1
|
Bento CM, van Calster K, Piller T, Oliveira GS, de Vooght L, Cappoen D, Cos P, Gomes MS, Silva T. Characterization of novel double-reporter strains of Mycobacterium abscessus for drug discovery: a study in mScarlet. Microbiol Spectr 2024; 12:e0036224. [PMID: 39189762 PMCID: PMC11448253 DOI: 10.1128/spectrum.00362-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024] Open
Abstract
Mycobacterium abscessus (Mab) is an emerging pathogen that poses a severe health threat, especially in people with cystic fibrosis and other chronic lung diseases. Available drugs are largely ineffective due to an exquisite intrinsic resistance, making Mab infections only comparable to multidrug-resistant tuberculosis. Current treatment is based on lengthy multidrug therapy, complicated by poor outcomes and high rates of treatment failure, recurrence, and mortality. Thus, finding new and more efficient drugs to combat this pathogen is urgent. However, drug discovery efforts targeting Mab have been limited, and traditional drug screening methods are labor-intensive, low-throughput, and do not reflect clinical effectiveness. Therefore, this work aimed to develop a new, efficient, and reliable tool for drug screening against Mab that can be used in vitro for identifying hits in a high-throughput manner and in vivo to select drug candidates for future clinical trials. We engineered two stable double-reporter strains of Mab capable of emitting strong fluorescent and luminescent signals. This is due to the expression of mScarlet protein and luciferase enzyme or the entire lux operon. Importantly, these strains maintain the same ground characteristics as the non-transformed Mab strain. We show that these new strains can be applied to various setups, from MIC determination in broth cultures and macrophage infection assays to in vivo infection (using the Galleria mellonella model). Using these strains enhances the potential for high-throughput screening of thousands of compounds in a fast and reliable way. IMPORTANCE Mycobacterium abscessus (Mab) is currently considered an "incurable nightmare." Its intrinsic resistance, high toxicity, long duration, and low cure rates of available therapies often lead to the clinical decision not to treat. Moreover, one of the significant drawbacks of anti-Mab drug development is the lack of correlation between in vitro susceptibility and clinical efficacy. Most drug screening assays are performed on Mab growing in liquid cultures. But being an intracellular pathogen, inducing granulomas and biofilm formation, the broth culture is far from ideal as in vitro drug-testing setup. This study presents new double-reporter Mab strains that allow direct real-time bacterial detection and quantification in a non-invasive way. These strains can be applied to an extensive range of experimental settings, far surpassing the utility of single-reporter bacteria. They can be used in all steps of the pre-clinical anti-Mab drug development pipeline, constituting a highly valuable tool to increase its success.
Collapse
Affiliation(s)
- Clara M Bento
- i3S-Instituto de Investigação e Inovação e Saúde, Universidade do Porto, Porto, Portugal
- IBMC-Instituto de Biologia Celular e Molecular, Universidade do Porto, Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCBiology), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Porto, Portugal
| | - Kevin van Calster
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Tatiana Piller
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Gabriel S Oliveira
- i3S-Instituto de Investigação e Inovação e Saúde, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Porto, Portugal
| | - Linda de Vooght
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Davie Cappoen
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - M Salomé Gomes
- i3S-Instituto de Investigação e Inovação e Saúde, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Porto, Portugal
| | - Tânia Silva
- i3S-Instituto de Investigação e Inovação e Saúde, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Porto, Portugal
| |
Collapse
|
2
|
Palčeková Z, Obregón-Henao A, De K, Walz A, Lam H, Philp J, Angala SK, Patterson J, Pearce C, Zuberogoitia S, Avanzi C, Nigou J, McNeil M, Muñoz Gutiérrez JF, Gilleron M, Wheat WH, Gonzalez-Juarrero M, Jackson M. Role of succinyl substituents in the mannose-capping of lipoarabinomannan and control of inflammation in Mycobacterium tuberculosis infection. PLoS Pathog 2023; 19:e1011636. [PMID: 37669276 PMCID: PMC10503756 DOI: 10.1371/journal.ppat.1011636] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 09/15/2023] [Accepted: 08/25/2023] [Indexed: 09/07/2023] Open
Abstract
The covalent modification of bacterial (lipo)polysaccharides with discrete substituents may impact their biosynthesis, export and/or biological activity. Whether mycobacteria use a similar strategy to control the biogenesis of its cell envelope polysaccharides and modulate their interaction with the host during infection is unknown despite the report of a number of tailoring substituents modifying the structure of these glycans. Here, we show that discrete succinyl substituents strategically positioned on Mycobacterium tuberculosis (Mtb) lipoarabinomannan govern the mannose-capping of this lipoglycan and, thus, much of the biological activity of the entire molecule. We further show that the absence of succinyl substituents on the two main cell envelope glycans of Mtb, arabinogalactan and lipoarabinomannan, leads to a significant increase of pro-inflammatory cytokines and chemokines in infected murine and human macrophages. Collectively, our results validate polysaccharide succinylation as a critical mechanism by which Mtb controls inflammation.
Collapse
Affiliation(s)
- Zuzana Palčeková
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Andrés Obregón-Henao
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Kavita De
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Amanda Walz
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Ha Lam
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jamie Philp
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Shiva Kumar Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Johnathan Patterson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Camron Pearce
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Sophie Zuberogoitia
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Charlotte Avanzi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Michael McNeil
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Juan F. Muñoz Gutiérrez
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Martine Gilleron
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - William H. Wheat
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
3
|
Marques-Neto LM, Piwowarska Z, Kanno AI, Moraes L, Trentini MM, Rodriguez D, Silva JLSC, Leite LCC. Thirty years of recombinant BCG: new trends for a centenary vaccine. Expert Rev Vaccines 2021; 20:1001-1011. [PMID: 34224293 DOI: 10.1080/14760584.2021.1951243] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Global perception of the potential for Bacille Calmette-Guérin (BCG), and consequently recombinant BCG (rBCG), in a variety of prophylactic and therapeutic applications has been increasing. A century of information on BCG, and three decades of experience with rBCG, has generated solid knowledge in this field.Area covered: Here, we review the current state of knowledge of BCG and rBCG development. Molecular tools have facilitated the expression of a variety of molecules in BCG, with the aim of improving its efficacy as a tuberculosis vaccine, generating polyvalent vaccines against other pathogens, including viruses, bacteria, and parasites, and developing immunotherapy approaches against noninvasive bladder cancer. BCG's recently appraised heterologous effects and prospects for expanding its application to other diseases are also addressed.Expert opinion: There are high expectations for new tuberculosis vaccines currently undergoing advanced clinical trials, which could change the prospects of the field. Systems biology could reveal effective biomarkers of protection, which would greatly support vaccine development. The development of appropriate large-scale production processes would further support implementation of new vaccines and rBCG products. The next few years should consolidate the broader applications of BCG and produce insights into improvements using the recombinant BCG technology.
Collapse
Affiliation(s)
| | - Zuzanna Piwowarska
- Laboratório De Desenvolvimento De Vacinas, Instituto Butantan, São Paulo, Brazil.,UnivLyon, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Alex I Kanno
- Laboratório De Desenvolvimento De Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Luana Moraes
- Laboratório De Desenvolvimento De Vacinas, Instituto Butantan, São Paulo, Brazil.,Programa De Pós-Graduação Interunidades Em Biotecnologia USP-Instituto Butantan-IPT, São Paulo, Brazil
| | - Monalisa M Trentini
- Laboratório De Desenvolvimento De Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Dunia Rodriguez
- Laboratório De Desenvolvimento De Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Jose L S C Silva
- Laboratório De Desenvolvimento De Vacinas, Instituto Butantan, São Paulo, Brazil.,Programa De Pós-Graduação Interunidades Em Biotecnologia USP-Instituto Butantan-IPT, São Paulo, Brazil
| | - Luciana C C Leite
- Laboratório De Desenvolvimento De Vacinas, Instituto Butantan, São Paulo, Brazil
| |
Collapse
|
4
|
New disease old vaccine: Is recombinant BCG vaccine an answer for COVID-19? Cell Immunol 2020; 356:104187. [PMID: 32745670 PMCID: PMC7386780 DOI: 10.1016/j.cellimm.2020.104187] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/13/2020] [Accepted: 07/24/2020] [Indexed: 12/25/2022]
Abstract
Mycobacterium bovis BCG, a live attenuated tuberculosis vaccine offers protection against disseminated TB in children. BCG exhibits heterologous protective effects against unrelated infections and reduces infant mortality due to non-mycobacterial infections. Recent reports have suggested that BCG vaccination might have protective effects against COVID-19, however it is highly unlikely that BCG vaccine in its current form can offer complete protection against SARS-CoV-2 infection due to the lack of specific immunity. Nonetheless, recombinant BCG strains expressing antigens of SARS-CoV-2 may offer protection against COVID-19 due to the activation of innate as well as specific adaptive immune response. Further proven safety records of BCG in humans, its adjuvant activity and low cost manufacturing makes it a frontrunner in the vaccine development to stop this pandemic. In this review we discuss about the heterologous effects of BCG, induction of trained immunity and its implication in development of a potential vaccine against COVID-19 pandemic.
Collapse
|
5
|
Grzegorzewicz AE, Gee C, Das S, Liu J, Belardinelli JM, Jones V, McNeil MR, Lee RE, Jackson M. Mechanisms of Resistance Associated with the Inhibition of the Dehydration Step of Type II Fatty Acid Synthase in Mycobacterium tuberculosis. ACS Infect Dis 2020; 6:195-204. [PMID: 31775512 DOI: 10.1021/acsinfecdis.9b00162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Isoxyl (ISO) and thiacetazone (TAC) are two antitubercular prodrugs that abolish mycolic acid biosynthesis and kill Mycobacterium tuberculosis (Mtb) through the inhibition of the essential type II fatty acid synthase (FAS-II) dehydratase HadAB. While mutations preventing ISO and TAC either from being converted to their active form or from covalently modifying their target are the most frequent spontaneous mutations associated with high-level resistance to both drugs, the molecular mechanisms underlying the high-level ISO and TAC resistance of Mtb strains harboring missense mutations in the second, nonessential, FAS-II dehydratase HadBC have remained unexplained. Using a combination of genetic, biochemical, and biophysical approaches and molecular dynamics simulation, we here show that all four reported resistance mutations in the HadC subunit of HadBC alter the stability and/or specific activity of the enzyme, allowing it in two cases (HadBCV85I and HadBCK157R) to compensate for a deficiency in HadAB in whole Mtb bacilli. The analysis of the mycolic acid profiles of Mtb strains expressing the mutated forms of HadC further points to alterations in the activity of the mycolic acid biosynthetic complex and suggests an additional contributing resistance mechanism whereby HadC mutations may reduce the accessibility of HadAB to ISO and TAC. Collectively, our results highlight the importance of developing optimized inhibitors of the dehydration step of FAS-II capable of inhibiting both dehydratases simultaneously, a goal that may be achievable given the structural resemblance of the two enzymes and their reliance on the same catalytic subunit HadB.
Collapse
Affiliation(s)
- Anna E. Grzegorzewicz
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682, United States
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, Wroclaw PL-53-114, Poland
| | - Clifford Gee
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Sourav Das
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Jiuyu Liu
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Juan Manuel Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682, United States
| | - Victoria Jones
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682, United States
| | - Michael R. McNeil
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682, United States
| | - Richard E. Lee
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682, United States
| |
Collapse
|
6
|
Covián C, Fernández-Fierro A, Retamal-Díaz A, Díaz FE, Vasquez AE, Lay MK, Riedel CA, González PA, Bueno SM, Kalergis AM. BCG-Induced Cross-Protection and Development of Trained Immunity: Implication for Vaccine Design. Front Immunol 2019; 10:2806. [PMID: 31849980 PMCID: PMC6896902 DOI: 10.3389/fimmu.2019.02806] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022] Open
Abstract
The Bacillus Calmette-Guérin (BCG) is a live attenuated tuberculosis vaccine that has the ability to induce non-specific cross-protection against pathogens that might be unrelated to the target disease. Vaccination with BCG reduces mortality in newborns and induces an improved innate immune response against microorganisms other than Mycobacterium tuberculosis, such as Candida albicans and Staphylococcus aureus. Innate immune cells, including monocytes and natural killer (NK) cells, contribute to this non-specific immune protection in a way that is independent of memory T or B cells. This phenomenon associated with a memory-like response in innate immune cells is known as "trained immunity." Epigenetic reprogramming through histone modification in the regulatory elements of particular genes has been reported as one of the mechanisms associated with the induction of trained immunity in both, humans and mice. Indeed, it has been shown that BCG vaccination induces changes in the methylation pattern of histones associated with specific genes in circulating monocytes leading to a "trained" state. Importantly, these modifications can lead to the expression and/or repression of genes that are related to increased protection against secondary infections after vaccination, with improved pathogen recognition and faster inflammatory responses. In this review, we discuss BCG-induced cross-protection and acquisition of trained immunity and potential heterologous effects of recombinant BCG vaccines.
