1
|
Caudill MT, Stoyanof ST, Caswell CC. Quorum sensing LuxR proteins VjbR and BabR jointly regulate Brucella abortus survival during infection. J Bacteriol 2025; 207:e0052724. [PMID: 40013834 PMCID: PMC11925318 DOI: 10.1128/jb.00527-24] [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: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 02/28/2025] Open
Abstract
Brucella abortus maintains an N-acetyl homoserine lactone quorum sensing system that consists of two LuxR proteins, VjbR and BabR, as well as two signals, dodecanoyl (C12 AHL) and 3-oxododecanoyl (3-OXO-C12 AHL) homoserine lactone. This system regulates major virulence factors that influence the bacteria's survival during infection. We generated the first strain that lacks both LuxR proteins and found a synergistic interaction for survival in the chronic infection C57BL/6 mouse model. Transcriptomic analyses of the ∆vjbR∆babR double-deletion strain, as well as the cognate single-deletion strains, in a rich medium with vehicle control or supplemented with an AHL signal revealed large-scale genetic dysregulation in all conditions. Moreover, the double mutant maintained a limited response to AHL, even in the absence of the LuxR proteins. We additionally found that quorum sensing regulates the denitrification pathway but found no in vitro differences in the ability of quorum sensing deletion strains to clear nitric oxide stress or grow under anoxic denitrifying conditions. Finally, we confirmed that BabR autoregulates its own expression, and that VjbR mildly represses BabR expression. Altogether, these experiments help further characterize the Brucella quorum sensing systems and indicate that further attention should be given to the joint interactions between VjbR and BabR in controlling virulence.IMPORTANCEBrucella abortus is a zoonotic bacterial pathogen that uses its quorum sensing to survive within hosts. This study further characterizes that system and indicates important future lines of inquiry. We found that both quorum sensing proteins, VjbR and BabR, coordinate to maintain survival, as well as document that both quorum sensing systems appear physiologically active.
Collapse
Affiliation(s)
- Mitchell T Caudill
- Center for One Health Research, Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - S Tristan Stoyanof
- Center for One Health Research, Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
| | - Clayton C Caswell
- Center for One Health Research, Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
| |
Collapse
|
2
|
Harms JS, Lasarev M, Warner T, Costa Oliveira S, Smith JA. Persistent articular infection and host reactive response contribute to Brucella -induced spondyloarthritis in SKG mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638825. [PMID: 40027658 PMCID: PMC11870484 DOI: 10.1101/2025.02.18.638825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Brucellosis, one of the most prevalent zoonotic diseases worldwide, often results in osteoarticular complications including large joint and axial arthritis mimicking spondyloarthritis. To model this chronic manifestation, we infected autoimmunity-prone SKG mice containing a mutation in the T-cell adaptor ZAP-70 with Brucella species. B. melitensis infection resulted in a fully penetrant, readily scoreable disease involving large joint wrist and foot arthritis, peri-ocular inflammation, and less frequent scaly paw rash. Infection with B. abortus resulted in delayed arthritis onset, and B. neotomae revealed sex differences, with more severe disease and a dose response in females. Heat-killed Brucella did not induce arthritis, evincing a requirement for viable infection. Across species, splenic CFU correlated well with final clinical score at 12 weeks (ρ=0.79 and p<0.001). In vivo imaging using luminescent B. neotomae revealed rapid colonization of the paws by one-week post-infection, more than a month prior to arthritis onset. Paw luminescence levels decreased after 2 weeks and then remained relatively static, even as clinical scores increased. Thus, the degree of arthritis did not strictly correlate with degree of paw infection but suggested an additional reactive component. Further, in examining a Brucella Δ tcpB mutant lacking a Type IV secretion system-dependent mediator, mice displayed an intermediate phenotype without significant differences in splenic CFU. Together these data suggest Brucella induced spondyloarthritis reflects both persistent colonization as well as excess host reactivity. Moreover, the sensitivity of the SKG model to different species and mutants will provide new opportunities for dissecting correlates of Brucella virulence and host immunity. Importance Brucellosis, a bacterial infection acquired from herd animals, remains one of the most common zoonotic diseases worldwide. Chronic infection often results in spondyloarthritis-like complications. Investigation into pathogenesis has been limited by the lack of overt disease in standard lab mice. We addressed this issue using spondyloarthritis-susceptible SKG mice. Upon infection with B. melitensis , SKG mice develop robust, fully penetrant large joint arthritis. Arthritis development required viable bacteria. Moreover, studies of colonization, gene expression and anatomic distribution using bioluminescent bacteria revealed active persistent infection in the mouse paws. However, peak paw infection occurred much earlier than arthritis onset, suggesting an added immune reactive component. Disease onset, severity and manifestations varied upon infection with different Brucella species and mutants. Together these results suggest this new model will be very useful to the scientific community for determining correlates of bacterial virulence leading to clinical disease.
Collapse
|
3
|
Sancho-Sánchez E, García-Arteaga K, Granados-Chinchilla F, Artavia G, Alfaro-Alarcón A, Villalobos-Villalobos A, Bouza-Mora L, Suárez-Esquivel M, Chacón-Díaz C, Guzmán-Verri C, Moreno E, Barquero-Calvo E. Reactivation of hidden-latent Brucella infection after doxycycline and streptomycin treatment in mice. Antimicrob Agents Chemother 2025; 69:e0130224. [PMID: 39745377 PMCID: PMC11823614 DOI: 10.1128/aac.01302-24] [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: 08/28/2024] [Accepted: 11/08/2024] [Indexed: 02/14/2025] Open
Abstract
Brucellosis has therapeutic challenges due to 3%-15% relapses/therapeutic failures (R/TF) after antibiotic treatment. Therefore, determining the antibiotic concentration in tissues, the physiopathological parameters, and the R/TF after treatment is relevant. After exploring different antibiotic quantities, we found that a combined dose of 100 µg/g of doxycycline (for 45 days) and 7.5 µg/g of streptomycin (for 14 days), respectively, achieved therapeutic levels of more than fourfold minimum inhibitory concentrations (MICs) against Brucella abortus in the spleen, liver, bone marrow, and plasma of mice, causing minimal pathophysiological effects. After 30 days of infection, mice received antibiotics, and hematological, histopathological, biochemical, and immunological analyses were performed. After antibiotic therapy, the pathological, hematological, immunological, and physiological profiles paralleled those described in human brucellosis. Treatment lowered antibody titers, reduced proinflammatory cytokines, and reduced inflammation in the target organs for a protracted period. No bacteria were detected in tissues 8 weeks after treatment, suggesting complete recovery. However, despite high doxycycline and streptomycin concentrations in tissues, relapses appeared in 100% of the animals after 182 days post-infection, estimated by the bacterial counts and PCR from organs. This proportion contrasts with the 15% R/TF observed in humans after antibiotic treatments. None of the B. abortus isolated from relapses showed augmented MICs or mutations coding for antibiotic resistance in chromosomal-relevant regions. We discuss whether our findings constitute a general phenomenon or differences in the exhaustive screening method for bacteria detection related to the murine model. Along these lines, we envision likely mechanisms of bacterial persistence in tissues after antibiotic treatment.
Collapse
Affiliation(s)
- Eugenia Sancho-Sánchez
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Kimberly García-Arteaga
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Fabio Granados-Chinchilla
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
- Escuela de Química, Universidad de Costa Rica, Sede Rodrigo Facio, San Pedro Montes de Oca, San José, Costa Rica
| | - Graciela Artavia
- Centro Nacional de Ciencia y Tecnología de Alimentos, Universidad de Costa Rica, San José, Costa Rica
| | - Alejandro Alfaro-Alarcón
- Departamento de Patología, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
- Berlin Institute of Health, Institute of Virology, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Laura Bouza-Mora
- Departamento de Análisis Clínicos, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Marcela Suárez-Esquivel
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Carlos Chacón-Díaz
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Caterina Guzmán-Verri
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Edgardo Moreno
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Elías Barquero-Calvo
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| |
Collapse
|
4
|
Ponzilacqua-Silva B, Dadelahi AS, Moley CR, Abushahba MFN, Skyberg JA. Metabolomic analysis of murine tissues infected with Brucella melitensis. PLoS One 2025; 20:e0314672. [PMID: 39869554 PMCID: PMC11771894 DOI: 10.1371/journal.pone.0314672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/07/2025] [Indexed: 01/29/2025] Open
Abstract
Brucella is a gram negative, facultative intracellular bacterial pathogen that constitutes a substantial threat to human and animal health. Brucella can replicate in a variety of tissues and can induce immune responses that alter host metabolite availability. Here, mice were infected with B. melitensis and murine spleens, livers, and female reproductive tracts were analyzed by GC-MS to determine tissue-specific metabolic changes at one-, two- and four- weeks post infection. The most remarkable changes were observed at two-weeks post-infection when relative to uninfected tissues, 42 of 329 detected metabolites in reproductive tracts were significantly altered by Brucella infection, while in spleens and livers, 68/205 and 139/330 metabolites were significantly changed, respectively. Several of the altered metabolites in host tissues were linked to the GABA shunt and glutaminolysis. Treatment of macrophages with GABA did not alter control of B. melitensis infection, and deletion of the putative GABA transporter BMEI0265 did not alter B. melitensis virulence. While glutaminolysis inhibition did not affect control of B. melitensis in macrophages, glutaminolysis was required for macrophage IL-1β production in response to B. melitensis. In summary, these results indicate that Brucella infection alters host tissue metabolism and that these changes could have effects on inflammation and the outcome of infection.
