for Subunit Vaccines? Pharm Res 2016; 33:2078-97. [DOI: 10.1007/s11095-016-1979-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/21/2016] [Indexed: 12/16/2022]
|
108
|
In vitro responses of chicken macrophage-like monocytes following exposure to pathogenic and non-pathogenic E. coli ghosts loaded with a rational design of conserved genetic materials of influenza and Newcastle disease viruses. Vet Immunol Immunopathol 2016; 176:5-17. [PMID: 27288852 DOI: 10.1016/j.vetimm.2016.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 04/30/2016] [Accepted: 05/06/2016] [Indexed: 11/21/2022]
Abstract
Avian influenza virus (AIV) and Newcastle disease virus (NDV) are two important viral diseases in the poultry industry. Therefore, new disease-fighting strategies, especially effective genetic vaccination, are in high demand. Bacterial Ghost (BG) is a promising platform for delivering genetic materials to macrophages, cells that are among the first to encounter these viruses. However, there is no investigation on the immune response of these macrophage-targeted treatments. Here, we investigated the effect of genetic materials of AIV and NDV on the gene expression profile of important pro-inflammatory cytokines, a chemokine, a transcription factor, major histocompatibility complexes, and the viability of the chicken macrophage-like monocyte cells (CMM). Our genetic construct contained the external domain of matrix protein 2 and nucleoprotein gene of AIV, and immunodominant epitopes of fusion and hemagglutinin-neuraminidase proteins of NDV (hereinafter referred to as pAIV-Vax), delivered via the pathogenic and non-pathogenic BGs (Escherichia coli O78K80 and E. coli TOP10 respectively). The results demonstrated that both types of BGs were able to efficiently deliver the construct to the CMM, although the pathogenic strain derived BG was a significantly better stimulant and delivery vehicle. Both BGs were safe regarding LPS toxicity and did not induce any cell death. Furthermore, the loaded BGs were more powerful in modulating the pro-inflammatory cytokines' responses and antigen presentation systems in comparison to the unloaded BGs. Nitric oxide production of the BG-stimulated cells was also comparable to those challenged by the live bacteria. According to the results, the combination of pAIV-Vax construct and E. coli O78K80 BG is promising in inducing a considerable innate and adaptive immune response against AIV-NDV and perhaps the pathogenic E. coli, provided that the current combination be a potential candidate for in vivo testing regarding the development of an effective trivalent DNA vaccine against avian influenza and Newcastle disease, as well as a bacterial ghost vaccine against avian pathogenic E. coli (APEC).
Collapse
|
109
|
Genital Chlamydia trachomatis: understanding the roles of innate and adaptive immunity in vaccine research. Clin Microbiol Rev 2016; 27:346-70. [PMID: 24696438 DOI: 10.1128/cmr.00105-13] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted disease worldwide, and despite significant advances in chlamydial research, a prophylactic vaccine has yet to be developed. This Gram-negative obligate intracellular bacterium, which often causes asymptomatic infection, may cause pelvic inflammatory disease (PID), ectopic pregnancies, scarring of the fallopian tubes, miscarriage, and infertility when left untreated. In the genital tract, Chlamydia trachomatis infects primarily epithelial cells and requires Th1 immunity for optimal clearance. This review first focuses on the immune cells important in a chlamydial infection. Second, we summarize the research and challenges associated with developing a chlamydial vaccine that elicits a protective Th1-mediated immune response without inducing adverse immunopathologies.
Collapse
|
110
|
Guo R, Geng S, Jiao H, Pan Z, Chen X, Jiao X. Evaluation of protective efficacy of a novel inactivated Salmonella Pullorum ghost vaccine against virulent challenge in chickens. Vet Immunol Immunopathol 2016; 173:27-33. [PMID: 27090623 DOI: 10.1016/j.vetimm.2016.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/18/2016] [Accepted: 03/25/2016] [Indexed: 12/14/2022]
Abstract
Salmonella Gallinarum biovar Pullorum is the causative agent of pullorum disease in poultry, an acute systemic disease that results in a high mortality rate in young chickens. Vaccines have been considered in many developing countries where levels of infection are high and eradication is not a realistic option. An attenuated strain combined with protein E-mediated cell lysis was used to generate a safety enhanced Salmonella Pullorum ghost vaccine. Immune responses and protection induced by ghost vaccine in chickens were investigated following a prime-boost immunization administered via intramuscular and oral routes. Chickens from vaccinated groups showed significant increases in antigen-specific IgG, especially after booster immunization. Lymphocyte proliferation responses were also significantly increased in all immunized groups at 2-weeks post-final vaccination. The Salmonella Pullorum ghost vaccine provided satisfactory protection against virulent Salmonella Pullorum infection, as shown by the robust stimulation of both humoral and cell-mediated immune responses as well as the reduction in the number of bacterial recovered post-challenge. Moreover, the immune effects and survival rates indicated intramuscular injection is more efficient than oral vaccination. In conclusion, our results suggest that Salmonella Pullorum ghosts may be used as a safe and effective novel inactivated vaccine candidate to protect against virulent Salmonella Pullorum infection.