Collapse
Affiliation(s)
- Camila Covián
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Ayleen Fernández-Fierro
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angello Retamal-Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián E Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Abel E Vasquez
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago, Chile
| | - Margarita K Lay
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
7
|
A Bivalent Recombinant Mycobacterium bovis BCG Expressing the S1 Subunit of the Pertussis Toxin Induces a Polyfunctional CD4 + T Cell Immune Response. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9630793. [PMID: 30941374 PMCID: PMC6420988 DOI: 10.1155/2019/9630793] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/30/2018] [Indexed: 12/15/2022]
Abstract
Background A recombinant BCG strain expressing the genetically detoxified S1 subunit of pertussis toxin 9K/129G (rBCG-S1PT), previously constructed by our research group, demonstrated the ability to develop high protection in mouse models of pertussis challenge which correlated with the induction of a Th1 immune response pattern. The Th1 immune response induced by rBCG-S1PT treatment was also confirmed in the murine orthotopic bladder cancer model, in which the intravesical instillation of rBCG-S1PT resulted in an improved antitumor effect. Based on these observations, we hypothesize that the reengineering of the S1PT expression in BCG could increase the efficiency of the protective Th1 immune response in order to develop a new alternative of immunotherapy in bladder cancer treatment. Objectives To construct rBCG strains expressing S1PT from extrachromosomal (rBCG-S1PT) and integrative vectors (rBCG-Sli), or their combination, generating the bivalent strain (rBCG-S1+S1i), and to evaluate the respective immunogenicity of rBCG strains in mice. Methods Mycobacterial plasmids were constructed by cloning the s1pt gene under integrative and extrachromosomal vectors and used to transform BCG, individually or in combination. Antigen expression and localization were confirmed by Western blot. Mice were immunized with wild-type BCG or the rBCG strains, and cytokines quantification and flow cytometry analysis were performed in splenocytes culture stimulated with mycobacterial-specific proteins. Findings S1PT expression was confirmed in all rBCG strains. The extrachromosomal vector directs S1PT to the cell wall-associated fraction, while the integrative vector directs its expression mainly to the intracellular fraction. Higher levels of IFN-γ were observed in the splenocytes culture from the group immunized with rBCG-S1i in comparison to BCG or rBCG-S1PT. rBCG-S1+S1i showed higher levels of CD4+ IFN-γ+ and double-positive CD4+ IFN-γ+ TNF-α+ T cells. Conclusions rBCG-S1+S1i was able to express the two forms of S1PT and elicited higher induction of polyfunctional CD4+ T cells, indicating enhanced immunogenicity and suggesting its use as immunotherapy for bladder cancer.
Collapse
|
8
|
Broset E, Saubi N, Guitart N, Aguilo N, Uranga S, Kilpeläinen A, Eto Y, Hanke T, Gonzalo-Asensio J, Martín C, Joseph-Munné J. MTBVAC-Based TB-HIV Vaccine Is Safe, Elicits HIV-T Cell Responses, and Protects against Mycobacterium tuberculosis in Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 13:253-264. [PMID: 30859110 PMCID: PMC6395831 DOI: 10.1016/j.omtm.2019.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 01/30/2019] [Indexed: 01/11/2023]
Abstract
The tuberculosis (TB) vaccine MTBVAC is the only live-attenuated Mycobacterium tuberculosis (Mtb)-based vaccine in clinical development, and it confers superior protection in different animal models compared to the current vaccine, BCG (Mycobacterium bovis bacillus Calmette-Guérin). With the aim of using MTBVAC as a vector for a dual TB-HIV vaccine, we constructed the recombinant MTBVAC.HIVA2auxo strain. First, we generated a lysine auxotroph of MTBVAC (MTBVACΔlys) by deleting the lysA gene. Then the auxotrophic MTBVACΔlys was transformed with the E. coli-mycobacterial vector p2auxo.HIVA, harboring the lysA-complementing gene and the HIV-1 clade A immunogen HIVA. This TB-HIV vaccine conferred similar efficacy to the parental strain MTBVAC against Mtb challenge in mice. MTBVAC.HIVA2auxo was safer than BCG and MTBVAC in severe combined immunodeficiency (SCID) mice, and it was shown to be maintained up to 42 bacterial generations in vitro and up to 100 days after inoculation in vivo. The MTBVAC.HIVA2auxo vaccine, boosted with modified vaccinia virus Ankara (MVA).HIVA, induced HIV-1 and Mtb-specific interferon-γ-producing T cell responses and polyfunctional HIV-1-specific CD8+ T cells producing interferon-γ (IFN-γ), tumor necrosis factor alpha (TNF-α), and CD107a in BALB/c mice. Here we describe new tools to develop combined vaccines against TB and HIV with the potential of expansion for other infectious diseases.
Collapse
Affiliation(s)
- Esther Broset
- Grupo de Genética de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, C/Domingo Miral s/n, Zaragoza 50009, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Zaragoza, Spain
| | - Narcís Saubi
- AIDS Research Group, Hospital Clínic de Barcelona/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain.,Red Temática de Investigación Cooperativa en SIDA (RD12/0017/0001), Spanish AIDS Network, Madrid, Spain
| | - Núria Guitart
- AIDS Research Group, Hospital Clínic de Barcelona/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain
| | - Nacho Aguilo
- Grupo de Genética de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, C/Domingo Miral s/n, Zaragoza 50009, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Santiago Uranga
- Grupo de Genética de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, C/Domingo Miral s/n, Zaragoza 50009, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Athina Kilpeläinen
- AIDS Research Group, Hospital Clínic de Barcelona/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain.,EAVI2020 European AIDS Vaccine Initiative H2020 Research Programme, London, UK
| | - Yoshiki Eto
- AIDS Research Group, Hospital Clínic de Barcelona/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jesús Gonzalo-Asensio
- Grupo de Genética de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, C/Domingo Miral s/n, Zaragoza 50009, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Zaragoza, Spain.,Servicio de Microbiología, Hospital Universitario Miguel Servet, IIS Aragón, Zaragoza, Spain
| | - Carlos Martín
- Grupo de Genética de Micobacterias, Departamento de Microbiología y Medicina Preventiva, Facultad de Medicina, Universidad de Zaragoza, C/Domingo Miral s/n, Zaragoza 50009, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Servicio de Microbiología, Hospital Universitario Miguel Servet, IIS Aragón, Zaragoza, Spain
| | - Joan Joseph-Munné
- AIDS Research Group, Hospital Clínic de Barcelona/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain.,EAVI2020 European AIDS Vaccine Initiative H2020 Research Programme, London, UK.,Servei de Malalties Infeccioses, Hospital Clínic de Barcelona, Catalonia, Spain
| |
Collapse
|
9
|
Kilpeläinen A, Maya-Hoyos M, Saubí N, Soto CY, Joseph Munne J. Advances and challenges in recombinant Mycobacterium bovis BCG-based HIV vaccine development: lessons learned. Expert Rev Vaccines 2018; 17:1005-1020. [PMID: 30300040 DOI: 10.1080/14760584.2018.1534588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Human Immunodeficiency Virus/Acquired Immune Deficiency Syndrome, tuberculosis, and malaria are responsible for most human deaths produced by infectious diseases worldwide. Vaccination against HIV requires generation of memory T cells and neutralizing antibodies, mucosal immunity, and stimulation of an innate immune responses. In this context, the use of Mycobacterium bovis bacillus Calmette-Guérin (BCG) as a live vaccine vehicle is a promising approach for T-cell induction. AREAS COVERED In this review, we provide a comprehensive summary of the literature regarding immunogenicity studies in animal models performed since 2005. Furthermore, we provide expert commentary and 5-year view on how the development of potential recombinant BCG-based HIV vaccines involves careful selection of the HIV antigen, expression vectors, promoters, BCG strain, preclinical animal models, influence of preexisting immunity, and safety issues, for the rational design of recombinant BCG:HIV vaccines to prevent HIV transmission in the general population. EXPERT COMMENTARY The three critical issues to be considered when developing a rBCG:HIV vaccine are codon optimization, antigen localization, and plasmid stability in vivo. The use of integrative expression vectors are likely to improve the mycobacterial vaccine stability and immunogenicity to develop not only recombinant BCG-based vaccines expressing second generation of HIV-1 immunogens but also other major pediatric pathogens to prime protective responses shortly following birth.
Collapse
Affiliation(s)
- Athina Kilpeläinen
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| | - Milena Maya-Hoyos
- b Chemistry Department, Faculty of Sciences , Universidad Nacional de Colombia, Ciudad Universitaria , Bogotá , Colombia
| | - Narcís Saubí
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| | - Carlos Y Soto
- b Chemistry Department, Faculty of Sciences , Universidad Nacional de Colombia, Ciudad Universitaria , Bogotá , Colombia
| | - Joan Joseph Munne
- a Catalan Center for HIV Vaccine Research and Development, AIDS Research Unit, Infectious Diseases Department, Hospital Clínic/IDIBAPS, School of Medicine , University of Barcelona , Barcelona , Spain
| |
Collapse
|
10
|
Liao TYA, Lau A, Sunil J, Hytönen V, Hmama Z. Expression of Exogenous Antigens in the Mycobacterium bovis BCG Vaccine via Non-genetic Surface Decoration with the Avidin-biotin System. J Vis Exp 2018. [PMID: 29443102 DOI: 10.3791/56421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Tuberculosis (TB) is a serious infectious disease and the only available vaccine M. bovis bacillus Calmette-Guérin (BCG) is safe and effective for protection against children's severe TB meningitis and some forms of disseminated TB, but fails to protect against pulmonary TB, which is the most prevalent form of the disease. Promising strategies to improve BCG currently rely either on its transformation with genes encoding immunodominant M. tuberculosis (Mtb)-specific antigens and/or complementation with genes encoding co-factors that would stimulate antigen presenting cells. Major limitations to these approaches include low efficiency, low stability, and the uncertain level of safety of expression vectors. In this study, we present an alternative approach to vaccine improvement, which consists of BCG complementation with exogenous proteins of interest on the surface of bacteria, rather than transformation with plasmids encoding corresponding genes. First, proteins of interest are expressed in fusion with monomeric avidin in standard E. coli expression systems and then used to decorate the surface of biotinylated BCG. Animal experiments using BCG surface decorated with surrogate ovalbumin antigen demonstrate that the modified bacterium is fully immunogenic and capable of inducing specific T cell responses. Altogether, the data presented here strongly support a novel and efficient method for reshaping the current BCG vaccine that replaces the laborious conventional approach of complementation with exogenous nucleic acids.