Collapse
Affiliation(s)
- Bárbara Ponzilacqua-Silva
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri
| | - Alexis S. Dadelahi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri
| | - Charles R. Moley
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri
| | - Mostafa F. N. Abushahba
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri
- Department of Zoonoses, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Jerod A. Skyberg
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| |
Collapse
|
5
|
Pellegrini JM, González-Espinoza G, Shayan RR, Hysenaj L, Rouma T, Arce-Gorvel V, Lelouard H, Popoff D, Zhao Y, Hanniffy S, Castillo-Zeledón A, Loperena-Barber M, Celis-Gutierrez J, Mionnet C, Bosilkovski M, Solera J, Muraille E, Barquero-Calvo E, Moreno E, Conde-Álvarez R, Moriyón I, Gorvel JP, Mémet S. Brucella abortus impairs T lymphocyte responsiveness by mobilizing IL-1RA-secreting omental neutrophils. Nat Commun 2025; 16:862. [PMID: 39833171 PMCID: PMC11747348 DOI: 10.1038/s41467-024-55799-2] [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: 08/01/2023] [Accepted: 12/30/2024] [Indexed: 01/22/2025] Open
Abstract
Immune evasion strategies of Brucella, the etiologic agent of brucellosis, a global zoonosis, remain partially understood. The omentum, a tertiary lymphoid organ part of visceral adipose tissue, has never been explored as a Brucella reservoir. We report that B. abortus infects and replicates within murine omental macrophages. Throughout the chronic phase of infection, the omentum accumulates macrophages, monocytes and neutrophils. The maintenance of PD-L1+Sca-1+ macrophages, monocytes and neutrophils in the omentum depends on the wadC-encoded determinant of Brucella LPS. We demonstrate that PD-L1+Sca-1+ murine omental neutrophils produce high levels of IL-1RA leading to T cell hyporesponsiveness. These findings corroborate brucellosis patient analysis of whole blood displaying upregulation of PDL1 and Ly6E genes, and of serum exhibiting high levels of IL-1RA. Overall, the omentum, a reservoir for B. abortus, promotes bacterial persistence and causes CD4+ and CD8+ T cell immunosuppression by IL-1RA secreted by PD-L1+Sca-1+ neutrophils.
Collapse
Affiliation(s)
| | | | | | - Lisiena Hysenaj
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Thomas Rouma
- Unité de Recherche en Biologie des Microorganismes (URBM)-Laboratoire d'Immunologie et de Microbiologie, NARILIS, University of Namur, Namur, Belgium
- Laboratoire de de Parasitologie, and ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles, Gosselies, Belgium
| | | | - Hugues Lelouard
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Dimitri Popoff
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Yun Zhao
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Sean Hanniffy
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Amanda Castillo-Zeledón
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Maite Loperena-Barber
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | | | - Cyrille Mionnet
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Mile Bosilkovski
- University Clinic for Infectious Diseases and Febrile Conditions, Skopje, Republic of North Macedonia
| | - Javier Solera
- Hospital General Universitario, Facultad de Medicina, Universidad Castilla la Mancha Albacete, Albacete, Spain
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes (URBM)-Laboratoire d'Immunologie et de Microbiologie, NARILIS, University of Namur, Namur, Belgium
- Laboratoire de de Parasitologie, and ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles, Gosselies, Belgium
| | - Elías Barquero-Calvo
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Edgardo Moreno
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Raquel Conde-Álvarez
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | - Ignacio Moriyón
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | | | - Sylvie Mémet
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
6
|
Uslu A, Sayın Z, Balevi A, Gulcu Y, Ergen F, Akıner I, Denizli O, Erganis O. Comparison of Long-Term Antibody Titers in Calves Treated with Different Conjunctival and Subcutaneous Brucella abortus S19 Vaccines. Animals (Basel) 2025; 15:212. [PMID: 39858212 PMCID: PMC11758667 DOI: 10.3390/ani15020212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Brucellosis is still the most common zoonosis worldwide despite advanced technology and animal husbandry. Since there is still no effective Brucella vaccine for humans, it is crucial to control the disease in ruminants through eradication and vaccination. Although some countries around the world have achieved this circumstance, every country aims to become free of Brucellosis through vaccination, animal movements, and various eradication measures. For this purpose, the Brucella abortus S19 strain has been used safely for about 100 years. However, due to the O-polysaccharide (OPS) antigen in its structure, the antibody response created by the vaccine causes confusion in serological tests. For this purpose, researchers have provided both mucosal immunity and short-term antibody response by using the B. abortus S19 vaccine in conjunctival form instead of subcutaneous form. This study aimed to determine how long the post-vaccination titer levels persisted in animals vaccinated with vaccines from 3 different companies and different routes. In this study, a total of 115 calves aged 3 to 4.5 months were created in five groups, with 23 animals in each group: group 1 (vaccine brand A), group 2 (vaccine brand B), and group 3 (vaccine brand C) received the two-dose conjunctival vaccine, group 4 received the single-dose subcutaneous vaccine (vaccine brand C), and group 5 received the subcutaneous vaccine (vaccine brand C) plus the booster dose conjunctival vaccine (vaccine brand B). Brucellosis antibody titers were monitored each 21 days until the cattle were 26-28 months old. The collected sera were analyzed using the Rose Bengal Plate Test (RBPT), Serum Agglutination Test (SAT), and Complement Fixation Test (CFT), which are the preferred serological methods for Brucellosis eradication plans worldwide. In the conjunctival vaccination groups, only 3 (13%) of the animals in group 1 developed antibody titers one month after vaccination, and there was no antibody response detected against Brucellosis in group 2 and group 3. In animals that were stimulated conjunctivally, the threshold value of 30 International CFT Units (ICFTUs) (for distinguishing between infective titers and vaccination titers) was observed in one animal each in group 1 and group 2 and 0 animal in group 3. It was found that antibody titers turn to Brucellosis negative in all conjunctival vaccine groups at 7 months after vaccination. In groups 4 and 5, the first-month serological screening detected over 30 ICFTUs in 17 (89.47%) animals and 16 (69.5%) animals, respectively. In group 4, CFT titers were found to fall below 30 on the 17th month and 9.3 on the 22nd month. On the 14th month, the CFT titers of group 5 were found to be below 30, and all animals in this group turned negative after the 19th month. It was found that the single dose B. abortus S19 subcutaneous vaccination in calves caused persistent antibodies in 5% of the population. It is believed that persistent and high antibody titers created by subcutaneous vaccines will cause false positivity and create confusion in Brucellosis eradication programs. Therefore, although there is no clear distinction between vaccinated and infected animals, it has been observed that conjunctival Brucellosis vaccines create more stable antibody titers and decrease rapidly compared to subcutaneous vaccines. Based on the results of this study and the advantages of conjunctival vaccines, more effective eradication programs and antibody monitoring can be carried out in vaccinated herds where Brucellosis outbreaks are observed.
Collapse
Affiliation(s)
- Ali Uslu
- Department of Microbiology, Faculty of Veterinary Medicine, Selcuk University, 42130 Konya, Turkey; (Z.S.); (A.B.); (O.D.); (O.E.)
| | - Zafer Sayın
- Department of Microbiology, Faculty of Veterinary Medicine, Selcuk University, 42130 Konya, Turkey; (Z.S.); (A.B.); (O.D.); (O.E.)
| | - Aslı Balevi
- Department of Microbiology, Faculty of Veterinary Medicine, Selcuk University, 42130 Konya, Turkey; (Z.S.); (A.B.); (O.D.); (O.E.)
| | - Yasin Gulcu
- Konya Veterinary Control and Research Institute, 42080 Konya, Turkey; (Y.G.); (I.A.)
| | - Fırat Ergen
- Gözlü Agricultural Enterprise, 42430 Konya, Turkey;
| | - Islam Akıner
- Konya Veterinary Control and Research Institute, 42080 Konya, Turkey; (Y.G.); (I.A.)
| | - Oguzhan Denizli
- Department of Microbiology, Faculty of Veterinary Medicine, Selcuk University, 42130 Konya, Turkey; (Z.S.); (A.B.); (O.D.); (O.E.)
| | - Osman Erganis
- Department of Microbiology, Faculty of Veterinary Medicine, Selcuk University, 42130 Konya, Turkey; (Z.S.); (A.B.); (O.D.); (O.E.)
| |
Collapse
|
7
|
Ciliberti MG, Santillo A, Caroprese M, Albenzio M. Buffalo Immune Competence Under Infectious and Non-Infectious Stressors. Animals (Basel) 2025; 15:163. [PMID: 39858163 PMCID: PMC11759140 DOI: 10.3390/ani15020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Immune competence is a critical aspect of protecting animals from the negative consequences of disease. The activation of the immune response from inflammation is part of adaptive homeostasis that serves to eliminate danger, induce tissue repair, and restore tissue homeostasis. Therefore, the main goal for the organism is to control both the induction and suppression of inflammation and resist the onset of disease. In this condition, modulators of inflammatory responses are produced, including small proteins called cytokines, which exert a pro- or anti-inflammatory action in a context-dependent manner. Indeed, the cytokine profile could be considered a useful biomarker to determine the pathophysiology of certain diseases, such as mastitis, endometritis, change-induced heat stress, and zoonoses. Recently, buffalo breeding has attracted the interest of the research communities due to their high resilience; however, little is known about the immune mechanism activated under specific stressors. This review describes the complex immune competence of the buffalo in the presence of the most common infectious and non-infectious stressors. In addition, a brief description of methods for early diagnosis of disease using cytokine quantification will be introduced.