Collapse
Affiliation(s)
- Rongxian Guo
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Shizhong Geng
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Hongmei Jiao
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China; School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
111
|
Meitz A, Sagmeister P, Lubitz W, Herwig C, Langemann T. Fed-Batch Production of Bacterial Ghosts Using Dielectric Spectroscopy for Dynamic Process Control. Microorganisms 2016; 4:microorganisms4020018. [PMID: 27681912 PMCID: PMC5029484 DOI: 10.3390/microorganisms4020018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/11/2016] [Accepted: 03/18/2016] [Indexed: 11/17/2022] Open
Abstract
The Bacterial Ghost (BG) platform technology evolved from a microbiological expression system incorporating the ϕX174 lysis gene E. E-lysis generates empty but structurally intact cell envelopes (BGs) from Gram-negative bacteria which have been suggested as candidate vaccines, immunotherapeutic agents or drug delivery vehicles. E-lysis is a highly dynamic and complex biological process that puts exceptional demands towards process understanding and control. The development of a both economic and robust fed-batch production process for BGs required a toolset capable of dealing with rapidly changing concentrations of viable biomass during the E-lysis phase. This challenge was addressed using a transfer function combining dielectric spectroscopy and soft-sensor based biomass estimation for monitoring the rapid decline of viable biomass during the E-lysis phase. The transfer function was implemented to a feed-controller, which followed the permittivity signal closely and was capable of maintaining a constant specific substrate uptake rate during lysis phase. With the described toolset, we were able to increase the yield of BG production processes by a factor of 8–10 when compared to currently used batch procedures reaching lysis efficiencies >98%. This provides elevated potentials for commercial application of the Bacterial Ghost platform technology.
Collapse
Affiliation(s)
- Andrea Meitz
- Research Center Pharmaceutical Engineering (RCPE) GmbH, Inffeldgasse 13, Graz A-8010, Austria.
| | - Patrick Sagmeister
- Research Division Biochemical Engineering, Institute of Chemical Engineering, Vienna University of Technology, Gumpendorfer Strasse 1A 166/4, Vienna A-1060, Austria.
| | - Werner Lubitz
- Biotech Innovation Research Development and Consulting (BIRD-C) GmbH & Co KG, Dr.-Bohr-Gasse 2-8, Vienna A-1030, Austria.
- Center of Molecular Biology, University of Vienna, Dr.-Bohr-Gasse 9, Vienna A-1030, Austria.
| | - Christoph Herwig
- Research Division Biochemical Engineering, Institute of Chemical Engineering, Vienna University of Technology, Gumpendorfer Strasse 1A 166/4, Vienna A-1060, Austria.
| | - Timo Langemann
- Research Center Pharmaceutical Engineering (RCPE) GmbH, Inffeldgasse 13, Graz A-8010, Austria.
- Biotech Innovation Research Development and Consulting (BIRD-C) GmbH & Co KG, Dr.-Bohr-Gasse 2-8, Vienna A-1030, Austria.
| |
Collapse
|
112
|
Ehgartner D, Herwig C, Neutsch L. At-line determination of spore inoculum quality in Penicillium chrysogenum bioprocesses. Appl Microbiol Biotechnol 2016; 100:5363-73. [DOI: 10.1007/s00253-016-7319-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 11/30/2022]
|
113
|
Inic-Kanada A, Stojanovic M, Schlacher S, Stein E, Belij-Rammerstorfer S, Marinkovic E, Lukic I, Montanaro J, Schuerer N, Bintner N, Kovacevic-Jovanovic V, Krnjaja O, Mayr UB, Lubitz W, Barisani-Asenbauer T. Delivery of a Chlamydial Adhesin N-PmpC Subunit Vaccine to the Ocular Mucosa Using Particulate Carriers. PLoS One 2015; 10:e0144380. [PMID: 26656797 PMCID: PMC4684359 DOI: 10.1371/journal.pone.0144380] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/17/2015] [Indexed: 11/18/2022] Open
Abstract
Trachoma, caused by the intracellular bacterium Chlamydia trachomatis (Ct), remains the world's leading preventable infectious cause of blindness. Recent attempts to develop effective vaccines rely on modified chlamydial antigen delivery platforms. As the mechanisms engaged in the pathology of the disease are not fully understood, designing a subunit vaccine specific to chlamydial antigens could improve safety for human use. We propose the delivery of chlamydia-specific antigens to the ocular mucosa using particulate carriers, bacterial ghosts (BGs). We therefore characterized humoral and cellular immune responses after conjunctival and subcutaneous immunization with a N-terminal portion (amino acid 1-893) of the chlamydial polymorphic membrane protein C (PmpC) of Ct serovar B, expressed in probiotic Escherichia coli Nissle 1917 bacterial ghosts (EcN BGs) in BALB/c mice. Three immunizations were performed at two-week intervals, and the immune responses were evaluated two weeks after the final immunization in mice. In a guinea pig model of ocular infection animals were immunized in the same manner as the mice, and protection against challenge was assessed two weeks after the last immunization. N-PmpC was successfully expressed within BGs and delivery to the ocular mucosa was well tolerated without signs of inflammation. N-PmpC-specific mucosal IgA levels in tears yielded significantly increased levels in the group immunized via the conjunctiva compared with the subcutaneously immunized mice. Immunization with N-PmpC EcN BGs via both immunization routes prompted the establishment of an N-PmpC-specific IFNγ immune response. Immunization via the conjunctiva resulted in a decrease in intensity of the transitional inflammatory reaction in conjunctiva of challenged guinea pigs compared with subcutaneously and non-immunized animals. The delivery of the chlamydial subunit vaccine to the ocular mucosa using a particulate carrier, such as BGs, induced both humoral and cellular immune responses. Further investigations are needed to improve the immunization scheme and dosage.