Collapse
Affiliation(s)
- Ting-Yu Angela Liao
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia
| | - Alice Lau
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia
| | - Joseph Sunil
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia
| | - Vesa Hytönen
- Institute of Biomedical Technology, University of Tampere
| | - Zakaria Hmama
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia;
| |
Collapse
|
11
|
Oliveira TL, Rizzi C, Dellagostin OA. Recombinant BCG vaccines: molecular features and their influence in the expression of foreign genes. Appl Microbiol Biotechnol 2017; 101:6865-6877. [PMID: 28779291 DOI: 10.1007/s00253-017-8439-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/13/2017] [Accepted: 07/15/2017] [Indexed: 01/17/2023]
Abstract
Recombinant Mycobacterium bovis BCG vaccines (rBCG) were first developed in the 1990s as a means of expressing antigens from multiple pathogens. This review examines the key structural factors of recombinant M. bovis that influence the expression of the heterologous antigens and the generation of genetic and functional stability in rBCG, which are crucial for inducing strong and lasting immune responses. The fundamental aim of this paper is to provide an overview of factors that affect the expression of recombinant proteins in BCG and the generation of the immune response against the target antigens, including mycobacterial promoters, location of foreign antigens, and stability of the vectors. The reporter systems that have been employed for evaluation of these molecular features in BCG are also reviewed here.
Collapse
Affiliation(s)
- Thaís Larré Oliveira
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Caroline Rizzi
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Odir Antônio Dellagostin
- Programa de Pós-Graduação em Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil. .,Unidade de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Campus Universitário, Caixa Postal 354, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
12
|
Rizzi C, Peiter AC, Oliveira TL, Seixas ACP, Leal KS, Hartwig DD, Seixas FK, Borsuk S, Dellagostin OA. Stable expression of Mycobacterium bovis antigen 85B in auxotrophic M. bovis bacillus Calmette-Guérin. Mem Inst Oswaldo Cruz 2017; 112:123-130. [PMID: 28177046 PMCID: PMC5293121 DOI: 10.1590/0074-02760160360] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/31/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Bovine tuberculosis (TB) is a zoonotic disease caused by Mycobacterium
bovis, responsible for causing major losses in livestock. A cost
effective alternative to control the disease could be herd vaccination. The
bacillus Calmette-Guérin (BCG) vaccine has a limited efficacy against bovine TB,
but can improved by over-expression of protective antigens. The M.
bovis antigen 85B demonstrates ability to induce protective immune
response against bovine TB in animal models. However, current systems for the
construction of recombinant BCG expressing multiple copies of the gene result in
strains of low genetic stability that rapidly lose the plasmid in vivo. Employing
antibiotic resistance as selective markers, these systems also compromise vaccine
safety. We previously reported the construction of a stable BCG expression system
using auxotrophic complementation as a selectable marker. OBJECTIVES The fundamental aim of this study was to construct strains of M.
bovis BCG Pasteur and the auxotrophic M. bovis BCG
ΔleuD expressing Ag85B and determine their stability in
vivo. METHODS Employing the auxotrophic system, we constructed rBCG strains that expressed
M. bovis Ag85B and compared their stability with a
conventional BCG strain in mice. Stability was measured in terms of bacterial
growth on the selective medium and retention of antigen expression. FINDINGS The auxotrophic complementation system was highly stable after 18 weeks, even
during in vivo growth, as the selective pressure and expression of antigen were
maintained comparing to the conventional vector. MAIN CONCLUSION The Ag85B continuous expression within the host may generate a stronger and
long-lasting immune response compared to conventional systems.
Collapse
Affiliation(s)
- Caroline Rizzi
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil
| | - Ana Carolina Peiter
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil
| | - Thaís Larré Oliveira
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil
| | - Amilton Clair Pinto Seixas
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil
| | - Karen Silva Leal
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil
| | - Daiane Drawanz Hartwig
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil.,Universidade Federal de Pelotas, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, RS, Brasil
| | - Fabiana Kommling Seixas
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil
| | - Sibele Borsuk
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil
| | - Odir Antônio Dellagostin
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Programa de Pós-Graduação em Biotecnologia, Pelotas, RS, Brasil
| |
Collapse
|
13
|
Mahant A, Saubi N, Eto Y, Guitart N, Gatell JM, Hanke T, Joseph J. Preclinical development of BCG.HIVA 2auxo.int, harboring an integrative expression vector, for a HIV-TB Pediatric vaccine. Enhancement of stability and specific HIV-1 T-cell immunity. Hum Vaccin Immunother 2017; 13:1798-1810. [PMID: 28426273 PMCID: PMC5557246 DOI: 10.1080/21645515.2017.1316911] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
One of the critical issues that should be addressed in the development of a BCG-based HIV vaccine is genetic plasmid stability. Therefore, to address this issue we have considered using integrative vectors and the auxotrophic mutant of BCG complemented with a plasmid carrying a wild-type complementing gene. In this study, we have constructed an integrative E. coli-mycobacterial shuttle plasmid, p2auxo.HIVAint, expressing the HIV-1 clade A immunogen HIVA. This shuttle vector uses an antibiotic resistance-free mechanism for plasmid selection and maintenance. It was first transformed into a glycine auxotrophic E. coli strain and subsequently transformed into a lysine auxotrophic Mycobacterium bovis BCG strain to generate the vaccine BCG.HIVA2auxo.int. Presence of the HIVA gene sequence and protein expression was confirmed. We demonstrated that the in vitro stability of the integrative plasmid p2auxo.HIVAint was increased 4-fold, as compared with the BCG strain harboring the episomal plasmid, and was genetically and phenotypically characterized. The BCG.HIVA2auxo.int vaccine in combination with modified vaccinia virus Ankara (MVA).HIVA was found to be safe and induced HIV-1 and Mycobacterium tuberculosis-specific interferon-γ-producing T-cell responses in adult BALB/c mice. We have engineered a more stable and immunogenic BCG-vectored vaccine using the prototype immunogen HIVA. Thus, the use of integrative expression vectors and the antibiotic-free plasmid selection system based on “double” auxotrophic complementation are likely to improve the mycobacterial vaccine stability in vivo and immunogenicity to develop not only recombinant BCG-based vaccines expressing second generation of HIV-1 immunogens but also other major pediatric pathogens to prime protective responses shortly following birth.
Collapse
Affiliation(s)
- Aakash Mahant
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Narcís Saubi
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Yoshiki Eto
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Núria Guitart
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Josep Ma Gatell
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| | - Tomáš Hanke
- b The Jenner Institute , University of Oxford , Oxford , UK
| | - Joan Joseph
- a AIDS Research Group, Hospital Clínic/IDIBAPS-HIVACAT, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain
| |
Collapse
|
14
|
Liao TYA, Lau A, Joseph S, Hytönen V, Hmama Z. Improving the Immunogenicity of the Mycobacterium bovis BCG Vaccine by Non-Genetic Bacterial Surface Decoration Using the Avidin-Biotin System. PLoS One 2015; 10:e0145833. [PMID: 26716832 PMCID: PMC4696857 DOI: 10.1371/journal.pone.0145833] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 12/09/2015] [Indexed: 12/18/2022] Open
Abstract
Current strategies to improve the current BCG vaccine attempt to over-express genes encoding specific M. tuberculosis (Mtb) antigens and/or regulators of antigen presentation function, which indeed have the potential to reshape BCG in many ways. However, these approaches often face serious difficulties, in particular the efficiency and stability of gene expression via nucleic acid complementation and safety concerns associated with the introduction of exogenous DNA. As an alternative, we developed a novel non-genetic approach for rapid and efficient display of exogenous proteins on bacterial cell surface. The technology involves expression of proteins of interest in fusion with a mutant version of monomeric avidin that has the feature of reversible binding to biotin. Fusion proteins are then used to decorate the surface of biotinylated BCG. Surface coating of BCG with recombinant proteins was highly reproducible and stable. It also resisted to the freeze-drying shock routinely used in manufacturing conventional BCG. Modifications of BCG surface did not affect its growth in culture media neither its survival within the host cell. Macrophages phagocytized coated BCG bacteria, which efficiently delivered their surface cargo of avidin fusion proteins to MHC class I and class II antigen presentation compartments. Thereafter, chimeric proteins corresponding to a surrogate antigen derived from ovalbumin and the Mtb specific ESAT6 antigen were generated and tested for immunogenicity in vaccinated mice. We found that BCG displaying ovalbumin antigen induces an immune response with a magnitude similar to that induced by BCG genetically expressing the same surrogate antigen. We also found that BCG decorated with Mtb specific antigen ESAT6 successfully induces the expansion of specific T cell responses. This novel technology, therefore, represents a practical and effective alternative to DNA-based gene expression for upgrading the current BCG vaccine.
Collapse
Affiliation(s)
- Ting-Yu Angela Liao
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Alice Lau
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Sunil Joseph
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Vesa Hytönen
- Institute of Biomedical Technology, University of Tampere, Tampere, Finland
| | - Zakaria Hmama
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
- * E-mail:
| |
Collapse
|
15
|
Zheng YQ, Naguib YW, Dong Y, Shi YC, Bou S, Cui Z. Applications of bacillus Calmette–Guerin and recombinant bacillus Calmette–Guerin in vaccine development and tumor immunotherapy. Expert Rev Vaccines 2015. [DOI: 10.1586/14760584.2015.1068124] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yuan-qiang Zheng
- 1Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
| | - Youssef W Naguib
- 2Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yixuan Dong
- 2Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yan-chun Shi
- 1Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
| | - Shorgan Bou
- 3National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, China
| | - Zhengrong Cui
- 1Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
- 2Pharmaceutics Division, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
16
|
Stable Expression of Lentiviral Antigens by Quality-Controlled Recombinant Mycobacterium bovis BCG Vectors. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:726-41. [PMID: 25924766 PMCID: PMC4478521 DOI: 10.1128/cvi.00075-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/22/2015] [Indexed: 12/14/2022]
Abstract
The well-established safety profile of the tuberculosis vaccine strain, Mycobacterium bovis bacille Calmette-Guérin (BCG), makes it an attractive vehicle for heterologous expression of antigens from clinically relevant pathogens. However, successful generation of recombinant BCG strains possessing consistent insert expression has encountered challenges in stability. Here, we describe a method for the development of large recombinant BCG accession lots which stably express the lentiviral antigens, human immunodeficiency virus (HIV) gp120 and simian immunodeficiency virus (SIV) Gag, using selectable leucine auxotrophic complementation. Successful establishment of vaccine stability stems from stringent quality control criteria which not only screen for highly stable complemented BCG ΔleuCD transformants but also thoroughly characterize postproduction quality. These parameters include consistent production of correctly sized antigen, retention of sequence-pure plasmid DNA, freeze-thaw recovery, enumeration of CFU, and assessment of cellular aggregates. Importantly, these quality assurance procedures were indicative of overall vaccine stability, were predictive for successful antigen expression in subsequent passaging both in vitro and in vivo, and correlated with induction of immune responses in murine models. This study has yielded a quality-controlled BCG ΔleuCD vaccine expressing HIV gp120 that retained stable full-length expression after 10(24)-fold amplification in vitro and following 60 days of growth in mice. A second vaccine lot expressed full-length SIV Gag for >10(68)-fold amplification in vitro and induced potent antigen-specific T cell populations in vaccinated mice. Production of large, well-defined recombinant BCG ΔleuCD lots can allow confidence that vaccine materials for immunogenicity and protection studies are not negatively affected by instability or differences between freshly grown production batches.
Collapse
|
17
|
Begnini KR, Buss JH, Collares T, Seixas FK. Recombinant Mycobacterium bovis BCG for immunotherapy in nonmuscle invasive bladder cancer. Appl Microbiol Biotechnol 2015; 99:3741-54. [DOI: 10.1007/s00253-015-6495-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 02/17/2015] [Accepted: 02/19/2015] [Indexed: 02/07/2023]
|
18
|
Zheng YQ, Naguib YW, Dong Y, Shi YC, Bou S, Cui Z. Applications of bacillus Calmette-Guerin and recombinant bacillus Calmette-Guerin in vaccine development and tumor immunotherapy. Expert Rev Vaccines 2015; 14:1255-75. [PMID: 26268434 PMCID: PMC4920355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Bacillus Calmette-Guerin (BCG) vaccines are attenuated live strains of Mycobacterium bovis and are among the most widely used vaccines in the world. BCG is proven to be effective in preventing severe infant meningitis and miliary tuberculosis. Intravesical instillation of BCG is also a standard treatment for non-muscle invasive bladder cancer. In the past few decades, recombinant BCG (rBCG) technology had been extensively applied to develop vaccine candidates for a variety of infectious diseases, including bacterial, viral, and parasite infections, and to improve the efficacy of BCG in bladder cancer therapy. This review is intended to show the vast applications of BCG and recombinant BCG (rBCG) in the prevention of infectious diseases and cancer immunotherapy, with a special emphasis on recent approaches and trends on both pre-clinical and clinical levels.