Collapse
Affiliation(s)
| | - Antonella Santillo
- Department of Agriculture, Food, Natural Resources, and Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy; (M.G.C.); (M.C.); (M.A.)
| | | | | |
Collapse
|
8
|
Yang J, Liu X, Yang H, Wang H, Xie B, Gao W, Xu M, Xu X, Liu B, Chen Z. Combination application of Pickering emulsion and BLS protein nanoparticles enhances vaccine efficacy. Int J Biol Macromol 2024; 283:137635. [PMID: 39547601 DOI: 10.1016/j.ijbiomac.2024.137635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
The deficiency of recombinant protein immune response could be compensated for by using nanoparticle platforms or adding immune enhancers, however existing vaccines or adjuvants struggle to elicit durable cellular immune responses. In this work, a protein nanoscaffold, lumazine synthase isolated from Brucella (BLS) was optimally designed that could facilitate cellular uptake of displayed antigens and the maturation of bone marrow-derived dendritic cells (BMDCs), and enhancing humoral immune responses. To enhance cellular immune response, chitosan hydrochloride-stabilized Pickering emulsion (CHSPE) was evaluated as an adjuvant for the BLS nanoscaffold-based vaccine. CHSPE could enhance the recruitment and activation of DCs at the injection site and the uptake of antigen and activation of DCs in the draining lymph nodes (dLNs), compared with commercial adjuvant ISA 206. In addition, CHSPE induced antigen-specific antibody levels comparable to those of ISA 206 and increased the ratio of central memory T cells (TCM) and effector memory T cells (TEM), and promoted the secretion of Th1-type cytokines. Moreover, CHSPE-formulated vaccine provided a similar level of protection to the live attenuated vaccine and was superior to that of ISA 206. Taken together, the combination of the BLS nanoscaffold and CHSPE provides a feasible strategy for recombinant protein subunit vaccine development.
Collapse
Affiliation(s)
- Jianghua Yang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China
| | - Xiang Liu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China
| | - Haoyan Yang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China
| | - Hao Wang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China
| | - Bin Xie
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China
| | - Weiyu Gao
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China
| | - Man Xu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China
| | - Xinling Xu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China
| | - Baoshan Liu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China.
| | - Zeliang Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University. Liaoning Province, Shenyang 110866, PR China; NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Key Laboratory of Tropical Diseases Control, School of public health, Sun Yat-sen University, Guangzhou 510275, PR China; Collaborative Innovation Center for Prevention and Control of Zoonoses, Jinzhou Medical University, Jinzhou 121001, PR China.
| |
Collapse
|
9
|
Elizalde-Bielsa A, Lázaro-Antón L, de Miguel MJ, Muñoz PM, Conde-Álvarez R, Zúñiga-Ripa A. Disruption of Erythritol Catabolism via the Deletion of Fructose-Bisphosphate Aldolase (Fba) and Transaldolase (Tal) as a Strategy to Improve the Brucella Rev1 Vaccine. Int J Mol Sci 2024; 25:11230. [PMID: 39457012 PMCID: PMC11508834 DOI: 10.3390/ijms252011230] [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: 08/30/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Brucellosis is a bacterial zoonosis caused by the genus Brucella, which mainly affects domestic animals. In these natural hosts, brucellae display a tropism towards the reproductive organs, such as the placenta, replicating in high numbers and leading to placentitis and abortion, an ability also exerted by the B. melitensis live-attenuated Rev1 strain, the only vaccine available for ovine brucellosis. It is broadly accepted that this tropism is mediated, at least in part, by the presence of certain preferred nutrients in the placenta, particularly erythritol, a polyol that is ultimately incorporated into the Brucella central carbon metabolism via two reactions dependent on transaldolase (Tal) or fructose-bisphosphate aldolase (Fba). In the light of these remarks, we propose that blocking the incorporation of erythritol into the central carbon metabolism of Rev1 by deleting the genes encoding Tal and Fba may impair the ability of the vaccine to proliferate massively in the placenta. Therefore, a Rev1ΔfbaΔtal double mutant was generated and confirmed to be unable to use erythritol. This mutant exhibited a reduced intracellular fitness both in BeWo trophoblasts and THP-1 macrophages. In the murine model, Rev1ΔfbaΔtal provided comparable protection to the Rev1 reference vaccine while inducing fewer adverse reproductive events in pregnant animals. Altogether, these results postulate the Rev1ΔfbaΔtal mutant as a reproductively safer Rev1-derived vaccine candidate to be studied in the natural host.
Collapse
Affiliation(s)
- Aitor Elizalde-Bielsa
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (A.E.-B.); (L.L.-A.)
| | - Leticia Lázaro-Antón
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (A.E.-B.); (L.L.-A.)
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95616, USA
| | - María Jesús de Miguel
- Department of Animal Science, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), 50059 Zaragoza, Spain; (M.J.d.M.); (P.M.M.)
- Instituto Agroalimentario de Aragón—IA2, CITA-Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Pilar M. Muñoz
- Department of Animal Science, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), 50059 Zaragoza, Spain; (M.J.d.M.); (P.M.M.)
- Instituto Agroalimentario de Aragón—IA2, CITA-Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Raquel Conde-Álvarez
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (A.E.-B.); (L.L.-A.)
| | - Amaia Zúñiga-Ripa
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (A.E.-B.); (L.L.-A.)
| |
Collapse
|
10
|
Poveda-Urkixo I, Mena-Bueno S, Ramírez GA, Zabalza-Baranguá A, Tsolis RM, Grilló MJ. Brucella melitensis Rev1Δwzm: Placental pathogenesis studies and safety in pregnant ewes. Vaccine 2024; 42:3710-3720. [PMID: 38755066 DOI: 10.1016/j.vaccine.2024.04.085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/20/2024] [Accepted: 04/27/2024] [Indexed: 05/18/2024]
Abstract
One of the main causes of human brucellosis is Brucella melitensis infecting small ruminants. To date, Rev1 is the only vaccine successfully used to control ovine and caprine brucellosis. However, it is pathogenic for pregnant animals, resulting in abortions and vaginal and milk shedding, as well as being infectious for humans. Therefore, there is an urgent need to develop an effective vaccine that is safer than Rev1. In efforts to further attenuate Rev1, we recently used wzm inactivation to generate a rough mutant (Rev1Δwzm) that retains a complete antigenic O-polysaccharide in the bacterial cytoplasm. The aim of the present study was to evaluate the placental pathogenicity of Rev1Δwzm in trophoblastic cells, throughout pregnancy in mice, and in ewes inoculated in different trimesters of pregnancy. This mutant was evaluated in comparison with the homologous 16MΔwzm derived from a virulent strain of B. melitensis and the naturally rough sheep pathogen B. ovis. Our results show that both wzm mutants triggered reduced cytotoxic, pro-apoptotic, and pro-inflammatory signaling in Bewo trophoblasts, as well as reduced relative expression of apoptosis genes. In mice, both wzm mutants produced infection but were rapidly cleared from the placenta, in which only Rev1Δwzm induced a low relative expression of pro-apoptotic and pro-inflammatory genes. In the 66 inoculated ewes, Rev1Δwzm was safe and immunogenic, displaying a transient serological interference in standard RBT but not CFT S-LPS tests; this serological response was minimized by conjunctival administration. In conclusion, these results support that B. melitensis Rev1Δwzm is a promising vaccine candidate for use in pregnant ewes and its efficacy against B. melitensis and B. ovis infections in sheep warrants further study.
Collapse
Affiliation(s)
- Irati Poveda-Urkixo
- Animal Health Department, Institute of Agrobiotechnology (IdAB; CSIC-Navarra Government), Avda. Pamplona 123, 31192 Mutilva, Navarra, Spain
| | - Sara Mena-Bueno
- Animal Health Department, Institute of Agrobiotechnology (IdAB; CSIC-Navarra Government), Avda. Pamplona 123, 31192 Mutilva, Navarra, Spain; Public University of Navarra (UPNA), Avda. de Pamplona 123, 31192 Mutilva, Spain
| | | | - Ana Zabalza-Baranguá
- Animal Health Department, Institute of Agrobiotechnology (IdAB; CSIC-Navarra Government), Avda. Pamplona 123, 31192 Mutilva, Navarra, Spain
| | - Renee M Tsolis
- Medical Microbiology and Immunology, University of California, Davis, 95616 USA
| | - María-Jesús Grilló
- Animal Health Department, Institute of Agrobiotechnology (IdAB; CSIC-Navarra Government), Avda. Pamplona 123, 31192 Mutilva, Navarra, Spain.