Collapse
Affiliation(s)
- Aleksandra Inic-Kanada
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centers of Expertise, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Marijana Stojanovic
- Department of Research and Development, Institute of Virology, Vaccines and Sera–TORLAK, Belgrade, Serbia
| | - Simone Schlacher
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centers of Expertise, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Stein
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centers of Expertise, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sandra Belij-Rammerstorfer
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centers of Expertise, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Emilija Marinkovic
- Department of Research and Development, Institute of Virology, Vaccines and Sera–TORLAK, Belgrade, Serbia
| | - Ivana Lukic
- Department of Research and Development, Institute of Virology, Vaccines and Sera–TORLAK, Belgrade, Serbia
| | - Jacqueline Montanaro
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centers of Expertise, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Nadine Schuerer
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centers of Expertise, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Nora Bintner
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centers of Expertise, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Vesna Kovacevic-Jovanovic
- Department of Research and Development, Institute of Virology, Vaccines and Sera–TORLAK, Belgrade, Serbia
| | - Ognjen Krnjaja
- Department of Research and Development, Institute of Virology, Vaccines and Sera–TORLAK, Belgrade, Serbia
| | | | | | - Talin Barisani-Asenbauer
- OCUVAC–Center of Ocular Inflammation and Infection, Laura Bassi Centers of Expertise, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
114
|
Sührer I, Langemann T, Lubitz W, Weuster-Botz D, Castiglione K. A novel one-step expression and immobilization method for the production of biocatalytic preparations. Microb Cell Fact 2015; 14:180. [PMID: 26577293 PMCID: PMC4650107 DOI: 10.1186/s12934-015-0371-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/28/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Whole cell biocatalysts and isolated enzymes are considered as state of the art in biocatalytic preparations for industrial applications. Whole cells as biocatalysts are disadvantageous if substrate or products are toxic to the cells or undesired byproducts are formed due to the cellular metabolism. The use of isolated enzymes in comparison is more expensive due to the required downstream processing. Immobilization of enzymes after purification increases preparation costs for biocatalysts significantly, but allows for the efficient reuse of the enzymes in the biocatalytic process. For a more rapid processing one-step expression and immobilization is desirable. RESULTS This study focused on the development of a new one-step expression and immobilization technique for enzymes on the example of the β-galactosidase from Escherichia coli K12. The enzyme was expressed in E. coli with a C-terminal membrane anchor originating from cytochrome b5 from rabbit liver and was thus in situ immobilized to the inner surface of the cytosolic membrane. Then, the expression of a lytic phage protein (gene E from PhiX174) caused the formation of a pore in the cell wall of E. coli, which resulted in release of the cytosol. The cellular envelopes with immobilized enzymes were retained. Batch and fed-batch processes were developed for efficient production of these biocatalysts. It was possible to obtain cellular envelopes with up to 27,200 ± 10,460 immobilized enzyme molecules per cellular envelope (753 ± 190 U/gdry weight). A thorough characterization of the effects of membrane immobilization was performed. Comparison to whole cells showed that mass transfer limitation was reduced in cellular envelopes due to the pore formation. CONCLUSION In this study the feasibility of a new one-step expression and immobilization technique for the generation of biocatalytic preparations was demonstrated. The technique could be a useful tool especially for enzyme systems, which are not suitable for whole-cell biocatalysts due to severe mass transfer limitations or undesired side reactions mediated by cytosolic enzymes.
Collapse
Affiliation(s)
- Ilka Sührer
- Institute of Biochemical Engineering, Technische Universität München, Boltzmannstr. 15, 85748, Garching, Germany.
| | - Timo Langemann
- BIRD-C GmbH & Co KG, Erne-Seder-Gasse 4/2, 1030, Vienna, Austria.
| | - Werner Lubitz
- BIRD-C GmbH & Co KG, Erne-Seder-Gasse 4/2, 1030, Vienna, Austria.
| | - Dirk Weuster-Botz
- Institute of Biochemical Engineering, Technische Universität München, Boltzmannstr. 15, 85748, Garching, Germany.
| | - Kathrin Castiglione
- Institute of Biochemical Engineering, Technische Universität München, Boltzmannstr. 15, 85748, Garching, Germany.
| |
Collapse
|
115
|
Langemann T, Mayr UB, Meitz A, Lubitz W, Herwig C. Multi-parameter flow cytometry as a process analytical technology (PAT) approach for the assessment of bacterial ghost production. Appl Microbiol Biotechnol 2015; 100:409-18. [PMID: 26521248 DOI: 10.1007/s00253-015-7089-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/29/2015] [Accepted: 10/13/2015] [Indexed: 10/22/2022]
Abstract
Flow cytometry (FCM) is a tool for the analysis of single-cell properties in a cell suspension. In this contribution, we present an improved FCM method for the assessment of E-lysis in Enterobacteriaceae. The result of the E-lysis process is empty bacterial envelopes-called bacterial ghosts (BGs)-that constitute potential products in the pharmaceutical field. BGs have reduced light scattering properties when compared with intact cells. In combination with viability information obtained from staining samples with the membrane potential-sensitive fluorescent dye bis-(1,3-dibutylarbituric acid) trimethine oxonol (DiBAC4(3)), the presented method allows to differentiate between populations of viable cells, dead cells, and BGs. Using a second fluorescent dye RH414 as a membrane marker, non-cellular background was excluded from the data which greatly improved the quality of the results. Using true volumetric absolute counting, the FCM data correlated well with cell count data obtained from colony-forming units (CFU) for viable populations. Applicability of the method to several Enterobacteriaceae (different Escherichia coli strains, Salmonella typhimurium, Shigella flexneri 2a) could be shown. The method was validated as a resilient process analytical technology (PAT) tool for the assessment of E-lysis and for particle counting during 20-l batch processes for the production of Escherichia coli Nissle 1917 BGs.