Collapse
Affiliation(s)
- Yuan-qiang Zheng
- Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, China
| | - Youssef W. Naguib
- Pharmaceutics Division, College of Pharmacy, the University of Texas at Austin, Austin, TX 78712, USA
| | - Yixuan Dong
- Pharmaceutics Division, College of Pharmacy, the University of Texas at Austin, Austin, TX 78712, USA
| | - Yan-chun Shi
- Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
| | - Shorgan Bou
- National Research Center for Animal Transgenic Biotechnology, Inner Mongolia University, Hohhot, China
| | - Zhengrong Cui
- Inner Mongolia Key Laboratory of Molecular Biology, Inner Mongolia Medical University, Hohhot 010059, China
- Pharmaceutics Division, College of Pharmacy, the University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
19
|
Belardinelli JM, Larrouy-Maumus G, Jones V, Sorio de Carvalho LP, McNeil MR, Jackson M. Biosynthesis and translocation of unsulfated acyltrehaloses in Mycobacterium tuberculosis. J Biol Chem 2014; 289:27952-65. [PMID: 25124040 PMCID: PMC4183827 DOI: 10.1074/jbc.m114.581199] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 08/12/2014] [Indexed: 11/25/2022] Open
Abstract
A number of species-specific polymethyl-branched fatty acid-containing trehalose esters populate the outer membrane of Mycobacterium tuberculosis. Among them, 2,3-diacyltrehaloses (DAT) and penta-acyltrehaloses (PAT) not only play a structural role in the cell envelope but also contribute to the ability of M. tuberculosis to multiply and persist in the infected host, promoting the intracellular survival of the bacterium and modulating host immune responses. The nature of the machinery, topology, and sequential order of the reactions leading to the biosynthesis, assembly, and export of these complex glycolipids to the cell surface are the object of the present study. Our genetic and biochemical evidence corroborates a model wherein the biosynthesis and translocation of DAT and PAT to the periplasmic space are coupled and topologically split across the plasma membrane. The formation of DAT occurs on the cytosolic face of the plasma membrane through the action of PapA3, FadD21, and Pks3/4; that of PAT occurs on the periplasmic face via transesterification reactions between DAT substrates catalyzed by the acyltransferase Chp2 (Rv1184c). The integral membrane transporter MmpL10 is essential for DAT to reach the cell surface, and its presence in the membrane is required for Chp2 to be active. Disruption of mmpL10 or chp2 leads to an important build-up of DAT inside the cells and to the formation of a novel form of unsulfated acyltrehalose esterified with polymethyl-branched fatty acids normally found in sulfolipids that is translocated to the cell surface.
Collapse
Affiliation(s)
- Juan Manuel Belardinelli
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682 and
| | - Gérald Larrouy-Maumus
- the Division of Mycobacterial Research, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Victoria Jones
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682 and
| | - Luiz Pedro Sorio de Carvalho
- the Division of Mycobacterial Research, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Michael R McNeil
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682 and
| | - Mary Jackson
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682 and
| |
Collapse
|
20
|
Chapman R, Bourn WR, Shephard E, Stutz H, Douglass N, Mgwebi T, Meyers A, Chin'ombe N, Williamson AL. The use of directed evolution to create a stable and immunogenic recombinant BCG expressing a modified HIV-1 Gag antigen. PLoS One 2014; 9:e103314. [PMID: 25061753 PMCID: PMC4111510 DOI: 10.1371/journal.pone.0103314] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/28/2014] [Indexed: 01/09/2023] Open
Abstract
Numerous features make Mycobacterium bovis BCG an attractive vaccine vector for HIV. It has a good safety profile, it elicits long-lasting cellular immune responses and in addition manufacturing costs are affordable. Despite these advantages it is often difficult to express viral antigens in BCG, which results in genetic instability and low immunogenicity. The aim of this study was to generate stable recombinant BCG (rBCG) that express high levels of HIV antigens, by modification of the HIV genes. A directed evolution process was applied to recombinant mycobacteria that expressed HIV-1 Gag fused to the green fluorescent protein (GFP). Higher growth rates and increased GFP expression were selected for. Through this process a modified Gag antigen was selected. Recombinant BCG that expressed the modified Gag (BCG[pWB106] and BCG[pWB206]) were more stable, produced higher levels of antigen and grew faster than those that expressed the unmodified Gag (BCG[pWB105]). The recombinant BCG that expressed the modified HIV-1 Gag induced 2 to 3 fold higher levels of Gag-specific CD4 T cells than those expressing the unmodified Gag (BCG[pWB105]). Mice primed with 107 CFU BCG[pWB206] and then boosted with MVA-Gag developed Gag-specific CD8 T cells with a frequency of 1343±17 SFU/106 splenocytes, 16 fold greater than the response induced with MVA-Gag alone. Levels of Gag-specific CD4 T cells were approximately 5 fold higher in mice primed with BCG[pWB206] and boosted with MVA-Gag than in those receiving the MVA-Gag boost alone. In addition mice vaccinated with BCG[pWB206] were protected from a surrogate vaccinia virus challenge.
Collapse
Affiliation(s)
- Rosamund Chapman
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- * E-mail:
| | - William R. Bourn
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Enid Shephard
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Medical Research Council, Cape Town, South Africa
- Department of Medicine Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Helen Stutz
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nicola Douglass
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Thandi Mgwebi
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ann Meyers
- Department of Molecular and Cell Biology, Faculty Of Science, University of Cape Town, Cape Town, South Africa
| | - Nyasha Chin'ombe
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Anna-Lise Williamson
- Division of Virology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Services, Cape Town, South Africa
| |
Collapse
|
21
|
Role for Gr-1+ cells in the control of high-dose Mycobacterium bovis recombinant BCG. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:1120-7. [PMID: 24920602 DOI: 10.1128/cvi.00363-14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mycobacterium bovis bacillus Calmette-Guérin (BCG) is an attractive target for development as a live vaccine vector delivering transgenic antigens from HIV and other pathogens. Most studies aimed at defining the clearance of BCG have been performed at doses between 10(2) and 10(4) CFU. Interestingly, however, recombinant BCG (rBCG) administered at doses of >10(6) CFU effectively generates antigen-specific T-cell responses and primes for heterologous boost responses. Thus, defining clearance at high doses might aid in the optimization of rBCG as a vector. In this study, we used bioluminescence imaging to examine the kinetics of rBCG transgene expression and clearance in mice immunized with 5 × 10(7) CFU rBCG expressing luciferase. Similar to studies using low-dose rBCG, our results demonstrate that the adaptive immune response is necessary for long-term control of rBCG beginning 9 days after immunizing mice. However, in contrast to these reports, we observed that the majority of mycobacterial antigen was eliminated prior to day 9. By examining knockout and antibody-mediated depletion mouse models, we demonstrate that the rapid clearance of rBCG occurs in the first 24 h and is mediated by Gr-1(+) cells. As Gr-1(+) granulocytes have been described as having no impact on BCG clearance at low doses, our results reveal an unappreciated role for Gr-1(+) neutrophils and inflammatory monocytes in the clearance of high-dose rBCG. This work demonstrates the potential of applying bioluminescence imaging to rBCG in order to gain an understanding of the immune response and increase the efficacy of rBCG as a vaccine vector.
Collapse
|
22
|
Saubi N, Gea-Mallorquí E, Ferrer P, Hurtado C, Sánchez-Úbeda S, Eto Y, Gatell JM, Hanke T, Joseph J. Engineering new mycobacterial vaccine design for HIV-TB pediatric vaccine vectored by lysine auxotroph of BCG. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14017. [PMID: 26015961 PMCID: PMC4362382 DOI: 10.1038/mtm.2014.17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/26/2014] [Indexed: 02/05/2023]
Abstract
In this study, we have engineered a new mycobacterial vaccine design by using an antibiotic-free plasmid selection system. We assembled a novel Escherichia coli (E. coli)–mycobacterial shuttle plasmid p2auxo.HIVA, expressing the HIV-1 clade A immunogen HIVA. This shuttle vector employs an antibiotic resistance-free mechanism for plasmid selection and maintenance based on glycine complementation in E. coli and lysine complementation in mycobacteria. This plasmid was first transformed into glycine auxotroph of E. coli strain and subsequently transformed into lysine auxotroph of Mycobacterium bovis BCG strain to generate vaccine BCG.HIVA2auxo. We demonstrated that the episomal plasmid p2auxo.HIVA was stable in vivo over a 7-week period and genetically and phenotypically characterized the BCG.HIVA2auxo vaccine strain. The BCG.HIVA2auxo vaccine in combination with modified vaccinia virus Ankara (MVA). HIVA was safe and induced HIV-1 and Mycobacterium tuberculosis-specific interferon-γ-producing T-cell responses in adult BALB/c mice. Polyfunctional HIV-1-specific CD8+ T cells, which produce interferon-γ and tumor necrosis factor-α and express the degranulation marker CD107a, were induced. Thus, we engineered a novel, safer, good laboratory practice–compatible BCG-vectored vaccine using prototype immunogen HIVA. This antibiotic-free plasmid selection system based on “double” auxotrophic complementation might be a new mycobacterial vaccine platform to develop not only recombinant BCG-based vaccines expressing second generation of HIV-1 immunogens but also other major pediatric pathogens to prime protective response soon after birth.
Collapse
Affiliation(s)
- Narcís Saubi
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Ester Gea-Mallorquí
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Pau Ferrer
- Department of Chemical Engineering, Group of Bioprocess Engineering and Applied Biocatalysis, School of Engineering, Autonomous University of Barcelona , Barcelona, Catalonia, Spain
| | - Carmen Hurtado
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Sara Sánchez-Úbeda
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Yoshiki Eto
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Josep M Gatell
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| | - Tomáš Hanke
- The Jenner Institute, University of Oxford , Oxford, UK ; MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford , Oxford, UK
| | - Joan Joseph
- AIDS Research Group, Hospital Clinic/HIVACAT, School of Medicine, University of Barcelona , Barcelona, Catalonia, Spain
| |
Collapse
|
23
|
Protective immunity induced by a recombinant BCG vaccine encoding the cyclophilin gene of Toxoplasma gondii. Vaccine 2013; 31:6065-71. [DOI: 10.1016/j.vaccine.2013.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 09/14/2013] [Accepted: 10/03/2013] [Indexed: 01/18/2023]
|
24
|
Doz E, Lombard R, Carreras F, Buzoni-Gatel D, Winter N. Mycobacteria-Infected Dendritic Cells Attract Neutrophils That Produce IL-10 and Specifically Shut Down Th17 CD4 T Cells through Their IL-10 Receptor. THE JOURNAL OF IMMUNOLOGY 2013; 191:3818-26. [DOI: 10.4049/jimmunol.1300527] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
25
|
Lecoeur H, Giraud E, Prévost MC, Milon G, Lang T. Reprogramming neutral lipid metabolism in mouse dendritic leucocytes hosting live Leishmania amazonensis amastigotes. PLoS Negl Trop Dis 2013; 7:e2276. [PMID: 23785538 PMCID: PMC3681733 DOI: 10.1371/journal.pntd.0002276] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 05/06/2013] [Indexed: 11/18/2022] Open
Abstract
Background After loading with live Leishmania (L) amazonensis amastigotes, mouse myeloid dendritic leucocytes/DLs are known to undergo reprogramming of their immune functions. In the study reported here, we investigated whether the presence of live L. amazonensis amastigotes in mouse bone marrow-derived DLs is able to trigger re-programming of DL lipid, and particularly neutral lipid metabolism. Methodology/Principal Findings Affymetrix-based transcriptional profiles were determined in C57BL/6 and DBA/2 mouse bone marrow-derived DLs that had been sorted from cultures exposed or not to live L. amazonensis amastigotes. This showed that live amastigote-hosting DLs exhibited a coordinated increase in: (i) long-chain fatty acids (LCFA) and cholesterol uptake/transport, (ii) LCFA and cholesterol (re)-esterification to triacyl-sn-glycerol (TAG) and cholesteryl esters (CE), respectively. As these neutral lipids are known to make up the lipid body (LB) core, oleic acid was added to DL cultures and LB accumulation was compared in live amastigote-hosting versus amastigote-free DLs by epi-fluorescence and transmission electron microscopy. This showed that LBs were both significantly larger and more numerous in live amastigote-hosting mouse dendritic leucocytes. Moreover, many of the larger LB showed intimate contact with the membrane of the parasitophorous vacuoles hosting the live L. amazonensis amastigotes. Conclusions/Significance As leucocyte LBs are known to be more than simple neutral lipid repositories, we set about addressing two related questions. Could LBs provide lipids to live amastigotes hosted within the DL parasitophorous vacuole and also deliver? Could LBs impact either directly or indirectly on the persistence of L. amazonensis amastigotes in rodent skin? Once they have gained entry to mammals, live Leishmania (L) amazonensis amastigotes are known to subvert both macrophages and dendritic leucocytes (DLs) as host cells. These L. amazonensis amastigotes then may or may not proliferate in these two phagocytic leucocyte lineages, but in both cases the otherwise versatile differentiation program of these lineages is known to be rapidly remodeled. Here, we describe the rapid reprogramming of C57BL/6 and DBA/2 mouse bone marrow-derived DLs, with a special focus on cytosolic lipid bodies (LBs) that are known to store neutral lipids such as triacyl-sn-glycerol (TAG) and cholesteryl esters (CE). After extracting RNA from carefully sorted amastigote-free DLs and L. amazonensis amastigote-hosting DLs, an Affymetrix-based analysis clearly showed a singular and coordinated increase in DL transcripts involved in (i) long-chain fatty acid uptake, transport and esterification to TAG and (ii) cholesterol uptake and esterification to cholesteryl esters. Oleic acid was added to check that neutral lipid metabolism was both rapidly increased and reprogrammed in amastigote-hosting DLs. It should be noted that the LBs in live amastigote-hosting DLs were more numerous, and that the largest of these LBs were in contact with live amastigote- hosting parasitophorous vacuoles. We further discuss these findings in the context of live L. amazonensis amastigote-rodent host interactions.