| |
Collapse
|
11
|
Maldonado-García JL, Alvarez-Herrera S, Pérez-Sánchez G, Becerril-Villanueva E, Pavón L, Tesoro-Cruz E, Girón-Pérez MI, Hurtado-Alvarado G, Damián-Morales G, López-Santiago R, Moreno-Lafont MC. Concomitant Treatment with Doxycycline and Rifampicin in Balb/c Mice Infected with Brucella abortus 2308 Fails to Reduce Inflammation and Motor Disability. Pharmaceuticals (Basel) 2024; 17:638. [PMID: 38794208 PMCID: PMC11123987 DOI: 10.3390/ph17050638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Brucellosis is an infection widely distributed around the world, and in some countries it is considered a public health problem. Brucellosis causes insidious symptoms that make it difficult to diagnose. Infection can also trigger chronic pain and neuropsychiatric complications. Antibiotics are not always effective to eradicate infection, contributing to chronicity. We aimed to investigate the effects of antibiotic treatment on proinflammatory cytokines, neurotransmitters, corticosterone, and behavior in a murine model of infecrion of B. abortus strain 2308. Four study groups were created: (a) control; (b) antibiotic control; (c) infected with B. abortus 2308; and (d) infected and treated with rifampicin and doxycycline. We determined B. abortus 2308 colony-forming units (CFUs), the count of dendritic cells, and macrophages in the spleen; serum levels of cytokines and corticosterone; levels of serotonin, dopamine, epinephrine, and norepinephrine in the brain; and equilibrium, physical strength, anxiety, and hopelessness tests. The infected and treated mice group was compared with the control and infected mice to assess whether treatment is sufficient to recover neuroimmunoendocrine parameters. Our results showed that despite the treatment of brucellosis with rifampicin and doxycycline, antibiotic-treated mice showed a persistence of B. abortus 2308 CFUs, an increased count in macrophage number, and higher circulating levels of corticosterone. Furthermore, the levels of IL-12, IL-6, and TNF-α remained higher. We found a decrease in muscular strength and equilibrium concomitant to changes in neurotransmitters in the hippocampus, cerebellum, and frontal cortex. Our data suggest that the remaining bacterial load after antibiotic administration favors inflammatory, neurochemical, and behavioral alterations, partly explaining the widespread and paradoxical symptomatology experienced by patients with chronic brucellosis.
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (G.D.-M.); (R.L.-S.)
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico
| | - Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente, Mexico City 14370, Mexico; (S.A.-H.); (G.P.-S.); (E.B.-V.)
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente, Mexico City 14370, Mexico; (S.A.-H.); (G.P.-S.); (E.B.-V.)
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente, Mexico City 14370, Mexico; (S.A.-H.); (G.P.-S.); (E.B.-V.)
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente, Mexico City 14370, Mexico; (S.A.-H.); (G.P.-S.); (E.B.-V.)
| | - Emiliano Tesoro-Cruz
- Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología, Centro Médico Nacional “La Raza”, IMSS, Mexico City 02990, Mexico;
| | | | - Gabriela Hurtado-Alvarado
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico;
| | - Gabriela Damián-Morales
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (G.D.-M.); (R.L.-S.)
| | - Rubén López-Santiago
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (G.D.-M.); (R.L.-S.)
| | - Martha C. Moreno-Lafont
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.M.-G.); (G.D.-M.); (R.L.-S.)
| |
Collapse
|
12
|
Elizalde-Bielsa A, Muñoz PM, Zúñiga-Ripa A, Conde-Álvarez R. A Review on the Methodology and Use of the Pregnant Mouse Model in the Study of Brucella Reproductive Pathogenesis and Its Abortifacient Effect. Microorganisms 2024; 12:866. [PMID: 38792696 PMCID: PMC11123383 DOI: 10.3390/microorganisms12050866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
Brucellosis is one of the most common and widespread bacterial zoonoses and is caused by Gram-negative bacteria belonging to the genus Brucella. These organisms are able to infect and replicate within the placenta, resulting in abortion, one of the main clinical signs of brucellosis. Although the mouse model is widely used to study Brucella virulence and, more recently, to evaluate the protection of new vaccines, there is no clear consensus on the experimental conditions (e.g., mouse strains, doses, routes of inoculation, infection/pregnancy time) and the natural host reproducibility of the pregnant mouse model for reproductive brucellosis. This lack of consensus calls for a review that integrates the major findings regarding the effect of Brucella wild-type and vaccine strains infections on mouse pregnancy. We found sufficient evidence on the utility of the pregnant mouse model to study Brucella-induced placentitis and abortion and propose suitable experimental conditions (dose, time of infection) and pregnancy outcome readouts for B. abortus and B. melitensis studies. Finally, we discuss the utility and limitations of the pregnant mouse as a predictive model for the abortifacient effect of live Brucella vaccines.
Collapse
Affiliation(s)
- Aitor Elizalde-Bielsa
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain;
| | - Pilar M. Muñoz
- Department of Animal Science, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), 50059 Zaragoza, Spain;
- Instituto Agroalimentario de Aragón—IA2, CITA-Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Amaia Zúñiga-Ripa
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain;
| | - Raquel Conde-Álvarez
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain;
| |
Collapse
|
13
|
Abushahba MFN, Dadelahi AS, Ponzilacqua-Silva B, Moley CR, Skyberg JA. Contrasting roles for IgM and B-cell MHCII expression in Brucella abortus S19 vaccine-mediated efficacy against B. melitensis infection. mSphere 2024; 9:e0075023. [PMID: 38349167 PMCID: PMC10964430 DOI: 10.1128/msphere.00750-23] [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: 12/01/2023] [Accepted: 01/16/2024] [Indexed: 02/15/2024] Open
Abstract
Brucellosis, caused by the bacterium Brucella, poses a significant global threat to both animal and human health. Although commercial live Brucella vaccines including S19, RB51, and Rev1 are available for animals, their unsuitability for human use and incomplete efficacy in animals necessitate the further study of vaccine-mediated immunity to Brucella. In this study, we employed in vivo B-cell depletion, as well as immunodeficient and transgenic mouse models, to comprehensively investigate the roles of B cells, antigen uptake and presentation, antibody production, and class switching in the context of S19-mediated immunity against brucellosis. We found that antibody production, and in particular secretory IgM plays a protective role in S19-mediated immunity against virulent Brucella melitensis early after the challenge in a manner associated with complement activation. While T follicular helper cell deficiency dampened IgG production and vaccine efficacy at later stages of the challenge, this effect appeared to be independent of antibody production and rather was associated with altered T-cell function. By contrast, B-cell MHCII expression negatively impacted vaccine efficacy at later timepoints after the challenge. In addition, B-cell depletion after vaccination, but before the challenge, enhanced S19-mediated protection against brucellosis, suggesting a deleterious role of B cells during the challenge phase. Collectively, our findings indicate antibody production is protective, while B-cell MHCII expression is deleterious, to live vaccine-mediated immunity against brucellosis. IMPORTANCE Brucella is a neglected zoonotic pathogen with a worldwide distribution. Our study delves into B-cell effector functions in live vaccine-mediated immunity against brucellosis. Notably, we found antibody production, particularly secretory IgM, confers protection against virulent Brucella melitensis in vaccinated mice, which was associated with complement activation. By contrast, B-cell MHCII expression negatively impacted vaccine efficacy. In addition, B-cell depletion after vaccination, but before the B. melitensis challenge, enhanced protection against infection, suggesting a detrimental B-cell role during the challenge phase. Interestingly, deficiency of T follicular helper cells, which are crucial for aiding germinal center B cells, dampened vaccine efficacy at later stages of challenge independent of antibody production. This study underscores contrasting and phase-dependent roles of B-cell effector functions in vaccine-mediated immunity against Brucella.
Collapse
Affiliation(s)
- Mostafa F. N. Abushahba
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, USA
- Department of Zoonoses, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Alexis S. Dadelahi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, USA
| | - Bárbara Ponzilacqua-Silva
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, USA
| | - Charles R. Moley
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, USA
| | - Jerod A. Skyberg
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
14
|
Carvalho TP, Toledo FAO, Bautista DFA, Silva MF, Oliveira JBS, Lima PA, Costa FB, Ribeiro NQ, Lee JY, Birbrair A, Paixão TA, Tsolis RM, Santos RL. Pericytes modulate endothelial inflammatory response during bacterial infection. mBio 2024; 15:e0325223. [PMID: 38289074 PMCID: PMC10936204 DOI: 10.1128/mbio.03252-23] [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: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 03/14/2024] Open
Abstract
Pericytes are located around blood vessels, in close contact with endothelial cells. We discovered that pericytes dampen pro-inflammatory endothelial cell responses. Endothelial cells co-cultured with pericytes had markedly reduced expression of adhesion molecules (PECAM-1 and ICAM-1) and proinflammatory cytokines (CCL-2 and IL-6) in response to bacterial stimuli (Brucella ovis, Listeria monocytogenes, or Escherichia coli lipopolysaccharide). Pericyte-depleted mice intraperitoneally inoculated with either B. ovis, a stealthy pathogen that does not trigger detectable inflammation, or Listeria monocytogenes, developed peritonitis. Further, during Citrobacter rodentium infection, pericyte-depleted mice developed severe intestinal inflammation, which was not evident in control mice. The anti-inflammatory effect of pericytes required connexin 43, as either chemical inhibition or silencing of connexin 43 abrogated pericyte-mediated suppression of endothelial inflammatory responses. Our results define a mechanism by which pericytes modulate inflammation during infection, which shifts our understanding of pericyte biology: from a structural cell to a pro-active player in modulating inflammation. IMPORTANCE A previously unknown mechanism by which pericytes modulate inflammation was discovered. The absence of pericytes or blocking interaction between pericytes and endothelium through connexin 43 results in stronger inflammation, which shifts our understanding of pericyte biology, from a structural cell to a player in controlling inflammation.