Collapse
Affiliation(s)
- Timo Langemann
- BIRD-C GmbH & Co KG, Dr.-Bohr-Gasse 2-8, A-1030, Vienna, Austria.,RCPE-Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, A-8010, Graz, Austria
| | | | - Andrea Meitz
- RCPE-Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, A-8010, Graz, Austria
| | - Werner Lubitz
- BIRD-C GmbH & Co KG, Dr.-Bohr-Gasse 2-8, A-1030, Vienna, Austria.,Center of Molecular Biology, University of Vienna, Dr.-Bohr-Gasse 9, A-1030, Vienna, Austria
| | - Christoph Herwig
- Research Division Biochemical Engineering, Institute of Chemical Engineering, Vienna University of Technology, Gumpendorfer Strasse 1A 166/4, A-1060, Vienna, Austria.
| |
Collapse
|
116
|
Walduck A, Andersen LP, Raghavan S. Inflammation, Immunity, and Vaccines for Helicobacter pylori Infection. Helicobacter 2015; 20 Suppl 1:17-25. [PMID: 26372820 DOI: 10.1111/hel.12252] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During the last year, a variety of studies have been published that increases our understanding of the basic mechanisms of immunity and inflammation in Helicobacter pylori infection and progression to gastric cancer. Innate immune regulation and epithelial cell response were covered by several studies that contribute with new insights in the host response to H. pylori infection. Also, the adaptive immune response to H. pylori and particularly the role of IL-22 have been addressed in some studies. These advances may improve vaccine development where new strategies have been published. Two major studies analyzed H. pylori genomes of 39 worldwide strains and looked at the protein profiles. In addition, multi-epitope vaccines for therapeutic use have been investigated. Studies on different adjuvants and delivery systems have also given us new insights. This review presents articles from the last year that reveal detailed insight into immunity and regulation of inflammation, the contribution of immune cells to the development of gastric cancer, and understanding mechanisms of vaccine-induced protection.
Collapse
Affiliation(s)
- Anna Walduck
- Health Innovations Research Institute, School of Applied Sciences RMIT University, Bundoora, Melbourne, Vic., Australia
| | - Leif P Andersen
- Department of Infection Control, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sukanya Raghavan
- Department of Microbiology and Immunology, Institute for Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
117
|
Montanaro J, Inic-Kanada A, Ladurner A, Stein E, Belij S, Bintner N, Schlacher S, Schuerer N, Mayr UB, Lubitz W, Leisch N, Barisani-Asenbauer T. Escherichia coli Nissle 1917 bacterial ghosts retain crucial surface properties and express chlamydial antigen: an imaging study of a delivery system for the ocular surface. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3741-54. [PMID: 26229437 PMCID: PMC4516183 DOI: 10.2147/dddt.s84370] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
To target chronic inflammatory ocular surface diseases, a drug delivery platform is needed that is safe, possesses immunomodulatory properties, and can be used either for drug delivery, or as a foreign antigen carrier. A new therapeutic approach that we have previously proposed uses nonliving bacterial ghosts (BGs) as a carrier-delivery system which can be engineered to carry foreign antigens and/or be loaded with therapeutic drugs. The parent strain chosen for development of our BG delivery system is the probiotic Escherichia coli strain Nissle 1917 (EcN), whose intrinsic properties trigger the innate immune system with the flagella and fimbriae used to attach and stimulate epithelial cells. In previous studies, we have shown that EcN BGs are safe for the ocular surface route, but evidence that EcN BGs retain flagella and fimbriae after transformation, has never been visually confirmed. In this study, we used different visualization techniques to determine whether flagella and fimbriae are retained on EcN BGs engineered either for drug delivery or as a foreign antigen carrier. We have also shown by immunoelectron microscopy that EcN retains two foreign antigens after processing to become EcN BGs. Furthermore, we demonstrated that BGs derived from EcN and expressing a foreign antigen attachment to conjunctival epithelial cells in vitro without causing reduced cell viability. These results are an important step in constructing a delivery system based on a nonliving probiotic that is suitable for use in ocular surface diseases pairing immunomodulation and targeted delivery.
Collapse
Affiliation(s)
- Jacqueline Montanaro
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Aleksandra Inic-Kanada
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Angela Ladurner
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Stein
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sandra Belij
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Nora Bintner
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Simone Schlacher
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Nadine Schuerer
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | | | | | - Nikolaus Leisch
- Department of Ecogenomics and Systems Biology, University of Vienna, Vienna, Austria
| | - Talin Barisani-Asenbauer
- Laura Bassi Centres of Expertise, OCUVAC - Centre of Ocular Inflammation and Infection, Centre for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
118
|
Abstract
Bacteria are perfect vessels for targeted cancer therapy. Conventional chemotherapy is limited by passive diffusion, and systemic administration causes severe side effects. Bacteria can overcome these obstacles by delivering therapeutic proteins specifically to tumors. Bacteria have been modified to produce proteins that directly kill cells, induce apoptosis via signaling pathways, and stimulate the immune system. These three modes of bacterial treatment have all been shown to reduce tumor growth in animal models. Bacteria have also been designed to convert nontoxic prodrugs to active therapeutic compounds. The ease of genetic manipulation enables creation of arrays of bacteria that release many new protein drugs. This versatility will allow targeting of multiple cancer pathways and will establish a platform for individualized cancer medicine.
Collapse
|
119
|
Protection against Shiga-Toxigenic Escherichia coli by Non-Genetically Modified Organism Receptor Mimic Bacterial Ghosts. Infect Immun 2015; 83:3526-33. [PMID: 26099582 DOI: 10.1128/iai.00669-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/16/2015] [Indexed: 11/20/2022] Open
Abstract
Shiga-toxigenic Escherichia coli (STEC) causes severe gastrointestinal infections in humans that may lead to life-threatening systemic sequelae, such as the hemolytic uremic syndrome (HUS). Rapid diagnosis of STEC infection early in the course of disease opens a window of opportunity for therapeutic intervention, for example, by administration of agents that neutralize Shiga toxin (Stx) in the gut lumen. We previously developed a recombinant bacterium that expresses a mimic of the Stx receptor globotriaosyl ceramide (Gb3) on its surface through modification of the lipopolysaccharide (A. W. Paton, R. Morona, and J. C. Paton, Nat Med 6:265-270, 2000, http://dx.doi.org/10.1038/73111). This construct was highly efficacious in vivo, protecting mice from otherwise fatal STEC disease, but the fact that it is a genetically modified organism (GMO) has been a barrier to clinical development. In the present study, we have overcome this issue by development of Gb3 receptor mimic bacterial ghosts (BGs) that are not classified as GMOs. Gb3-BGs neutralized Stx1 and Stx2 in vitro with high efficiency, whereas alternative Gb3-expressing non-GMO subbacterial particles (minicells and outer membrane blebs) were ineffective. Gb3-BGs were highly efficacious in a murine model of STEC disease. All mice (10/10) treated with Gb3-BGs survived challenge with a highly virulent O113:H21 STEC strain and showed no pathological signs of renal injury. In contrast, 6/10 mice treated with control BGs succumbed to STEC challenge, and survivors exhibited significant weight loss, neutrophilia, and histopathological evidence of renal damage. Thus, Gb3-BGs offer a non-GMO approach to treatment of STEC infection in humans, particularly in an outbreak setting.