Collapse
Affiliation(s)
- Hervé Lecoeur
- Institut Pasteur, Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Paris, France
- * E-mail: (HL); (TL)
| | - Emilie Giraud
- Institut Pasteur, Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Paris, France
| | - Marie-Christine Prévost
- Institut Pasteur, Département Biologie Cellulaire et Infection, Plateforme de Microscopie Ultrastructurale, Paris, France
| | - Geneviève Milon
- Institut Pasteur, Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Paris, France
| | - Thierry Lang
- Institut Pasteur, Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Paris, France
- Institut Pasteur, Département Infection et Epidémiologie, Laboratoire des Processus Infectieux à Trypanosomatidés, Paris, France
- * E-mail: (HL); (TL)
| |
Collapse
|
26
|
Giraud E, Lecoeur H, Soubigou G, Coppée JY, Milon G, Prina E, Lang T. Distinct transcriptional signatures of bone marrow-derived C57BL/6 and DBA/2 dendritic leucocytes hosting live Leishmania amazonensis amastigotes. PLoS Negl Trop Dis 2012; 6:e1980. [PMID: 23272268 PMCID: PMC3521701 DOI: 10.1371/journal.pntd.0001980] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 11/05/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/OBJECTIVES The inoculation of a low number (10(4)) of L. amazonensis metacyclic promastigotes into the dermis of C57BL/6 and DBA/2 mouse ear pinna results in distinct outcome as assessed by the parasite load values and ear pinna macroscopic features monitored from days 4 to 22-phase 1 and from days 22 to 80/100-phase 2. While in C57BL/6 mice, the amastigote population size was increasing progressively, in DBA/2 mice, it was rapidly controlled. This latter rapid control did not prevent intracellular amastigotes to persist in the ear pinna and in the ear-draining lymph node/ear-DLN. The objectives of the present analysis was to compare the dendritic leukocytes-dependant immune processes that could account for the distinct outcome during the phase 1, namely, when phagocytic dendritic leucocytes of C57BL/6 and DBA/2 mice have been subverted as live amastigotes-hosting cells. METHODOLOGY/PRINCIPAL FINDINGS Being aware of the very low frequency of the tissues' dendritic leucocytes/DLs, bone marrow-derived C57BL/6 and DBA/2 DLs were first generated and exposed or not to live DsRed2 expressing L. amazonensis amastigotes. Once sorted from the four bone marrow cultures, the DLs were compared by Affymetrix-based transcriptomic analyses and flow cytometry. C57BL/6 and DBA/2 DLs cells hosting live L. amazonensis amastigotes do display distinct transcriptional signatures and markers that could contribute to the distinct features observed in C57BL/6 versus DBA/2 ear pinna and in the ear pinna-DLNs during the first phase post L. amazonensis inoculation. CONCLUSIONS/SIGNIFICANCE The distinct features captured in vitro from homogenous populations of C57BL/6 and DBA/2 DLs hosting live amastigotes do offer solid resources for further comparing, in vivo, in biologically sound conditions, functions that range from leukocyte mobilization within the ear pinna, the distinct emigration from the ear pinna to the DLN of live amastigotes-hosting DLs, and their unique signalling functions to either naive or primed T lymphocytes.
Collapse
Affiliation(s)
- Emilie Giraud
- Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Institut Pasteur, Paris, France
| | - Hervé Lecoeur
- Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Institut Pasteur, Paris, France
| | | | - Jean-Yves Coppée
- Plateforme Transcriptome et Epigénome, Institut Pasteur, Paris, France
| | - Geneviève Milon
- Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Institut Pasteur, Paris, France
| | - Eric Prina
- Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Institut Pasteur, Paris, France
| | - Thierry Lang
- Département de Parasitologie et Mycologie, Laboratoire Immunophysiologie et Parasitisme, Institut Pasteur, Paris, France
- * E-mail:
| |
Collapse
|
27
|
Rizzi C, Bianco MV, Blanco FC, Soria M, Gravisaco MJ, Montenegro V, Vagnoni L, Buddle B, Garbaccio S, Delgado F, Leal KS, Cataldi AA, Dellagostin OA, Bigi F. Vaccination with a BCG strain overexpressing Ag85B protects cattle against Mycobacterium bovis challenge. PLoS One 2012; 7:e51396. [PMID: 23251517 PMCID: PMC3519572 DOI: 10.1371/journal.pone.0051396] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 10/31/2012] [Indexed: 12/29/2022] Open
Abstract
Mycobacterium bovis is the causative agent of tuberculosis in cattle but also infects other animals, including humans. Previous studies in cattle have demonstrated that the protection induced by BCG is not complete. In order to improve the protection efficacy of BCG, in this study we overexpressed Ag85B in a BCG Pasteur strain, by using an expression system based on the use of an auxotrophic strain for the leucine amino acid, and complementation with leuD. We found that vaccination of cattle with BCG overexpressing Ag85B induced higher production of IL-17 and IL-4 mRNA upon purified protein derivative (PPDB) stimulation of peripheral blood mononuclear cells (PBMCs) than vaccination with BCG. Moreover, the IL-17 mRNA expression after vaccination negatively correlated with disease severity resulting from a subsequent challenge with M. bovis, suggesting that this cytokine is a potential biomarker of cattle protection against bovine tuberculosis. Importantly, vaccination with the recombinant BCG vaccine protected cattle better than the wild-type BCG Pasteur.
Collapse
Affiliation(s)
- Caroline Rizzi
- Núcleo de Biotecnologia, CDTec, Universidade Federal de Pelotas, Pelotas, Brazil
| | - María Verónica Bianco
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| | - Federico Carlos Blanco
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| | - Marcelo Soria
- Microbiología Agrícola, Facultad de Agronomía, Universidad de Buenos Aires, INBA-CONICET, Ciudad de Buenos Aires, Argentina
| | - María José Gravisaco
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| | - Valeria Montenegro
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| | - Lucas Vagnoni
- Instituto de Patobiología, CICVyA- INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| | - Bryce Buddle
- AgResearch, Hopkirk Research Institute, Palmerston North, New Zealand
| | - Sergio Garbaccio
- Instituto de Patobiología, CICVyA- INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| | - Fernando Delgado
- Instituto de Patobiología, CICVyA- INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| | - Karen Silva Leal
- Núcleo de Biotecnologia, CDTec, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Angel Adrián Cataldi
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| | | | - Fabiana Bigi
- Instituto de Biotecnología, CICVyA-INTA, N. Repetto y De los Reseros, Buenos Aires, Argentina
| |
Collapse
|
28
|
Jung C, Meinzer U, Montcuquet N, Thachil E, Château D, Thiébaut R, Roy M, Alnabhani Z, Berrebi D, Dussaillant M, Pedruzzi E, Thenet S, Cerf-Bensussan N, Hugot JP, Barreau F. Yersinia pseudotuberculosis disrupts intestinal barrier integrity through hematopoietic TLR-2 signaling. J Clin Invest 2012; 122:2239-51. [PMID: 22565313 DOI: 10.1172/jci58147] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 03/28/2012] [Indexed: 01/01/2023] Open
Abstract
Intestinal barrier function requires intricate cooperation between intestinal epithelial cells and immune cells. Enteropathogens are able to invade the intestinal lymphoid tissue known as Peyer's patches (PPs) and disrupt the integrity of the intestinal barrier. However, the underlying molecular mechanisms of this process are poorly understood. In mice infected with Yersinia pseudotuberculosis, we found that PP barrier dysfunction is dependent on the Yersinia virulence plasmid and the expression of TLR-2 by hematopoietic cells, but not by intestinal epithelial cells. Upon TLR-2 stimulation, Y. pseudotuberculosis-infected monocytes activated caspase-1 and produced IL-1β. In turn, IL-1β increased NF-κB and myosin light chain kinase activation in intestinal epithelial cells, thus disrupting the intestinal barrier by opening the tight junctions. Therefore, Y. pseudotuberculosis subverts intestinal barrier function by altering the interplay between immune and epithelial cells during infection.
Collapse
|
29
|
de la Paz Santangelo M, Gest PM, Guerin ME, Coinçon M, Pham H, Ryan G, Puckett SE, Spencer JS, Gonzalez-Juarrero M, Daher R, Lenaerts AJ, Schnappinger D, Therisod M, Ehrt S, Sygusch J, Jackson M. Glycolytic and non-glycolytic functions of Mycobacterium tuberculosis fructose-1,6-bisphosphate aldolase, an essential enzyme produced by replicating and non-replicating bacilli. J Biol Chem 2011; 286:40219-31. [PMID: 21949126 PMCID: PMC3220552 DOI: 10.1074/jbc.m111.259440] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 09/09/2011] [Indexed: 12/29/2022] Open
Abstract
The search for antituberculosis drugs active against persistent bacilli has led to our interest in metallodependent class II fructose-1,6-bisphosphate aldolase (FBA-tb), a key enzyme of gluconeogenesis absent from mammalian cells. Knock-out experiments at the fba-tb locus indicated that this gene is required for the growth of Mycobacterium tuberculosis on gluconeogenetic substrates and in glucose-containing medium. Surface labeling and enzymatic activity measurements revealed that this enzyme was exported to the cell surface of M. tuberculosis and produced under various axenic growth conditions including oxygen depletion and hence by non-replicating bacilli. Importantly, FBA-tb was also produced in vivo in the lungs of infected guinea pigs and mice. FBA-tb bound human plasmin(ogen) and protected FBA-tb-bound plasmin from regulation by α(2)-antiplasmin, suggestive of an involvement of this enzyme in host/pathogen interactions. The crystal structures of FBA-tb in the native form and in complex with a hydroxamate substrate analog were determined to 2.35- and 1.9-Å resolution, respectively. Whereas inhibitor attachment had no effect on the plasminogen binding activity of FBA-tb, it competed with the natural substrate of the enzyme, fructose 1,6-bisphosphate, and substantiated a previously unknown reaction mechanism associated with metallodependent aldolases involving recruitment of the catalytic zinc ion by the substrate upon active site binding. Altogether, our results highlight the potential of FBA-tb as a novel therapeutic target against both replicating and non-replicating bacilli.