Collapse
Affiliation(s)
- Thaynara P. Carvalho
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Frank A. O. Toledo
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Diego F. A. Bautista
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Monique F. Silva
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jefferson B. S. Oliveira
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pâmela A. Lima
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fabíola B. Costa
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Noelly Q. Ribeiro
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jee-Yon Lee
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Alexander Birbrair
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tatiane A. Paixão
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Reneé M. Tsolis
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Renato L. Santos
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| |
Collapse
|
15
|
Marinho FV, Brito C, de Araujo ACVSC, Oliveira SC. Guanylate-binding protein-5 is involved in inflammasome activation by bacterial DNA but only the cooperation of multiple GBPs accounts for control of Brucella abortus infection. Front Immunol 2024; 15:1341464. [PMID: 38404575 PMCID: PMC10885698 DOI: 10.3389/fimmu.2024.1341464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Guanylate-binding proteins (GBPs) are produced in response to pro-inflammatory signals, mainly interferons. The most studied cluster of GBPs in mice is on chromosome 3. It comprises the genes for GBP1-to-3, GBP5 and GBP7. In humans, all GBPs are present in a single cluster on chromosome 1. Brucella abortus is a Gram-negative bacterium known to cause brucellosis, a debilitating disease that affects both humans and animals. Our group demonstrated previously that GBPs present on murine chromosome 3 (GBPchr3) is important to disrupt Brucella-containing vacuole and GBP5 itself is important to Brucella intracellular LPS recognition. In this work, we investigated further the role of GBPs during B. abortus infection. Methods and results We observed that all GBPs from murine chromosome 3 are significantly upregulated in response to B. abortus infection in mouse bone marrow-derived macrophages. Of note, GBP5 presents the highest expression level in all time points evaluated. However, only GBPchr3-/- cells presented increased bacterial burden compared to wild-type macrophages. Brucella DNA is an important Pathogen-Associated Molecular Pattern that could be available for inflammasome activation after BCV disruption mediated by GBPs. In this regard, we observed reduced IL-1β production in the absence of GBP2 or GBP5, as well as in GBPchr3-/- murine macrophages. Similar result was showed by THP-1 macrophages with downregulation of GBP2 and GBP5 mediated by siRNA. Furthermore, significant reduction on caspase-1 p20 levels, LDH release and Gasdermin-D conversion into its mature form (p30 N-terminal subunit) was observed only in GBPchr3-/- macrophages. In an in vivo perspective, we found that GBPchr3-/- mice had increased B. abortus burden and higher number of granulomas per area of liver tissue, indicating increased disease severity. Discussion/conclusion Altogether, these results demonstrate that although GBP5 presents a high expression pattern and is involved in inflammasome activation by bacterial DNA in macrophages, the cooperation of multiple GBPs from murine chromosome 3 is necessary for full control of Brucella abortus infection.
Collapse
Affiliation(s)
- Fabio V. Marinho
- Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Brito
- Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Carolina V. S. C. de Araujo
- Instituto de Ciências Biológicas, Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto de Ciências Biomédicas, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Sergio C. Oliveira
- Instituto de Ciências Biológicas, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto de Ciências Biomédicas, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Yang J, Wang Y, Hou Y, Sun M, Xia T, Wu X. Evasion of host defense by Brucella. CELL INSIGHT 2024; 3:100143. [PMID: 38250017 PMCID: PMC10797155 DOI: 10.1016/j.cellin.2023.100143] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024]
Abstract
Brucella , an adept intracellular pathogen, causes brucellosis, a zoonotic disease leading to significant global impacts on animal welfare and the economy. Regrettably, there is currently no approved and effective vaccine for human use. The ability of Brucella to evade host defenses is essential for establishing chronic infection and ensuring stable intracellular growth. Brucella employs various mechanisms to evade and undermine the innate and adaptive immune responses of the host through modulating the activation of pattern recognition receptors (PRRs), inflammatory responses, or the activation of immune cells like dendritic cells (DCs) to inhibit antigen presentation. Moreover, it regulates multiple cellular processes such as apoptosis, pyroptosis, and autophagy to establish persistent infection within host cells. This review summarizes the recently discovered mechanisms employed by Brucella to subvert host immune responses and research progress on vaccines, with the aim of advancing our understanding of brucellosis and facilitating the development of more effective vaccines and therapeutic approaches against Brucella .
Collapse
Affiliation(s)
- Jinke Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yue Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yuanpan Hou
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Mengyao Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Tian Xia
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Xin Wu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| |
Collapse
|
17
|
Wang S, Yin Y, Zai X, Gu Y, Guo F, Shao F, Zhang Y, Li Y, Li R, Zhang J, Xu J, Chen W. A novel Galleria mellonella experimental model for zoonotic pathogen Brucella. Virulence 2023; 14:2268496. [PMID: 37817444 PMCID: PMC10599192 DOI: 10.1080/21505594.2023.2268496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023] Open
Abstract
Brucellosis is a major threat to public health and animal husbandry. Several in vivo vertebrate models, such as mice, guinea pigs, and nonhuman primates, have been used to study Brucella pathogenesis, bacteria-host interactions, and vaccine efficacy. However, these models have limitations whereas the invertebrate Galleria mellonella model is a cost-effective and ethical alternative. The aim of the present study was to examine the invertebrate G. mellonella as an in vivo infection model for Brucella. Infection assays were employed to validate the fitness of the larval model for Brucella infection and virulence evaluation. The protective efficacy of immune sera was evaluated by pre-incubated with a lethal dose of bacteria before infection. The consistency between the mouse model and the larval model was confirmed by assessing the protective efficacy of two Brucella vaccine strains. The results show that G. mellonella could be infected by Brucella strains, in a dose- and temperature-dependent way. Moreover, this larval model can effectively evaluate the virulence of Brucella strains in a manner consistent with that of mammalian infection models. Importantly, this model can assess the protective efficacy of vaccine immune sera within a day. Further investigation implied that haemolymph played a crucial role in the protective efficacy of immune sera. In conclusion, G. mellonella could serve as a quick, efficient, and reliable model for evaluating the virulence of Brucella strains and efficacy of immune sera in an ethical manner.
Collapse
Affiliation(s)
- Shuyi Wang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Ying Yin
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaodong Zai
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yanfei Gu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Fengyu Guo
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Fangze Shao
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yue Zhang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yaohui Li
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Ruihua Li
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Jun Zhang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Junjie Xu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Wei Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
18
|
Blasco JM, Moreno E, Muñoz PM, Conde-Álvarez R, Moriyón I. A review of three decades of use of the cattle brucellosis rough vaccine Brucella abortus RB51: myths and facts. BMC Vet Res 2023; 19:211. [PMID: 37853407 PMCID: PMC10583465 DOI: 10.1186/s12917-023-03773-3] [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: 08/14/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023] Open
Abstract
Cattle brucellosis is a severe zoonosis of worldwide distribution caused by Brucella abortus and B. melitensis. In some countries with appropriate infrastructure, animal tagging and movement control, eradication was possible through efficient diagnosis and vaccination with B. abortus S19, usually combined with test-and-slaughter (T/S). Although S19 elicits anti-smooth lipopolysaccharide antibodies that may interfere in the differentiation of infected and vaccinated animals (DIVA), this issue is minimized using appropriate S19 vaccination protocols and irrelevant when high-prevalence makes mass vaccination necessary or when eradication requisites are not met. However, S19 has been broadly replaced by vaccine RB51 (a rifampin-resistant rough mutant) as it is widely accepted that is DIVA, safe and as protective as S19. These RB51 properties are critically reviewed here using the evidence accumulated in the last 35 years. Controlled experiments and field evidence shows that RB51 interferes in immunosorbent assays (iELISA, cELISA and others) and in complement fixation, issues accentuated by revaccinating animals previously immunized with RB51 or S19. Moreover, contacts with virulent brucellae elicit anti-smooth lipopolysaccharide antibodies in RB51 vaccinated animals. Thus, accepting that RB51 is truly DIVA results in extended diagnostic confusions and, when combined with T/S, unnecessary over-culling. Studies supporting the safety of RB51 are flawed and, on the contrary, there is solid evidence that RB51 is excreted in milk and abortifacient in pregnant animals, thus being released in abortions and vaginal fluids. These problems are accentuated by the RB51 virulence in humans, lack diagnostic serological tests detecting these infections and RB51 rifampicin resistance. In controlled experiments, protection by RB51 compares unfavorably with S19 and lasts less than four years with no evidence that RB51-revaccination bolsters immunity, and field studies reporting its usefulness are flawed. There is no evidence that RB51 protects cattle against B. melitensis, infection common when raised together with small ruminants. Finally, data acumulated during cattle brucellosis eradication in Spain shows that S19-T/S is far more efficacious than RB51-T/S, which does not differ from T/S alone. We conclude that the assumption that RB51 is DIVA, safe, and efficaceous results from the uncritical repetition of imperfectly examined evidence, and advise against its use.