Collapse
|
120
|
Generation of a Novel Staphylococcus aureus Ghost Vaccine and Examination of Its Immunogenicity against Virulent Challenge in Rats. Infect Immun 2015; 83:2957-65. [PMID: 25964469 DOI: 10.1128/iai.00009-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/02/2015] [Indexed: 01/05/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive pathogen that causes a wide range of infections in humans and animals. Bacterial ghosts are nonliving, empty cell envelopes and are well represented as novel vaccine candidates. In this study, we examined the immunogenicity and protective efficacy of S. aureus ghosts (SAGs) against a virulent challenge in rats. Nonliving SAGs were generated by using the MIC of sodium hydroxide. The formation of a transmembrane lysis tunnel structure in SAGs was visualized by scanning electron microscopy. To investigate these SAGs as a vaccine candidate, rats were divided into four groups, A (nonimmunized control), B (orally immunized), C (subcutaneously immunized), and D (intravenously immunized). The IgG antibody responses were significantly stronger in the SAG-immunized groups than in the nonimmunized control group (P < 0.05). Moreover, a significant increase in the populations of CD4(+) and CD8(+) T cells was observed in all three immunized groups (P < 0.05). We also found that serum bactericidal antibodies were significantly elicited in the SAG-immunized groups (P < 0.05). Most importantly, the bacterial loads in the immunized groups were significantly lower than those in the nonimmunized control group (P < 0.01). These results suggest that immunization with SAGs induces immune responses and provides protection against a virulent S. aureus challenge.
Collapse
|
121
|
Zhu W, Zhang Y, Liu X. Efficient production of safety-enhanced Escherichia coli ghosts by tandem expression of PhiX 174 mutant gene E and staphylococcal nuclease A gene. Microbiol Res 2015; 176:7-13. [PMID: 26070687 DOI: 10.1016/j.micres.2015.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/16/2015] [Accepted: 03/16/2015] [Indexed: 01/23/2023]
Abstract
The application of bacterial ghosts as vaccines is limited because of their low lysis efficiency and production and the presence of pathogenic islands and/or antibiotic resistance genes within ghost preparations. To overcome these problems, a new lysis plasmid with fusion gene of mutant gene E and staphylococcal nuclease A gene (mE-L-SNA) were constructed and characterized. The new plasmid pBV-mELS could efficiently induce the genetic inactivation of Escherichia coli cultures, accompanied by the intracellular degradation of the genetic material of host cells, devoid of the presence of pathogenic islands and antibiotic resistance genes within ghost preparations. Furthermore, the lysis efficiency of the plasmid pBV-mELS was not affected by bacterial concentration and could reach 99.99995% for E. coli at late-log phase. However, when the 74-bp non-encoding region of the gene mE-L-SNA were deleted or the first T nucleotide of the gene mE-L-SNA were substituted, these resulting genes lost the function of bacteriolysis, which suggested the 74-bp region of the gene mE-L-SNA, especially the first T nucleotide, played a crucial role in enhancement of bacteriolysis. The lysis system with the gene mE-L-SNA had predominance for large-scale production of safety-enhanced bacterial ghosts. The strategy may provide a promising avenue for efficient production of safe bacterial ghost vaccines.
Collapse
Affiliation(s)
- Wenxing Zhu
- Shandong Provincial Key Lab of Microbial Engineering, School of Bioengineering, Qilu University of Technology, Jinan 250353, PR China
| | - Yuyu Zhang
- Shandong Key Lab of Animal Disease Control and Breeding, Shandong Academy of Agricultural Sciences, Jinan 250100, PR China
| | - Xinli Liu
- Shandong Provincial Key Lab of Microbial Engineering, School of Bioengineering, Qilu University of Technology, Jinan 250353, PR China.
| |
Collapse
|
122
|
Wang J, Ding K, Chen Y, Zhang L, Liu Z, Xue A, Gu W, Yang X, Li X, Huang J, Xing C, Cao Y, Chen M. Detection of thrombin with an aptamer-based macromolecule biosensor using bacterial ghost system. ACS Synth Biol 2014; 3:963-5. [PMID: 25524099 DOI: 10.1021/sb500018f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A rapid on-site detection of exogenous proteins without the need for equipped laboratories or skilled personnel would benefit many areas. We built a rapid protein detection platform based on aptamer-induced inner-membrane scaffolds dimerization by virtue of bacterial ghost system. When the detection platform was coincubated with two kinds of aptamers targeting two different sites of thrombin, green fluorescence or β-lactamase activity were yielded with two different designs. The latter was detected by commercially available testing strips.