Collapse
Affiliation(s)
- Maria de la Paz Santangelo
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682
- Instituto de Biotecnología, Centro de Investigación en Ciencias Veterinarias y Agronómicas-Instituto Nacional de Tecnología Agropecuaria, 1686 Buenos Aires, Argentina
| | - Petra M. Gest
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682
| | - Marcelo E. Guerin
- Unidad de Biofísica, Centro Mixto Consejo Superior de Investigaciones Científicas-Universidad del País Vasco/Euskal Herriko Unibertsitatea (CSIC-UPV/EHU), Barrio Sarriena s/n, Leioa, 48940 Bizkaia, Spain
- Departamento de Bioquímica, Universidad del País Vasco, Aptdo. 644, 48080 Bilbao, Spain
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Mathieu Coinçon
- Département de Biochimie, Université de Montréal, CP 6128, Station centre-ville, Montréal PQ H3C 3J7, Canada
| | - Ha Pham
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682
| | - Gavin Ryan
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682
| | - Susan E. Puckett
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York 10065
| | - John S. Spencer
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682
| | - Mercedes Gonzalez-Juarrero
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682
| | - Racha Daher
- Laboratoire de Chimie Bioorganique et Bioinorganique-Institut de Chimie Moléculaire et des Matériaux d'Orsay, UMR 8182, Université Paris Sud, 91405 Orsay, France and
| | - Anne J. Lenaerts
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682
| | - Dirk Schnappinger
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York 10065
| | - Michel Therisod
- Laboratoire de Chimie Bioorganique et Bioinorganique-Institut de Chimie Moléculaire et des Matériaux d'Orsay, UMR 8182, Université Paris Sud, 91405 Orsay, France and
| | - Sabine Ehrt
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York 10065
| | - Jurgen Sygusch
- Département de Biochimie, Université de Montréal, CP 6128, Station centre-ville, Montréal PQ H3C 3J7, Canada
| | - Mary Jackson
- From the Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523-1682
| |
Collapse
|
30
|
Nurul AA, Norazmi MN. Immunogenicity and in vitro protective efficacy of recombinant Mycobacterium bovis bacille Calmette Guerin (rBCG) expressing the 19 kDa merozoite surface protein-1 (MSP-1(19)) antigen of Plasmodium falciparum. Parasitol Res 2011; 108:887-97. [PMID: 21057812 DOI: 10.1007/s00436-010-2130-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 10/13/2010] [Indexed: 10/18/2022]
Abstract
Vaccine development against the blood-stage malaria parasite is aimed at reducing the pathology of the disease. We constructed a recombinant Mycobacterium bovis bacille Calmette Guerin (rBCG) expressing the 19 kDa C-terminus of Plasmodium falciparum merozoite surface protein-1 (MSP-1(19)) to evaluate its protective ability against merozoite invasion of red blood cells in vitro. A mutated version of MSP-1(19), previously shown to induce the production of inhibitory but not blocking antibodies, was cloned into a suitable shuttle plasmid and transformed into BCG Japan (designated rBCG016). A native version of the molecule was also cloned into BCG (rBCG026). Recombinant BCG expressing the mutated version of MSP-1(19) (rBCG016) elicited enhanced specific immune response against the epitope in BALB/c mice as compared to rBCG expressing the native version of the epitope (rBCG026). Sera from rBCG016-immunized mice contained significant levels of specific IgG, especially of the IgG2a subclass, against MSP-1(19) as determined by enzyme-linked immunosorbent assay. The sera was reactive with fixed P. falciparum merozoites as demonstrated by indirect immunofluorescence assay (IFA) and inhibited merozoite invasion of erythrocytes in vitro. Furthermore, lymphocytes from rBCG016-immunized mice demonstrated higher proliferative response against the MSP-1(19) antigen as compared to those of rBCG026- and BCG-immunized animals. rBCG expressing the mutated version of MSP-1(19) of P. falciparum induced enhanced humoral and cellular responses against the parasites paving the way for the rational use of rBCG as a blood-stage malaria vaccine candidate.
Collapse
Affiliation(s)
- Asma Abdullah Nurul
- School of Dental Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | | |
Collapse
|
31
|
Expression of MPB83 from Mycobacterium bovis in Brucella abortus S19 induces specific cellular immune response against the recombinant antigen in BALB/c mice. Microbes Infect 2010; 12:1236-43. [PMID: 20888425 DOI: 10.1016/j.micinf.2010.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 09/07/2010] [Accepted: 09/14/2010] [Indexed: 11/20/2022]
Abstract
Immunodominant MPB83 antigen from Mycobacterium bovis was expressed as a chimeric protein fused to either β-galactosidase, outer membrane lipoprotein OMP19 or periplasmic protein BP26 in gram-negative Brucella abortus S19, in all cases driven by each gene's own promoter. All fusion proteins were successfully expressed and localized in the expected subcellular fraction. Moreover, OMP19-MPB83 was processed as a lipoprotein when expressed in B. abortus. Splenocytes from BALB/c mice immunized with the recombinant S19 strains carrying the genes coding for the heterologous antigens in replicative plasmids, showed equally specific INF-γ production in response to MPB83 stimulation. Association to the lipid moiety of OMP19 presented no advantage in terms of immunogenicity for MPB83. In contrast, fusion to BP26, which was encoded by an integrative plasmid, resulted in a weaker immune response. None of the constructions affected the survival rate or the infection pattern of Brucella. We concluded that B. abortus S19 is an appropriate candidate for the expression of M. bovis antigens both associated to the membrane or cytosolic fraction and may provide the basis for a future combined vaccine for bovine brucellosis and tuberculosis.
Collapse
|
32
|
Abstract
Elucidating the function of mycobacterial proteins, determining their contribution to virulence, and developing new vaccine candidates has been facilitated by systems permitting the heterologous expression of genes in mycobacteria. Mycobacterium bovis bacille Calmette Guérin (BCG) and Mycobacterium smegmatis have commonly been employed as host systems for the heterologous expression of mycobacterial genes as well as genes from other bacteria, viruses, and mammalian cells. Vectors that permit strong, constitutive expression of genes have been developed, and more recently systems that allow tightly regulated induction of gene expression have become available. In this chapter, we describe two complementary techniques relevant to the field of gene expression in mycobacteria. We first outline the methodology used for the expression and specific detection of recombinant products expressed in BCG. The expression vectors described use an epitope tag fused to the C-terminal end of the foreign protein, ablating the need for additional reagents to detect the recombinant product. Second, we describe the inducible expression of genes in recombinant M. smegmatis and the subsequent purification of gene products using affinity chromatography.
Collapse
|
33
|
Andreu N, Zelmer A, Fletcher T, Elkington PT, Ward TH, Ripoll J, Parish T, Bancroft GJ, Schaible U, Robertson BD, Wiles S. Optimisation of bioluminescent reporters for use with mycobacteria. PLoS One 2010; 5:e10777. [PMID: 20520722 PMCID: PMC2875389 DOI: 10.1371/journal.pone.0010777] [Citation(s) in RCA: 245] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 04/27/2010] [Indexed: 01/01/2023] Open
Abstract
Background Mycobacterium tuberculosis, the causative agent of tuberculosis, still represents a major public health threat in many countries. Bioluminescence, the production of light by luciferase-catalyzed reactions, is a versatile reporter technology with multiple applications both in vitro and in vivo. In vivo bioluminescence imaging (BLI) represents one of its most outstanding uses by allowing the non-invasive localization of luciferase-expressing cells within a live animal. Despite the extensive use of luminescent reporters in mycobacteria, the resultant luminescent strains have not been fully applied to BLI. Methodology/Principal Findings One of the main obstacles to the use of bioluminescence for in vivo imaging is the achievement of reporter protein expression levels high enough to obtain a signal that can be detected externally. Therefore, as a first step in the application of this technology to the study of mycobacterial infection in vivo, we have optimised the use of firefly, Gaussia and bacterial luciferases in mycobacteria using a combination of vectors, promoters, and codon-optimised genes. We report for the first time the functional expression of the whole bacterial lux operon in Mycobacterium tuberculosis and M. smegmatis thus allowing the development of auto-luminescent mycobacteria. We demonstrate that the Gaussia luciferase is secreted from bacterial cells and that this secretion does not require a signal sequence. Finally we prove that the signal produced by recombinant mycobacteria expressing either the firefly or bacterial luciferases can be non-invasively detected in the lungs of infected mice by bioluminescence imaging. Conclusions/Significance While much work remains to be done, the finding that both firefly and bacterial luciferases can be detected non-invasively in live mice is an important first step to using these reporters to study the pathogenesis of M. tuberculosis and other mycobacterial species in vivo. Furthermore, the development of auto-luminescent mycobacteria has enormous ramifications for high throughput mycobacterial drug screening assays which are currently carried out either in a destructive manner using LuxAB or the firefly luciferase.
Collapse
Affiliation(s)
- Nuria Andreu
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Andrea Zelmer
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Taryn Fletcher
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Paul T. Elkington
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Theresa H. Ward
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Jorge Ripoll
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Tanya Parish
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Infectious Diseases Research Institute, Seattle, Washington, United States of America
| | - Gregory J. Bancroft
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ulrich Schaible
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Molecular Infection Research, Research Center Borstel, Borstel, Germany
| | | | - Siouxsie Wiles
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
34
|
Lecoeur H, de La Llave E, Osorio Y Fortéa J, Goyard S, Kiefer-Biasizzo H, Balazuc AM, Milon G, Prina E, Lang T. Sorting of Leishmania-bearing dendritic cells reveals subtle parasite-induced modulation of host-cell gene expression. Microbes Infect 2010; 12:46-54. [PMID: 19786115 DOI: 10.1016/j.micinf.2009.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 09/04/2009] [Accepted: 09/20/2009] [Indexed: 11/26/2022]
Abstract
Once in the mouse skin, Leishmania (L) amazonensis amastigotes are hosted by professional mononuclear phagocytes such as dendritic cells (DCs). When monitored after parasite inoculation, the frequency of amastigote-hosting DCs is very low (<1%) in both the skin and skin-draining lymph nodes. Therefore, we designed and validated an efficient procedure to purify live amastigotes-hosting DCs with the objective to facilitate quantitative and qualitative analysis of such rare cells. To this end, a L. amazonensis transgenic parasite expressing DsRed2 fluorescent protein was generated and added to mouse bone marrow-derived DC cultures. Then, a high speed sorting procedure, performed in BSL-2 containment, was setup to pick out only DCs hosting live amastigotes. This study reveals, for the first time, a unique transcript pattern from sorted live amastigotes-hosting DCs that would have been undetectable in unsorted samples. It was indeed possible to highlight a significant and coordinated up-regulation of L-arginine transporter and arginase2 transcripts in Leishmania-hosting DCs compared to un-parasitized DCs. These results indicate that arginine catabolism for polyamine generation is dominating over L-arginine catabolism for NO generation. In conclusion, this approach provides a powerful method for further characterisation, of amastigote-hosting DCs in the skin and the skin-draining lymph nodes.
Collapse
Affiliation(s)
- Hervé Lecoeur
- Institut Pasteur, Unité d'Immunophysiologie et Parasitisme Intracellulaire, Département de Parasitologie et Mycologie, Paris Cedex 15, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Construction of an unmarked recombinant BCG expressing a pertussis antigen by auxotrophic complementation: protection against Bordetella pertussis challenge in neonates. Vaccine 2009; 27:7346-51. [PMID: 19782111 DOI: 10.1016/j.vaccine.2009.09.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 09/02/2009] [Accepted: 09/13/2009] [Indexed: 10/20/2022]
Abstract
Mycobacterium bovis BCG has long been investigated as a candidate for heterologous antigen presentation. We have previously described an rBCG-Pertussis that confers protection against challenge with Bordetella pertussis in neonate and adult mice. In order to obtain stable expression in vivo, we constructed an unmarked BCG lysine auxotrophic and a complementation vector containing the lysine and the genetically detoxified S1 pertussis toxin genes, both under control of the same promoter. Complemented BCG-Delta lysine growth and expression of the pertussis antigen were stable, without the use of an antibiotic marker. Our results show that the complemented rBCG-Delta lysA-S1PT-lysA(+)(kan(-)), which is now suitable to be evaluated in clinical trials, maintains similar characteristics of the original rBCG-pNL71S1PT strain, such as the antigen expression level, cellular immune response and protection against the same model challenge in neonatal-immunized mice.