Collapse
Affiliation(s)
- J M Blasco
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, España
| | - E Moreno
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - P M Muñoz
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, España
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, España
| | - R Conde-Álvarez
- Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - I Moriyón
- Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
19
|
Euba B, Gil-Campillo C, Asensio-López J, López-López N, Sen-Kilic E, Díez-Martínez R, Burgui S, Barbier M, Garmendia J. In Vivo Genome-Wide Gene Expression Profiling Reveals That Haemophilus influenzae Purine Synthesis Pathway Benefits Its Infectivity within the Airways. Microbiol Spectr 2023; 11:e0082323. [PMID: 37195232 PMCID: PMC10269889 DOI: 10.1128/spectrum.00823-23] [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: 02/23/2023] [Accepted: 04/27/2023] [Indexed: 05/18/2023] Open
Abstract
Haemophilus influenzae is a human-adapted bacterial pathogen that causes airway infections. Bacterial and host elements associated with the fitness of H. influenzae within the host lung are not well understood. Here, we exploited the strength of in vivo-omic analyses to study host-microbe interactions during infection. We used in vivo transcriptome sequencing (RNA-seq) for genome-wide profiling of both host and bacterial gene expression during mouse lung infection. Profiling of murine lung gene expression upon infection showed upregulation of lung inflammatory response and ribosomal organization genes, and downregulation of cell adhesion and cytoskeleton genes. Transcriptomic analysis of bacteria recovered from bronchoalveolar lavage fluid samples from infected mice showed a significant metabolic rewiring during infection, which was highly different from that obtained upon bacterial in vitro growth in an artificial sputum medium suitable for H. influenzae. In vivo RNA-seq revealed upregulation of bacterial de novo purine biosynthesis, genes involved in non-aromatic amino acid biosynthesis, and part of the natural competence machinery. In contrast, the expression of genes involved in fatty acid and cell wall synthesis and lipooligosaccharide decoration was downregulated. Correlations between upregulated gene expression and mutant attenuation in vivo were established, as observed upon purH gene inactivation leading to purine auxotrophy. Likewise, the purine analogs 6-thioguanine and 6-mercaptopurine reduced H. influenzae viability in a dose-dependent manner. These data expand our understanding of H. influenzae requirements during infection. In particular, H. influenzae exploits purine nucleotide synthesis as a fitness determinant, raising the possibility of purine synthesis as an anti-H. influenzae target. IMPORTANCE In vivo-omic strategies offer great opportunities for increased understanding of host-pathogen interplay and for identification of therapeutic targets. Here, using transcriptome sequencing, we profiled host and pathogen gene expression during H. influenzae infection within the murine airways. Lung pro-inflammatory gene expression reprogramming was observed. Moreover, we uncovered bacterial metabolic requirements during infection. In particular, we identified purine synthesis as a key player, highlighting that H. influenzae may face restrictions in purine nucleotide availability within the host airways. Therefore, blocking this biosynthetic process may have therapeutic potential, as supported by the observed inhibitory effect of 6-thioguanine and 6-mercaptopurine on H. influenzae growth. Together, we present key outcomes and challenges for implementing in vivo-omics in bacterial airway pathogenesis. Our findings provide metabolic insights into H. influenzae infection biology, raising the possibility of purine synthesis as an anti-H. influenzae target and of purine analog repurposing as an antimicrobial strategy against this pathogen.
Collapse
Affiliation(s)
- Begoña Euba
- Instituto de Agrobiotecnología (IDAB), Consejo Superior de Investigaciones Científicas (CSIC)-Gobierno de Navarra, Mutilva, Spain
| | - Celia Gil-Campillo
- Instituto de Agrobiotecnología (IDAB), Consejo Superior de Investigaciones Científicas (CSIC)-Gobierno de Navarra, Mutilva, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Javier Asensio-López
- Instituto de Agrobiotecnología (IDAB), Consejo Superior de Investigaciones Científicas (CSIC)-Gobierno de Navarra, Mutilva, Spain
- Asociación de la Industria Navarra (AIN)-Gobierno de Navarra, Cordovilla, Spain
| | - Nahikari López-López
- Instituto de Agrobiotecnología (IDAB), Consejo Superior de Investigaciones Científicas (CSIC)-Gobierno de Navarra, Mutilva, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Emel Sen-Kilic
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | | | - Saioa Burgui
- Asociación de la Industria Navarra (AIN)-Gobierno de Navarra, Cordovilla, Spain
| | - Mariette Barbier
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Junkal Garmendia
- Instituto de Agrobiotecnología (IDAB), Consejo Superior de Investigaciones Científicas (CSIC)-Gobierno de Navarra, Mutilva, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Conexión Nanomedicina-CSIC, Madrid, Spain
| |
Collapse
|
20
|
de Carvalho TP, da Silva LA, Castanheira TLL, de Souza TD, da Paixão TA, Lazaro-Anton L, Tsolis RM, Santos RL. Cell and Tissue Tropism of Brucella spp. Infect Immun 2023; 91:e0006223. [PMID: 37129522 PMCID: PMC10187126 DOI: 10.1128/iai.00062-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Brucella spp. are facultatively intracellular bacteria that can infect, survive, and multiply in various host cell types in vivo and/or in vitro. The genus Brucella has markedly expanded in recent years with the identification of novel species and hosts, which has revealed additional information about the cell and tissue tropism of these pathogens. Classically, Brucella spp. are considered to have tropism for organs that contain large populations of phagocytes such as lymph nodes, spleen, and liver, as well as for organs of the genital system, including the uterus, epididymis, testis, and placenta. However, experimental infections of several different cultured cell types indicate that Brucella may actually have a broader cell tropism than previously thought. Indeed, recent studies indicate that certain Brucella species in particular hosts may display a pantropic distribution in vivo. This review discusses the available knowledge on cell and tissue tropism of Brucella spp. in natural infections of various host species, as well as in experimental animal models and cultured cells.
Collapse
Affiliation(s)
- Thaynara Parente de Carvalho
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Laice Alves da Silva
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaís Larissa Lourenço Castanheira
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto Federal de Educação Ciência e Tecnologia do Norte de Minas Gerais, Salinas, Brazil
| | - Tayse Domingues de Souza
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tatiane Alves da Paixão
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leticia Lazaro-Anton
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Renee M. Tsolis
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| | - Renato Lima Santos
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Medical Microbiology and Immunology, University of California – Davis, Davis, California, USA
| |
Collapse
|
21
|
Wang H, Clapp B, Hoffman C, Yang X, Pascual DW. A Single Nasal Dose Vaccination with a Brucella abortus Mutant Potently Protects against Pulmonary Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1576-1588. [PMID: 37036290 PMCID: PMC10159994 DOI: 10.4049/jimmunol.2300071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023]
Abstract
The Brucella abortus double-mutant (ΔznuA ΔnorD Brucella abortus-lacZ [znBAZ]) was assessed for its protective efficacy after vaccination with a single nasal dose. Superior protection was achieved in znBAZ-vaccinated mice against pulmonary, wild-type B. abortus 2308 challenge when compared with conventional livestock Brucella abortus vaccines, the smooth S19 (smooth B. abortus strain 19 vaccine) and rough RB51 (rough mutant vaccine strain of B. abortus) strains. Nasal znBAZ vaccination reduced splenic and lung colonization by wild-type brucellae by >3-4 logs. In contrast, S19 reduced lung colonization by only 32-fold, and RB51 failed to reduce colonization. One profound attribute of znBAZ vaccination was the >3-fold increase in pulmonary CD8+ T cells when compared with other vaccinated groups. S19 vaccination increased only CD4+ T cells. All vaccines induced IFN-γ and TNF-α production by CD4+ T cells, but only znBAZ vaccination enhanced the recruitment of polyfunctional CD8+ T cells, by >100-fold. IL-17 by both CD4+ and CD8+ T cells was also induced by subsequent znBAZ vaccination. These results demonstrate that, in addition to achieving protective immunity by CD4+ T cells, CD8+ T cells, specifically resident memory T cells, also confer protection against brucellosis. The protection obtained by znBAZ vaccination was attributed to IFN-γ-producing CD8+ T cells, because depletion of CD8+ T cells throughout vaccination and challenge phases abrogated protection. The stimulation of only CD4+ T cells by RB51- and S19-vaccinated mice proved insufficient in protecting against pulmonary B. abortus 2308 challenge. Thus, nasal znBAZ vaccination offers an alternative means to elicit protection against brucellosis.
Collapse
Affiliation(s)
- Hongbin Wang
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| | - Beata Clapp
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| | - Carol Hoffman
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| | - Xinghong Yang
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| | - David W. Pascual
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL
| |
Collapse
|
22
|
Montero‐Blay A, Blanco JD, Rodriguez‐Arce I, Lastrucci C, Piñero‐Lambea C, Lluch‐Senar M, Serrano L. Bacterial expression of a designed single-chain IL-10 prevents severe lung inflammation. Mol Syst Biol 2023; 19:e11037. [PMID: 36598022 PMCID: PMC9834763 DOI: 10.15252/msb.202211037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 01/05/2023] Open
Abstract
Interleukin-10 (IL-10) is an anti-inflammatory cytokine that is active as a swapped domain dimer and is used in bacterial therapy of gut inflammation. IL-10 can be used as treatment of a wide range of pulmonary diseases. Here we have developed a non-pathogenic chassis (CV8) of the human lung bacterium Mycoplasma pneumoniae (MPN) to treat lung diseases. We find that IL-10 expression by MPN has a limited impact on the lung inflammatory response in mice. To solve these issues, we rationally designed a single-chain IL-10 (SC-IL10) with or without surface mutations, using our protein design software (ModelX and FoldX). As compared to the IL-10 WT, the designed SC-IL10 molecules increase the effective expression in MPN four-fold, and the activity in mouse and human cell lines between 10 and 60 times, depending on the cell line. The SC-IL10 molecules expressed in the mouse lung by CV8 in vivo have a powerful anti-inflammatory effect on Pseudomonas aeruginosa lung infection. This rational design strategy could be used to other molecules with immunomodulatory properties used in bacterial therapy.