Collapse
Affiliation(s)
- Jiasheng Wang
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Ke Ding
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Yujie Chen
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Lifeng Zhang
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Zukai Liu
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Angli Xue
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Wenjia Gu
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Xiaoyue Yang
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Xihan Li
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Jin Huang
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Congcong Xing
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Yunlong Cao
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| | - Ming Chen
- Zhejiang
University Team (ZJU-China) for the International Genetically
Engineered Machine Competition (iGEM), ‡College of Life Sciences, §School of Medicine, ∥Department of Chemical
and Biological Engineering, ⊥College of Agriculture and Biotechnology, #Department of Chemistry, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, ○Department of Physics, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
123
|
Jawale CV, Lee JH. Characterization of a Salmonella Typhimurium ghost carrying an adjuvant protein as a vaccine candidate for the protection of chickens against virulent challenge. Avian Pathol 2014; 43:506-13. [PMID: 25245884 DOI: 10.1080/03079457.2014.966303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this study we describe the generation of a safe, immunogenic, genetically inactivated Salmonella Typhimurium ghost vaccine candidate carrying the Escherichia coli heat-labile enterotoxin B subunit (LTB) protein as an adjuvant molecule. An asd(+) p15A ori(-) plasmid pJHL187-LTB harbouring the E lysis gene cassette and a foreign antigen delivery cassette containing the eltB gene was used to transform a Δasd Salmonella Typhimurium (JOL1311) strain to construct the ghost strain, JOL1499. Incubation of mid-logarithmic phase JOL1499 cultures at 42°C resulted in co-expression of the eltB and E lysis genes, leading to the generation of Salmonella Typhimurium ghost cells carrying the LTB protein (Salmonella Typhimurium-LTB ghost). The production of LTB in Salmonella Typhimurium-LTB ghost preparations was confirmed by western blot analysis, and functional activity of the LTB protein to bind with GM1 receptors was determined by means of GM1 enzyme-linked immunosorbent assay. Efficacy of the Salmonella Typhimurium-LTB ghost as a vaccine candidate was evaluated in a chicken model using 56 chickens at 5 weeks old, which were divided into four groups (n = 14): group A was designated the non-vaccinated control group, whereas the birds in groups B, C, and D were immunized intramuscularly with 10(9), 10(8), and 10(7) ghost cells, respectively. Compared with the non-immunized chickens (group A), immunized chickens (groups B, C and D) exhibited increased titres of plasma IgG and intestinal secretory IgA antibodies. After oral challenge with 10(9) colony-forming units of a virulent Salmonella Typhimurium strain, the vaccinated group B birds showed a decrease in internal organ colonization with the challenge strain.
Collapse
Affiliation(s)
- Chetan V Jawale
- a College of Veterinary Medicine , Chonbuk National University , Jeonju , Republic of Korea
| | | |
Collapse
|
124
|
Abstract
Bacterial ghosts are empty cell envelopes of Gram-negative bacteria that can be used as vehicles for antigen delivery. Ghosts are generated by releasing the bacterial cytoplasmic contents through a channel in the cell envelope that is created by the controlled production of the bacteriophage ϕX174 lysis protein E. While ghosts possess all the immunostimulatory surface properties of the original host strain, they do not pose any of the infectious threats associated with live vaccines. Recently, we have engineered the Escherichia coli autotransporter hemoglobin protease (Hbp) into a platform for the efficient surface display of heterologous proteins in Gram-negative bacteria, HbpD. Using the Mycobacterium tuberculosis vaccine target ESAT6 (early secreted antigenic target of 6 kDa), we have explored the application of HbpD to decorate E. coli and Salmonella ghosts with antigens. The use of different promoter systems enabled the concerted production of HbpD-ESAT6 and lysis protein E. Ghost formation was monitored by determining lysis efficiency based on CFU, the localization of a set of cellular markers, fluorescence microscopy, flow cytometry, and electron microscopy. Hbp-mediated surface display of ESAT6 was monitored using a combination of a protease accessibility assay, fluorescence microscopy, flow cytometry and (immuno-)electron microscopy. Here, we show that the concerted production of HbpD and lysis protein E in E. coli and Salmonella can be used to produce ghosts that efficiently display antigens on their surface. This system holds promise for the development of safe and cost-effective vaccines with optimal intrinsic adjuvant activity and exposure of heterologous antigens to the immune system.
Collapse
|
125
|
Chen J, Li N, She F. Helicobacter pylori outer inflammatory protein DNA vaccine-loaded bacterial ghost enhances immune protective efficacy in C57BL/6 mice. Vaccine 2014; 32:6054-60. [PMID: 25236588 DOI: 10.1016/j.vaccine.2014.09.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/28/2014] [Accepted: 09/05/2014] [Indexed: 12/28/2022]
Abstract
Helicobacter pylori (H. pylori) infection is associated with incidents of gastrointestinal diseases in half of the human population. However, management of its infection remains a challenge. Hence, it is necessary to develop an efficient vaccine to fight against this pathogen. In the present study, a novel vaccine based on the production of attenuated Salmonella typhimurium bacterial ghost (SL7207-BG), delivering H. pylori outer inflammatory protein gene (oipA) encoded DNA vaccine was developed, and the efficiency was evaluated in C57BL/6 mice. Significant higher levels of IgG2a/IgG1 antibodies and IFN-γ/IL-4 cytokines were detected after mice were oral administered with oipA DNA vaccine loaded SL7207-BG, indicating that a mixed Th1/Th2 immune response was elicited. When challenged with infective doses H. pylori strain SS1, the ghost based vaccine was capable of reducing bacterium colonization in the vaccinated mice. In addition, codon-optimized oipA plasmid loaded SL7207-BG significantly eliminates H. pylori colonization density in mice model. Thus, it has been demonstrated that this novel bacterial ghost based DNA vaccine could be used as a promising vaccine candidate for the control of H. pylori infection.