Collapse
|
36
|
Recombinant Mycobacterium bovis BCG. Vaccine 2009; 27:6495-503. [PMID: 19720367 DOI: 10.1016/j.vaccine.2009.08.044] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 08/11/2009] [Accepted: 08/12/2009] [Indexed: 12/14/2022]
Abstract
The Bacillus Calmette-Guerin (BCG) is an attenuated strain of Mycobacterium bovis that has been broadly used as a vaccine against human tuberculosis. This live bacterial vaccine is able to establish a persistent infection and induces both cellular and humoral immune responses. The development of mycobacterial genetic systems to express foreign antigens and the adjuvanticity of BCG are the basis of the potential use of this attenuated mycobacterium as a recombinant vaccine. Over the years, a range of strategies has been developed to allow controlled and stable expression of viral, bacterial and parasite antigens in BCG. Herein, we review the strategies developed to express heterologous antigens in BCG and the immune response elicited by recombinant BCG constructs. In addition, the use of recombinant BCG as an immunomodulator and future perspectives of BCG as a recombinant vaccine vector are discussed.
Collapse
|
37
|
Griffin S, Williamson AL, Chapman R. Optimisation of a mycobacterial replicon increases foreign antigen expression in mycobacteria. Tuberculosis (Edinb) 2009; 89:225-32. [DOI: 10.1016/j.tube.2009.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 03/31/2009] [Accepted: 03/31/2009] [Indexed: 10/20/2022]
|
38
|
Gousset K, Schiff E, Langevin C, Marijanovic Z, Caputo A, Browman DT, Chenouard N, de Chaumont F, Martino A, Enninga J, Olivo-Marin JC, Männel D, Zurzolo C. Prions hijack tunnelling nanotubes for intercellular spread. Nat Cell Biol 2009; 11:328-36. [PMID: 19198598 DOI: 10.1038/ncb1841] [Citation(s) in RCA: 477] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Accepted: 01/26/2009] [Indexed: 01/09/2023]
Abstract
In variant Creutzfeldt-Jakob disease, prions (PrP(Sc)) enter the body with contaminated foodstuffs and can spread from the intestinal entry site to the central nervous system (CNS) by intercellular transfer from the lymphoid system to the peripheral nervous system (PNS). Although several means and different cell types have been proposed to have a role, the mechanism of cell-to-cell spreading remains elusive. Tunnelling nanotubes (TNTs) have been identified between cells, both in vitro and in vivo, and may represent a conserved means of cell-to-cell communication. Here we show that TNTs allow transfer of exogenous and endogenous PrP(Sc) between infected and naive neuronal CAD cells. Significantly, transfer of endogenous PrP(Sc) aggregates was detected exclusively when cells chronically infected with the 139A mouse prion strain were connected to mouse CAD cells by means of TNTs, identifying TNTs as an efficient route for PrP(Sc) spreading in neuronal cells. In addition, we detected the transfer of labelled PrP(Sc) from bone marrow-derived dendritic cells to primary neurons connected through TNTs. Because dendritic cells can interact with peripheral neurons in lymphoid organs, TNT-mediated intercellular transfer would allow neurons to transport prions retrogradely to the CNS. We therefore propose that TNTs are involved in the spreading of PrP(Sc) within neurons in the CNS and from the peripheral site of entry to the PNS by neuroimmune interactions with dendritic cells.
Collapse
Affiliation(s)
- Karine Gousset
- Unité de Trafic Membranaire et Pathogénèse, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kocíncová D, Winter N, Euphrasie D, Daffé M, Reyrat JM, Etienne G. The cell surface-exposed glycopeptidolipids confer a selective advantage to the smooth variants of Mycobacterium smegmatis in vitro. FEMS Microbiol Lett 2008; 290:39-44. [PMID: 19025562 DOI: 10.1111/j.1574-6968.2008.01396.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The cell surface of mycobacteria is quite rich in lipids. Glycopeptidolipids, surface-exposed lipids that typify some mycobacterial species, have been associated with a phenotypic switch between rough and smooth colony morphotypes. This conversion in Mycobacterium smegmatis is correlated with the absence/presence of glycopeptidolipids on the cell surface and is due to insertion sequence mobility. Here, we show that the occurrence of a high amount of glycopeptidolipids in the smooth variant leads to lower invasion abilities and lower internalization by macrophages. We further show that the high production of glycopeptidolipids on the cell surface can confer a selective advantage to the smooth variant when grown in vitro. This higher fitness under the laboratory condition might explain the selection of smooth variants in several independent laboratories. The implications of these findings are discussed.
Collapse
Affiliation(s)
- Dana Kocíncová
- Faculté de Médecine, Université Paris Descartes, Paris, France
| | | | | | | | | | | |
Collapse
|
40
|
Badell E, Nicolle F, Clark S, Majlessi L, Boudou F, Martino A, Castello-Branco L, Leclerc C, Lewis DJM, Marsh PD, Gicquel B, Winter N. Protection against tuberculosis induced by oral prime with Mycobacterium bovis BCG and intranasal subunit boost based on the vaccine candidate Ag85B-ESAT-6 does not correlate with circulating IFN-gamma producing T-cells. Vaccine 2008; 27:28-37. [PMID: 18977269 DOI: 10.1016/j.vaccine.2008.10.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 09/15/2008] [Accepted: 10/14/2008] [Indexed: 12/17/2022]
Abstract
The potent IFN-gamma inducing fusion antigen Ag85B-ESAT-6 (85B6) is a lead subunit candidate to improve current vaccination against Mycobacterium tuberculosis (Mtb). The recombinant M. bovis BCG strain Myc3504 was constructed to secrete 85B6. It was based on commercial BCG strain Moreau Rio de Janeiro (BCG(MoWT)) which remains available for human oral administration. Myc 3504 induced higher levels of 85B6-specific IFN-gamma circulating T-cells as compared to BCG(MoWT). A novel needle-free mucosal immunization regimen combining oral prime with Myc3504 or BCG(MoWT) with intranasal boost with LTK-63-adjuvanted 85B6 was compared to subcutaneous prime-boost immunization. Strikingly whereas parenteral immunization induced sustained levels of 85B6-specific IFN-gamma secretion by circulating T-cells, mucosal regimens induced barely detectable IFN-gamma. Despite this, mice and guinea pigs immunized with the mucosal regimens were as efficiently protected against aerosol Mtb challenge as parenterally immunized animals. After Mtb challenge, anti-ESAT-6 IFN-gamma responses sharply increased in non-vaccinated mice as a hallmark of infection. Parenterally immunized mice that controlled Mtb infection, displayed anti-ESAT-6 IFN-gamma responses as high as non-immunized infected mice, compromising the possible use of ESAT-6 as a diagnostic tool. Interestingly, in mucosally immunized mice that were equally protected, post-challenge ESAT-6-specific IFN-gamma T-cell response remained low.
Collapse
Affiliation(s)
- Edgar Badell
- Institut Pasteur, Unité de Génétique Mycobactérienne, 25-28 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nod2 mediates susceptibility to Yersinia pseudotuberculosis in mice. PLoS One 2008; 3:e2769. [PMID: 18648508 PMCID: PMC2447872 DOI: 10.1371/journal.pone.0002769] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Accepted: 06/16/2008] [Indexed: 01/07/2023] Open
Abstract
Nucleotide oligomerisation domain 2 (NOD2) is a component of the innate immunity known to be involved in the homeostasis of Peyer patches (PPs) in mice. However, little is known about its role during gut infection in vivo. Yersinia pseudotuberculosis is an enteropathogen causing gastroenteritis, adenolymphitis and septicaemia which is able to invade its host through PPs. We investigated the role of Nod2 during Y. pseudotuberculosis infection. Death was delayed in Nod2 deleted and Crohn's disease associated Nod2 mutated mice orogastrically inoculated with Y. pseudotuberculosis. In PPs, the local immune response was characterized by a higher KC level and a more intense infiltration by neutrophils and macrophages. The apoptotic and bacterial cell counts were decreased. Finally, Nod2 deleted mice had a lower systemic bacterial dissemination and less damage of the haematopoeitic organs. This resistance phenotype was lost in case of intraperitoneal infection. We concluded that Nod2 contributes to the susceptibility to Y. pseudotuberculosis in mice.
Collapse
|
42
|
Morel C, Badell E, Abadie V, Robledo M, Setterblad N, Gluckman JC, Gicquel B, Boudaly S, Winter N. Mycobacterium bovis BCG-infected neutrophils and dendritic cells cooperate to induce specific T cell responses in humans and mice. Eur J Immunol 2008; 38:437-47. [PMID: 18203135 DOI: 10.1002/eji.200737905] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neutrophils are increasingly thought to modulate dendritic cell (DC) functions. We investigated the role of the neutrophil-DC partnership in the response to Mycobacterium bovis BCG-the vaccine used against tuberculosis. We compared neutrophil-DC crosstalk in humans and mice, searching for functional differences. In both species, neutrophils captured fluorescent BCG-enhanced green fluorescent protein (EGFP) and were more phagocytic than DC. Non-apoptotic BCG-infected neutrophils clustered with immature DC, establishing intimate contacts with dendrites, at which fluorescent intact bacilli were observed. Physical interactions between neutrophils and DC were required for DC activation. Human BCG-infected DC produced interleukin (IL)-10, an inhibitory cytokine, whereas DC exposed to BCG-infected neutrophils produced low to undetectable amounts of the cytokine. Mouse BCG-infected neutrophils induced sustained IL-2 production by DC. Human DC exposed to BCG-infected neutrophils stimulated recall T cell reactivity from vaccinated donors. Mouse DC infected with recombinant ovalbumin (OVA)-producing BCG (rBCG(ova)) elicited proliferation of TCR-OVA-transgenic CD4 and CD8 T cells. Moreover, exposing DC to rBCG(ova)-infected neutrophils enhanced OVA presentation. Thus, in mice and humans, neutrophils help DC to cross-present BCG antigens to T cells. Our results suggest that this "ménage à trois" involving neutrophils, DC and T cells plays a major role in the immune response to BCG.
Collapse
Affiliation(s)
- Céline Morel
- Unité Mixte de Recherche 7151 CNRS, Université Paris 7, Laboratoire d'immunologie cellulaire et immunopathologie de l'Ecole Pratique des Hautes Etudes, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Scandurra G, Britton W, Triccas J. Inactivation of the Mycobacterium tuberculosis fadB4 gene results in increased virulence in host cell and mice. Microbes Infect 2008; 10:38-44. [DOI: 10.1016/j.micinf.2007.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 10/02/2007] [Accepted: 10/03/2007] [Indexed: 10/22/2022]
|
44
|
Borsuk S, Mendum TA, Fagundes MQ, Michelon M, Cunha CW, McFadden J, Dellagostin OA. Auxotrophic complementation as a selectable marker for stable expression of foreign antigens in Mycobacterium bovis BCG. Tuberculosis (Edinb) 2007; 87:474-80. [PMID: 17888740 DOI: 10.1016/j.tube.2007.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 06/20/2007] [Accepted: 07/19/2007] [Indexed: 10/22/2022]
Abstract
Mycobacterium bovis BCG has the potential to be an effective live vector for multivalent vaccines. However, most mycobacterial cloning vectors rely on antibiotic resistance genes as selectable markers, which would be undesirable in any practical vaccine. Here we report the use of auxotrophic complementation as a selectable marker that would be suitable for use in a recombinant vaccine. A BCG auxotrophic for the amino acid leucine was constructed by knocking out the leuD gene by unmarked homologous recombination. Expression of leuD on a plasmid not only allowed complementation, but also acted as a selectable marker. Removal of the kanamycin resistance gene, which remained necessary for plasmid manipulations in Escherichia coli, was accomplished by two different methods: restriction enzyme digestion followed by re-ligation before BCG transformation, or by Cre-loxP in vitro recombination mediated by the bacteriophage P1 Cre Recombinase. Stability of the plasmid was evaluated during in vitro and in vivo growth of the recombinant BCG in comparison to selection by antibiotic resistance. The new system was highly stable even during in vivo growth, as the selective pressure is maintained, whereas the conventional vector was unstable in the absence of selective pressure. This new system will now allow the construction of potential recombinante vaccine strains using stable multicopy plasmid vectors without the inclusion of antibiotic resistance markers.