Collapse
Affiliation(s)
- Ariadna Montero‐Blay
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Javier Delgado Blanco
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Irene Rodriguez‐Arce
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Claire Lastrucci
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Carlos Piñero‐Lambea
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Maria Lluch‐Senar
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Luis Serrano
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- ICREABarcelonaSpain
| |
Collapse
|
23
|
Li S, Huang J, Wang K, Liu Y, Guo Y, Li X, Wu J, Sun P, Wang Y, Zhu L, Wang H. A bioconjugate vaccine against Brucella abortus produced by engineered Escherichia coli. Front Bioeng Biotechnol 2023; 11:1121074. [PMID: 36911199 PMCID: PMC9995886 DOI: 10.3389/fbioe.2023.1121074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Brucellosis, mainly caused by Brucella, is a widespread zoonotic disease worldwide, with no available effective vaccine for human use. Recently, bioconjugate vaccines against Brucella have been prepared in Yersinia enterocolitica O:9 (YeO9), whose O-antigen structure is similar to that of Brucella abortus. However, the pathogenicity of YeO9 still hinders the large-scale production of these bioconjugate vaccines. Here, an attractive system for the preparation of bioconjugate vaccines against Brucella was established in engineered E. coli. Briefly, the OPS gene cluster of YeO9 was modularized into five individual fragments and reassembled using synthetic biological methods through standardized interfaces, then introduced into E. coli. After confirming the synthesis of targeted antigenic polysaccharides, the exogenous protein glycosylation system (PglL system) was used to prepare the bioconjugate vaccines. A series of experiments were conducted to demonstrate that the bioconjugate vaccine could effectively evoke humoral immune responses and induce the production of specific antibodies against B. abortus A19 lipopolysaccharide. Furthermore, the bioconjugate vaccines provide protective roles in both lethal and non-lethal challenge of B. abortus A19 strain. Using the engineered E. coli as a safer chassis to prepare bioconjugate vaccines against B. abortus paves the way for future industrial applications.
Collapse
Affiliation(s)
- Shulei Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China.,The Third Medical Center, PLA General Hospital, Beijing, China.,Department of Clinical Laboratory, The Third Medical Centre of Chinese PLA General Hospital, The Training Site for Postgraduate of Jin Zhou Medical University, Beijing, China
| | - Jing Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China.,Beijing Minhai Biotechnology Co., Ltd., Beijing, China
| | - Kangfeng Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Yan Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Xiang Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Jun Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Peng Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Yufei Wang
- The Third Medical Center, PLA General Hospital, Beijing, China.,Department of Clinical Laboratory, The Third Medical Centre of Chinese PLA General Hospital, The Training Site for Postgraduate of Jin Zhou Medical University, Beijing, China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
24
|
Guo X, Zeng H, Li M, Xiao Y, Gu G, Song Z, Shuai X, Guo J, Huang Q, Zhou B, Chu Y, Jiao H. The mechanism of chronic intracellular infection with Brucella spp. Front Cell Infect Microbiol 2023; 13:1129172. [PMID: 37143745 PMCID: PMC10151771 DOI: 10.3389/fcimb.2023.1129172] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Globally, brucellosis is a widespread zoonotic disease. It is prevalent in more than 170 countries and regions. It mostly damages an animal's reproductive system and causes extreme economic losses to the animal husbandry industry. Once inside cells, Brucella resides in a vacuole, designated the BCV, which interacts with components of the endocytic and secretory pathways to ensure bacterial survival. Numerous studies conducted recently have revealed that Brucella's ability to cause a chronic infection depends on how it interacts with the host. This paper describes the immune system, apoptosis, and metabolic control of host cells as part of the mechanism of Brucella survival in host cells. Brucella contributes to both the body's non-specific and specific immunity during chronic infection, and it can aid in its survival by causing the body's immune system to become suppressed. In addition, Brucella regulates apoptosis to avoid being detected by the host immune system. The BvrR/BvrS, VjbR, BlxR, and BPE123 proteins enable Brucella to fine-tune its metabolism while also ensuring its survival and replication and improving its ability to adapt to the intracellular environment.
Collapse
Affiliation(s)
- Xiaoyi Guo
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Hui Zeng
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mengjuan Li
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yu Xiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Guojing Gu
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenhui Song
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xuehong Shuai
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jianhua Guo
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qingzhou Huang
- The College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Bo Zhou
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| | - Yuefeng Chu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| | - Hanwei Jiao
- The College of Veterinary Medicine, Southwest University, Chongqing, China
- The Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Bo Zhou, ; Yuefeng Chu, ; Hanwei Jiao,
| |
Collapse
|
25
|
Ma C, Li H, Lu S, Li X, Wang S, Wang W. Ocular Lesions in Brucella Infection: A Review of the Literature. Infect Drug Resist 2022; 15:7601-7617. [PMID: 36579126 PMCID: PMC9791996 DOI: 10.2147/idr.s394497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Ocular lesions due to Brucella infection are uncommon and easily overlooked in clinical management, but must be differentiated from non-infectious eye diseases and treated promptly to protect the patient's vision. We reviewed the relevant literature and identified 47 patients with ocular complications of Brucella infection. Among them, 28 showed ocular neuropathy, 15 presented with uveitis, and four patients displayed other ocular symptoms. Ocular symptoms accompanying Brucella infection require prompt diagnosis and treatment. The main methods of diagnosis are intraocular fluid tests and blood tests. Early diagnosis and treatment with suitable antibiotics are central to protecting the patient's vision. Notably, in terms of mechanism of injury, Brucella infection is chronic and cannot be eliminated by phagocytes, and can cause damage to the eye by inducing autoimmune reactions, antigen-antibody complex production, release of endogenous and exogenous toxins, and bacterial production of septic thrombi in the tissues. In this review, we summarize the ocular symptoms, diagnosis, treatment and prognosis of Brucella infection, and discuss the mechanisms of Brucella in ocular lesions, providing a reference for the diagnosis and treatment of Brucella ocular lesions.
Collapse
Affiliation(s)
- Chao Ma
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Haoyu Li
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, Hunan, People’s Republic of China
| | - Shuwen Lu
- Department of Ophthalmology, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, People’s Republic of China
| | - Xian Li
- Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester, England
- School of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, the University of Manchester, Manchester, England
| | - Shuai Wang
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Wenzhan Wang
- Department of Ophthalmology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| |
Collapse
|
26
|
Hensel ME, Stranahan LW, Edwards JF, Arenas-Gamboa AM. Intratracheal inoculation results in Brucella-associated reproductive disease in male mouse and guinea pig models of infection. Front Microbiol 2022; 13:1029199. [DOI: 10.3389/fmicb.2022.1029199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Brucella species are considered a significant cause of reproductive pathology in male and female animals. Importantly, Brucella melitensis can induce reproductive disease in humans. Reproductive pathogenesis and evaluation of newly developed countermeasures against brucellosis studies have traditionally utilized female animal models. However, any potential, new intervention for use in humans would need to be evaluated in both sexes. Therefore, animal models for male reproductive brucellosis are desperately needed to understand disease progression. Accordingly, we evaluated guinea pigs and mice using B. melitensis 16 M in an intratracheal model of inoculation at different stages of infection (peracute, acute, and chronic) with an emphasis on determining the effect to the male reproductive organs. Aerosol inoculation resulted in colonization of the reproductive organs (testicle, epididymis, prostate) in both species. Infection peaked during the peracute (1-week post-infection [p.i.]) and acute (2-weeks p.i.) stages of infection in the mouse in spleen, epididymis, prostate, and testicle, but colonization was poorly associated with inflammation. In the guinea pig, peak infection was during the acute stage (4-weeks p.i.) and resulted in inflammation that disrupted spermatogenesis chronically. To determine if vaccine efficacy could be evaluated using these models, males were vaccinated using subcutaneous injection with vaccine candidate 16 MΔvjbR at 109 CFU/100 μl followed by intratracheal challenge with 16 M at 107. Interestingly, vaccination efficacy varied between species and reproductive organs demonstrating the value of evaluating vaccine candidates in multiple models and sexes. Vaccination resulted in a significant reduction in colonization in the mouse, but this could not be correlated with a decrease in inflammation. Due to the ability to evaluate for both colonization and inflammation, guinea pigs seemed the better model not only for assessing host-pathogen interactions but also for future vaccine development efforts.