Collapse
Affiliation(s)
- Jiansen Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, Fujian, China; Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, Fujian, China; Department of Nosocomial Infection Control, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, China
| | - Neng Li
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, Fujian, China; Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, Fujian, China
| | - Feifei She
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, Fujian, China; Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, Fujian, China.
| |
Collapse
|
126
|
Bergmann-Leitner ES, Leitner WW. Adjuvants in the Driver's Seat: How Magnitude, Type, Fine Specificity and Longevity of Immune Responses Are Driven by Distinct Classes of Immune Potentiators. Vaccines (Basel) 2014; 2:252-96. [PMID: 26344620 PMCID: PMC4494256 DOI: 10.3390/vaccines2020252] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/20/2014] [Accepted: 03/28/2014] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which vaccine adjuvants enhance immune responses has historically been considered to be the creation of an antigen depot. From here, the antigen is slowly released and provided to immune cells over an extended period of time. This "depot" was formed by associating the antigen with substances able to persist at the injection site, such as aluminum salts or emulsions. The identification of Pathogen-Associated Molecular Patterns (PAMPs) has greatly advanced our understanding of how adjuvants work beyond the simple concept of extended antigen release and has accelerated the development of novel adjuvants. This review focuses on the mode of action of different adjuvant classes in regards to the stimulation of specific immune cell subsets, the biasing of immune responses towards cellular or humoral immune response, the ability to mediate epitope spreading and the induction of persistent immunological memory. A better understanding of how particular adjuvants mediate their biological effects will eventually allow them to be selected for specific vaccines in a targeted and rational manner.
Collapse
Affiliation(s)
- Elke S Bergmann-Leitner
- US Military Malaria Research Program, Malaria Vaccine Branch, 503 Robert Grant Ave, 3W65, Silver Spring, MD 20910, USA.
| | - Wolfgang W Leitner
- Division on Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 6610 Rockledge Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
127
|
Muhammad A, Champeimont J, Mayr UB, Lubitz W, Kudela P. Bacterial ghosts as carriers of protein subunit and DNA-encoded antigens for vaccine applications. Expert Rev Vaccines 2014; 11:97-116. [DOI: 10.1586/erv.11.149] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
128
|
Abstract
Over the past three decades, a powerful array of techniques has been developed for expressing heterologous proteins and saccharides on the surface of bacteria. Surface-engineered bacteria, in turn, have proven useful in a variety of settings, including high-throughput screening, biofuel production, and vaccinology. In this chapter, we provide a comprehensive review of methods for displaying polypeptides and sugars on the bacterial cell surface, and discuss the many innovative applications these methods have found to date. While already an important biotechnological tool, we believe bacterial surface display may be further improved through integration with emerging methodology in other fields, such as protein engineering and synthetic chemistry. Ultimately, we envision bacterial display becoming a multidisciplinary platform with the potential to transform basic and applied research in bacteriology, biotechnology, and biomedicine.
Collapse
|
129
|
Guo J, Jia R. A novel inducible expression system for the functional study of toxic gene in bacteria. World J Microbiol Biotechnol 2013; 30:1527-31. [DOI: 10.1007/s11274-013-1573-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 11/30/2013] [Indexed: 10/25/2022]
|
130
|
Bergmann-Leitner ES, Hosie H, Trichilo J, Deriso E, Ranallo RT, Alefantis T, Savranskaya T, Grewal P, Ockenhouse CF, Venkatesan MM, Delvecchio VG, Angov E. Self-adjuvanting bacterial vectors expressing pre-erythrocytic antigens induce sterile protection against malaria. Front Immunol 2013; 4:176. [PMID: 23847617 PMCID: PMC3701146 DOI: 10.3389/fimmu.2013.00176] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 06/18/2013] [Indexed: 11/24/2022] Open
Abstract
Genetically inactivated, Gram-negative bacteria that express malaria vaccine candidates represent a promising novel self-adjuvanting vaccine approach. Antigens expressed on particulate bacterial carriers not only target directly to antigen-presenting cells but also provide a strong danger signal thus circumventing the requirement for potent extraneous adjuvants. E. coli expressing malarial antigens resulted in the induction of either Th1 or Th2 biased responses that were dependent on both antigen and sub-cellular localization. Some of these constructs induced higher quality humoral responses compared to recombinant protein and most importantly they were able to induce sterile protection against sporozoite challenge in a murine model of malaria. In light of these encouraging results, two major Plasmodium falciparum pre-erythrocytic malaria vaccine targets, the Cell-Traversal protein for Ookinetes and Sporozoites (CelTOS) fused to the Maltose-binding protein in the periplasmic space and the Circumsporozoite Protein (CSP) fused to the Outer membrane (OM) protein A in the OM were expressed in a clinically relevant, attenuated Shigella strain (Shigella flexneri 2a). This type of live-attenuated vector has previously undergone clinical investigations as a vaccine against shigellosis. Using this novel delivery platform for malaria, we find that vaccination with the whole-organism represents an effective vaccination alternative that induces protective efficacy against sporozoite challenge. Shigella GeMI-Vax expressing malaria targets warrant further evaluation to determine their full potential as a dual disease, multivalent, self-adjuvanting vaccine system, against both shigellosis, and malaria.