Collapse
Affiliation(s)
- Sibele Borsuk
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP-354, 96010-900 Pelotas, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
45
|
Dennehy M, Bourn W, Steele D, Williamson AL. Evaluation of recombinant BCG expressing rotavirus VP6 as an anti-rotavirus vaccine. Vaccine 2007; 25:3646-57. [PMID: 17339069 DOI: 10.1016/j.vaccine.2007.01.087] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2006] [Revised: 12/19/2006] [Accepted: 01/11/2007] [Indexed: 11/16/2022]
Abstract
Recombinant BCG expressing rotavirus VP6 was explored as an anti-rotavirus vaccine in a mouse model. Three promoters and five ribosome-binding sites were used in episomal and integrative E. coli-mycobacterium shuttle vectors to express VP6 in BCG. The VP6 gene was configured for accumulation within the BCG cytoplasm, secretion from the BCG cell or targeting to the BCG cell membrane. Vectors were assessed in terms of stability, levels of antigen production, immunogenicity and protection in mice. Gross instability occurred in episomal vectors utilizing the hsp60 promoter. However, three integrative vectors using the same expression system and two episomal vectors using inducible promoters were successfully recovered from BCG. Growth rates of the former were not detectably reduced. Growth rates of the latter were considerably reduced, implying the existence of a significant metabolic load. In the absence of selection, loss rate of these plasmids was high. VP6 production levels (0.04-1.78% of total cytoplasmic protein) were on the lower end of the range reported for other rBCG. One episomal and one integrated vaccine reduced viral shedding in intraperitoneally vaccinated mice challenged with rotavirus. Compared to controls, infection-associated faecal shedding of virus was reduced by 66% and 62%, respectively. These protective vectors differ in promoter, ribosome-binding site and antigen production level, but both link the VP6 protein to the 19kDa lipoprotein signal sequence, suggesting that transport of VP6 to the BCG membrane is important for induction of a protective immune response. Protection occurred in the absence of detectable anti-rotavirus antibody in serum or faeces, implicating cellular immunity in protection.
Collapse
Affiliation(s)
- Maureen Dennehy
- Division of Medical Virology, Department of Clinical Laboratory Sciences, Faculty of Health Sciences, University of Cape Town, and National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa.
| | | | | | | |
Collapse
|
46
|
Santangelo MP, McIntosh D, Bigi F, Armôa GRG, Campos ASD, Ruybal P, Dellagostin OA, McFadden J, Mendum T, Gicquel B, Winter N, Farber M, Cataldi A. Mycobacterium bovis BCG as a delivery system for the RAP-1 antigen from Babesia bovis. Vaccine 2007; 25:1104-13. [PMID: 17049681 DOI: 10.1016/j.vaccine.2006.09.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Revised: 09/15/2006] [Accepted: 09/15/2006] [Indexed: 11/20/2022]
Abstract
Babesia bovis is the causative agent of babesiosis, a tick-borne disease that is a major cause of loss to livestock production in Latin America. Vaccination against Babesia species represents a major challenge against cattle morbidity and mortality in enzootic areas. The aim of this study was to evaluate the capacity of Bacille Calmette-Guerin (BCG) to deliver the rhoptry associated protein (RAP-1) antigen of B. bovis and to stimulate specific cellular and humoral immune responses in mice. Two of five mycobacterial expression vectors efficiently expressed the antigen. These constructs were subsequently studied in vivo following three immunization protocols. The construct with the greatest in vivo stability proved to be the one that induced the strongest immune responses. Our data support the hypothesis that specific T lymphocyte priming by rBCG can be employed as a component of a combined vaccine strategy to induce long-lasting humoral and cellular immune responsiveness towards B. bovis and encourage further work on the application of rBCG to the development of Babesia vaccines.
Collapse
Affiliation(s)
- M P Santangelo
- Institute of Biotechnology, CICVyA-INTA, Los Reseros y Las Cabañas, 1712 Castelar, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Modification of the mycobacteriophage Ms6 attP core allows the integration of multiple vectors into different tRNAala T-loops in slow- and fast-growing mycobacteria. BMC Mol Biol 2006; 7:47. [PMID: 17173678 PMCID: PMC1762012 DOI: 10.1186/1471-2199-7-47] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 12/15/2006] [Indexed: 11/17/2022] Open
Abstract
Background Mycobacteriophage Ms6 integrates into Mycobacterium smegmatis and M. bovis BCG chromosome at the 3' end of tRNAala genes. Homologous recombination occurs between the phage attP core and the attB site located in the T-loop. Integration-proficient vectors derived from Ms6 are useful genetic tools, but their insertion sites in the BCG chromosome remain poorly defined. The primary objective of this study was to identify Ms6 target genes in M. smegmatis and BCG. We then aimed to modify the attP site in Ms6-derived vectors, to switch integration to other tRNAala loci. This provided the basis for the development of recombinant M. bovis BCG strains expressing several reporter genes inserted into different tRNAala genes. Results The three tRNAala genes are highly conserved in M. smegmatis and BCG. However, in the T-loop of tRNAalaU and tRNAalaV containing the attB site, a single base difference was observed between the two species. We observed that the tRNAalaU gene was the only site into which Ms6-derived integration-proficient vectors integrated in M. smegmatis, whereas in BCG, the tRNAalaV gene was used as the target. No integration occurred in the BCG tRNAalaU T-loop, despite a difference of only one base from the 26-base Ms6 attP core. We mutated the attP core to give a perfect match with the other tRNAala T-loops from M. smegmatis and BCG. Modification of the seven-base T-loop decreased integration efficiency, identifying this site as a possible site of strand exchange. Finally, two Ms6 vectors were constructed to integrate two reporter genes into the tRNAalaU and tRNAalaV T-loops of the same BCG chromosome. Conclusion Small changes in the 7 bp T-loop attP site of Ms6 made it possible to use another attB site, albeit with a lower integration efficiency. These molecular studies on BCG tRNAala genes made it possible to create valuable tools for the site-directed insertion of several genes in the same BCG strain. These tools will be useful for the development of novel multivalent vaccines and genetic studies.
Collapse
|
48
|
Varaldo PB, Miyaji EN, Vilar MM, Campos AS, Dias WO, Armôa GRG, Tendler M, Leite LCC, McIntosh D. Mycobacterial codon optimization of the gene encoding the Sm14 antigen of Schistosoma mansoni in recombinant Mycobacterium bovis Bacille Calmette-Guérin enhances protein expression but not protection against cercarial challenge in mice. ACTA ACUST UNITED AC 2006; 48:132-9. [PMID: 16965361 DOI: 10.1111/j.1574-695x.2006.00133.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A mycobacterial codon-optimized gene encoding the Sm14 antigen of Schistosoma mansoni was generated using oligonucleotide assembly. This synthetic gene enhanced approximately fourfold the protein expression level in recombinant Mycobacterium bovis Bacille Calmette-Guérin (rBCG) when compared to that obtained using the native gene in the same expression vector. Immunization of mice with rBCG expressing Sm14 via the synthetic gene induced specific cellular Th1-predominant immune responses, as determined by interferon-gamma production of Sm14-stimulated splenocytes, which were comparable to those recorded in animals immunized with an rBCG strain expressing the native gene. Administration of a single dose of the rBCG-Sm14 construct carrying the synthetic gene conferred protection against cercarial challenge in outbred Swiss mice, at a level equivalent to those provided by either a single dose of rBCG expressing the native gene or three doses of Escherichia coli-derived recombinant Sm14. Our data demonstrated that despite improving the level of antigen expression, the codon optimization strategy did not result in enhanced immunity or protection against cercarial S. mansoni challenge.
Collapse
Affiliation(s)
- Paula B Varaldo
- Centro de Biotecnologia, Instituto Butantan, Sao Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Campoy S, Aranda J, Alvarez G, Barbé J, Llagostera M. Isolation and sequencing of a temperate transducing phage for Pasteurella multocida. Appl Environ Microbiol 2006; 72:3154-60. [PMID: 16672452 PMCID: PMC1472319 DOI: 10.1128/aem.72.5.3154-3160.2006] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A temperate bacteriophage (F108) has been isolated through mitomycin C induction of a Pasteurella multocida serogroup A strain. F108 has a typical morphology of the family Myoviridae, presenting a hexagonal head and a long contractile tail. F108 is able to infect all P. multocida serogroup A strains tested but not those belonging to other serotypes. Bacteriophage F108, the first P. multocida phage sequenced so far, presents a 30,505-bp double-stranded DNA genome with cohesive ends (CTTCCTCCCC cos site). The F108 genome shows the highest homology with those of Haemophilus influenzae HP1 and HP2 phages. Furthermore, an F108 prophage attachment site in the P. multocida chromosome has been established to be inside a gene encoding tRNA(Leu). By using several chromosomal markers that are spread along the P. multocida chromosome, it has been demonstrated that F108 is able to perform generalized transduction. This fact, together with the absence of pathogenic genes in the F108 genome, makes this bacteriophage a valuable tool for P. multocida genetic manipulation.
Collapse
Affiliation(s)
- Susana Campoy
- Centre de Recerca en Sanitat Animal (CReSA), Universitat Autònoma de Barcelona, Edifici C, Bellaterra, 08193 Barcelona, Spain
| | | | | | | | | |
Collapse
|
50
|
Michelon A, Conceição FR, Binsfeld PC, da Cunha CW, Moreira AN, Argondizzo AP, McIntosh D, Armôa GRG, Campos AS, Farber M, McFadden J, Dellagostin OA. Immunogenicity of Mycobacterium bovis BCG expressing Anaplasma marginale MSP1a antigen. Vaccine 2006; 24:6332-9. [PMID: 16781025 DOI: 10.1016/j.vaccine.2006.05.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 05/11/2006] [Accepted: 05/16/2006] [Indexed: 11/16/2022]
Abstract
Humoral and cellular immune responses of mice inoculated with recombinant Mycobacterium bovis BCG expressing the MSP1a antigen of Anaplasma marginale were evaluated. The msp1a gene was amplified by PCR and cloned into the mycobacterial expression vectors pUS2000 and pMIP12. Immunization of isogenic BALB/c mice with the rBCG/pUS2000-msp1a construct induced significant seroconversion to MSP1a (p<0.001), which was 26 times above pre-immunization levels at day 63 post-initial immunization and which remained stable for the duration of the experiment (6 months). In contrast, rBCG/pMIP12-msp1a induced seroconversion at a level of 6 times above pre-immunization values, which peaked at day 63. Western blot analysis showed that sera derived from mice vaccinated with either rBCG construct recognized both native and recombinant forms of A. marginale MSP1a. In contrast to the humoral response data, immunization with rBCG/pMIP12-msp1a was found to induce a markedly stronger cellular response than that recorded for BCG/pUS2000-msp1a. These observations clearly demonstrated the immunogenicity of recombinant BCG expressing the MSP1a antigen and suggested that the immune responses were influenced by the level of antigen expression. The results of this research warrant studies of recombinant M. bovis BCG expressing MSP1a in cattle to test for protective antibody production for control of bovine anaplasmosis.
Collapse
Affiliation(s)
- André Michelon
- Centro de Biotecnologia, Universidade Federal de Pelotas, CP 354, 96010-900 Pelotas, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|