Collapse
|
131
|
Liu HF, Li W, Lu MB, Yu LJ. Pharmacokinetics and risk evaluation of DNA vaccine against Schistosoma japonicum. Parasitol Res 2012; 112:59-67. [PMID: 22990210 DOI: 10.1007/s00436-012-3104-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 08/24/2012] [Indexed: 02/03/2023]
Abstract
DNA plasmid immunization is a novel approach of preventive and therapeutic vaccine. More than 100 DNA vaccines have been on preclinical or clinical phase trials, and four kinds of DNA vaccines for livestock have been approved by USDA, CFIA, and APVMA. Schistosomiasis is a worldwide parasitic disease, and vaccine immunization is supposed to be a promising approach to control the health crisis. On the basis of former preclinical studies, we further focused on the pharmacokinetics and risk evaluation of DNA vaccine in vivo. In the present study, enhanced green fluorescent protein (EGFP) report gene was fused with Schistosoma japonicum 23 kDa transmembrane protein antigen gene (Sj23) and constructed into DNA vaccine pVIVO2-Sj23.EGFP. After intramuscularly injecting 100 μg of purified DNA vaccine plasmid to immunizate BALB/c mice, we studied the tissue distribution of DNA plasmid and expressed Sj23.EGFP antigen, the persistence time of elicited antibodies, and the risk of DNA vaccine transferred into intestinal microorganisms. The results showed that DNA vaccine plasmid could be distributed into all tissues of the body after injection; however, only few organs including the injected muscle were detected DNA vaccine at postimmunization until the 100 days by PCR technology; the detection of green fluorescence protein displayed that DNA vaccine could be expressed in almost every tissue and organs; the ELISA assay indicated the immune antibody against Sj23 could persist over 70 days; and the DNA vaccine transferring intestinal flora results was negative. The results indicated that the DNA vaccine has systemic protection and long-lasting effectivity and is safe to intestinal flora.
Collapse
Affiliation(s)
- Hai-Feng Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China.
| | | | | | | |
Collapse
|
132
|
Paton AW, Morona R, Paton JC. Bioengineered microbes in disease therapy. Trends Mol Med 2012; 18:417-25. [DOI: 10.1016/j.molmed.2012.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 05/11/2012] [Accepted: 05/15/2012] [Indexed: 01/30/2023]
|
133
|
Generation of biotechnology-derived Flavobacterium columnare ghosts by PhiX174 gene E-mediated inactivation and the potential as vaccine candidates against infection in grass carp. J Biomed Biotechnol 2012; 2012:760730. [PMID: 22719209 PMCID: PMC3376489 DOI: 10.1155/2012/760730] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/05/2012] [Indexed: 11/17/2022] Open
Abstract
Flavobacterium columnare is a bacterial pathogen causing high mortality rates for many freshwater fish species. Fish vaccination with a safe and effective vaccine is a potential approach for prevention and control of fish disease. Here, in order to produce bacterial ghost vaccine, a specific Flavobacterium lysis plasmid pBV-E-cat was constructed by cloning PhiX174 lysis gene E and the cat gene with the promoter of F. columnare into the prokaryotic expression vector pBV220. The plasmid was successfully electroporated into the strain F. columnare G4cpN22 after curing of its endogenous plasmid. F. columnare G4cpN22 ghosts (FCGs) were generated for the first time by gene E-mediated lysis, and the vaccine potential of FCG was investigated in grass carp (Ctenopharyngodon idellus) by intraperitoneal route. Fish immunized with FCG showed significantly higher serum agglutination titers and bactericidal activity than fish immunized with FKC or PBS. Most importantly, after challenge with the parent strain G4, the relative percent survival (RPS) of fish in FCG group (70.9%) was significantly higher than FKC group (41.9%). These results showed that FCG could confer immune protection against F. columnare infection. As a nonliving whole cell envelope preparation, FCG may provide an ideal alternative to pathogen-based vaccines against columnaris in aquaculture.
Collapse
|
134
|
Abstract
Cell systems have recently emerged as biological drug carriers, as an interesting alternative to other systems such as micro- and nano-particles. Different cells, such as carrier erythrocytes, bacterial ghosts and genetically engineered stem and dendritic cells have been used. They provide sustained release and specific delivery of drugs, enzymatic systems and genetic material to certain organs and tissues. Cell systems have potential applications for the treatment of cancer, HIV, intracellular infections, cardiovascular diseases, Parkinson’s disease or in gene therapy. Carrier erythrocytes containing enzymes such us L-asparaginase, or drugs such as corticosteroids have been successfully used in humans. Bacterial ghosts have been widely used in the field of vaccines and also with drugs such as doxorubicin. Genetically engineered stem cells have been tested for cancer treatment and dendritic cells for immunotherapeutic vaccines. Although further research and more clinical trials are necessary, cell-based platforms are a promising strategy for drug delivery.
Collapse
|
135
|
Kudela P, Koller VJ, Mayr UB, Nepp J, Lubitz W, Barisani-Asenbauer T. Bacterial Ghosts as antigen and drug delivery system for ocular surface diseases: Effective internalization of Bacterial Ghosts by human conjunctival epithelial cells. J Biotechnol 2011; 153:167-75. [PMID: 21501636 DOI: 10.1016/j.jbiotec.2011.03.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 03/03/2011] [Accepted: 03/29/2011] [Indexed: 11/29/2022]
Abstract
The purpose of the presented investigation was to examine the efficiency of the novel carrier system Bacterial Ghosts (BGs), which are empty bacterial cell envelopes of Gram-negative bacteria to target human conjunctival epithelial cells, as well as to test the endocytic capacity of conjunctival cells after co-incubation with BGs generated from different bacterial species, and to foreclose potential cytotoxic effects caused by BGs. The efficiency of conjunctival cells to internalize BGs was investigated using the Chang conjunctival epithelial cell line and primary human conjunctiva-derived epithelial cells (HCDECs) as in vitro model. A high capacity of HCDECs to functionally internalize BGs was detected with the level of internalization depending on the type of species used for BGs generation. Detailed analysis showed no cytotoxic effect of BGs on HCDECs independently of the used bacterial species. Moreover, co-incubation with BGs did not enhance expression of both MHC class I and class II molecules by HCDECs, but increased expression of ICAM-1. The high rates of BG's internalization by HCDECs with no BG-mediated cytotoxic impact designate this carrier system to be a promising candidate for an ocular surface drug delivery system. BGs could be useful for future therapeutic ocular surface applications and eye-specific disease vaccine development including DNA transfer.
Collapse
Affiliation(s)
- Pavol Kudela
- Department of Ophthalmology and Optometry, Medical University of Vienna, A-1090 Vienna, Austria.
| | | | | | | | | | | |
Collapse
|