Collapse
|
27
|
Goodwin ZI, Yang X, Hoffman C, Pascual DW. Live mucosal vaccination stimulates potent protection via varied CD4+ and CD8+ T cell subsets against wild-type Brucella melitensis 16M challenge. Front Immunol 2022; 13:995327. [PMID: 36263034 PMCID: PMC9574439 DOI: 10.3389/fimmu.2022.995327] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/31/2022] [Indexed: 12/01/2022] Open
Abstract
Re-emerging zoonotic pathogen Brucella spp. continues to impact developing countries and persists in expanding populations of wildlife species in the US, constantly threatening infection of our domestic herds. The development of improved animal and human vaccines remains a priority. In this study, immunity to a novel live attenuated B. melitensis strain, termed znBM-mC, was characterized. An oral prime, intranasal (IN) boost strategy conferred exquisite protection against pulmonary challenge, with wild-type (wt) B. melitensis providing nearly complete protection in the lungs and spleens from brucellae colonization. Vaccination with znBM-mC showed an IFN-γ+ CD8+ T-cell bias in the lungs as opposed to Rev 1-vaccinated mice showing IFN-γ+ CD4+ T-cell inclination. Lung CD4+ and CD8+ effector memory T cells (TEMs) increased over 200-fold; and lung CD4+ and CD8+ resident memory T cells (TRMs) increased more than 250- and 150-fold, respectively. These T cells served as the primary producers of IFN-γ in the lungs, which was essential for vaccine clearance and the predominant cytokine generated pre-and post-challenge with wt B. melitensis 16M; znBM-mC growth could not be arrested in IFN-γ−/− mice. Increases in lung TNF-α and IL-17 were also induced, with IL-17 being mostly derived from CD4+ T cells. Vaccination of CD4−/−, CD8−/−, and B6 mice with znBM-mC conferred full protection in the lungs and spleens post-pulmonary challenge with virulent B. melitensis; vaccination of IL-17−/− mice resulted in the protection of the lungs, but not the spleen. These data demonstrate the efficacy of mucosal vaccine administration for the generation of protective memory T cells against wt B. melitensis.
Collapse
|
28
|
Reyes AWB, Kim H, Huy TXN, Nguyen TT, Min W, Lee D, Hur J, Lee JH, Kim S. The In Vitro and In Vivo Effect of Lipoxygenase Pathway Inhibitors Nordihydroguaiaretic Acid and Its Derivative Tetra- O-methyl Nordihydroguaiaretic Acid against Brucella abortus 544. J Microbiol Biotechnol 2022; 32:1126-1133. [PMID: 36039381 PMCID: PMC9628969 DOI: 10.4014/jmb.2207.07026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/09/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022]
Abstract
This study investigated the contribution of lipoxygenase (LOX) inhibitors, nordihydroguaiaretic acid (NDGA), tetra-O-methyl nordihydroguaiaretic acid (M4N) and zileuton (ZIL), and thromboxane A2 (TXA2) inhibitor 4,5-diphenylimidazole (DPI) in the proliferation of Brucella abortus infection. None of the compounds affected the uptake of Brucella into the macrophages. We determined the effect of neutralizing leukotriene B4 (LTB4) receptor and showed that the uptake of the bacteria was inhibited at 30 min post-infection. M4N treatment attenuated intracellular survival of Brucella at 2 h post-incubation but it was not observed in the succeeding time points. DPI treatment showed reduced survival of Brucella at 24 h post-incubation while blocking LTB4 receptor was observed to have a lower intracellular growth at 48 h post-incubation suggesting different action of the inhibitors in the course of the survival of Brucella within the cells. Reduced proliferation of the bacteria in the spleens of mice was observed in animals treated with ZIL or DPI. Increased serum cytokine level of TNF-α and MCP-1 was observed in mice treated with M4N or ZIL while a lower IFN-γ level in ZIL-treated mice and a higher IL-12 serum level in DPI-treated mice were observed at 7 d post-infection. At 14 d post-infection, ZIL-treated mice displayed reduced serum level of IL-12 and IL-10. Overall, inhibition of 5-LOX or TXA2 or a combination therapy promises a potential alternative therapy against B. abortus infection. Furthermore, strong ligands for LTB4 receptor could also be a good candidate for the control of Brucella infection.
Collapse
Affiliation(s)
- Alisha Wehdnesday Bernardo Reyes
- Department of Veterinary Paraclinical Sciences, College of Veterinary Medicine, University of the Philippines Los Baños, College, Laguna 4031, Philippines
| | - Heejin Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Tran Xuan Ngoc Huy
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Trang Thi Nguyen
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Wongi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Dongho Lee
- College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Jin Hur
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Suk Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea,Corresponding author Phone +82-55-772-2359 Fax: +82-55-772-2349 E-mail:
| |
Collapse
|
29
|
Mena-Bueno S, Poveda-Urkixo I, Irazoki O, Palacios L, Cava F, Zabalza-Baranguá A, Grilló MJ. Brucella melitensis Wzm/Wzt System: Changes in the Bacterial Envelope Lead to Improved Rev1Δwzm Vaccine Properties. Front Microbiol 2022; 13:908495. [PMID: 35875565 PMCID: PMC9306315 DOI: 10.3389/fmicb.2022.908495] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
The lipopolysaccharide (LPS) O-polysaccharide (O-PS) is the main virulence factor in Brucella. After synthesis in the cytoplasmic membrane, O-PS is exported to the periplasm by the Wzm/Wzt system, where it is assembled into a LPS. This translocation also engages a bactoprenol carrier required for further biosynthesis pathways, such as cell wall biogenesis. Targeting O-PS export by blockage holds great potential for vaccine development, but little is known about the biological implications of each Wzm/Wzt moiety. To improve this knowledge and to elucidate its potential application as a vaccine, we constructed and studied wzm/wzt single- and double-deletion mutants, using the attenuated strain Brucella melitensis Rev1 as the parental strain. This allowed us to describe the composition of Brucella peptidoglycan for the first time. We observed that these mutants lack external O-PS yet trigger changes in genetic transcription and in phenotypic properties associated with the outer membrane and cell wall. The three mutants are highly attenuated; unexpectedly, Rev1Δwzm also excels as an immunogenic and effective vaccine against B. melitensis and Brucella ovis in mice, revealing that low persistence is not at odds with efficacy. Rev1Δwzm is attenuated in BeWo trophoblasts, does not infect mouse placentas, and is safe in pregnant ewes. Overall, these attributes and the minimal serological interference induced in sheep make Rev1Δwzm a highly promising vaccine candidate.
Collapse
Affiliation(s)
- Sara Mena-Bueno
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
- Agronomy, Biotecnology and Food Department, Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Irati Poveda-Urkixo
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
| | - Oihane Irazoki
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Leyre Palacios
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
| | - Felipe Cava
- Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden
| | - Ana Zabalza-Baranguá
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
| | - María Jesús Grilló
- Animal Health Department, Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Pamplona, Spain
- *Correspondence: María Jesús Grilló,
| |
Collapse
|
30
|
Celastrol mitigates staphyloxanthin biosynthesis and biofilm formation in Staphylococcus aureus via targeting key regulators of virulence; in vitro and in vivo approach. BMC Microbiol 2022; 22:106. [PMID: 35421933 PMCID: PMC9011992 DOI: 10.1186/s12866-022-02515-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/01/2022] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
Staphylococcus aureus is a leading cause of human infections. The spread of antibiotic-resistant staphylococci has driven the search for novel strategies to supersede antibiotics use. Thus, targeting bacterial virulence rather than viability could be a possible alternative.
Results
The influence of celastrol on staphyloxanthin (STX) biosynthesis, biofilm formation, antibiotic susceptibility and host pathogenesis in S. aureus has been investigated. Celastrol efficiently reduced STX biosynthesis in S. aureus. Liquid chromatography-mass spectrometry (LC–MS) and molecular docking revealed that celastrol inhibits STX biosynthesis through its effect on CrtM. Quantitative measurement of STX intermediates showed a significant pigment inhibition via interference of celastrol with CrtM and accumulation of its substrate, farnesyl diphosphate. Importantly, celastrol-treated S. aureus was more sensitive to environmental stresses and human blood killing than untreated bacteria. Similarly, inhibition of STX upon celastrol treatment rendered S. aureus more susceptible to membrane targeting antibiotics. In addition to its anti-pigment capability, celastrol exhibits significant anti-biofilm activity against S. aureus as indicated by crystal violet assay and microscopy. Celastrol-treated cells showed deficient exopolysaccharide production and cell hydrophobicity. Moreover, celastrol markedly synergized the action of conventional antibiotics against S. aureus and reduced bacterial pathogenesis in vivo using mice infection model. These findings were further validated using qRT-PCR, demonstrating that celastrol could alter the expression of STX biosynthesis genes as well as biofilm formation related genes and bacterial virulence.
Conclusions
Celastrol is a novel anti-virulent agent against S. aureus suggesting, a prospective therapeutic role for celastrol as a multi-targeted anti-pathogenic agent.
Collapse
|
31
|
Tumor Necrosis Factor Alpha Contributes to Inflammatory Pathology in the Placenta during Brucella abortus Infection. Infect Immun 2022; 90:e0001322. [PMID: 35100011 PMCID: PMC8929372 DOI: 10.1128/iai.00013-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Research on Brucella pathogenesis has focused primarily on its ability to cause persistent intracellular infection of the mononuclear phagocyte system. At these sites, Brucella abortus evades innate immunity, which results in low-level inflammation and chronic infection of phagocytes. In contrast, the host response in the placenta during infection is characterized by severe inflammation and extensive extracellular replication of B. abortus. Despite the importance of reproductive disease caused by Brucella infection, our knowledge of the mechanisms involved in placental inflammation and abortion is limited. To understand the immune responses specifically driving placental pathology, we modeled placental B. abortus infection in pregnant mice. B. abortus infection caused an increase in the production of tumor necrosis factor alpha (TNF-α), specifically in the placenta. We found that placental expression levels of Tnfa and circulating TNF-α were dependent on the induction of endoplasmic reticulum stress and the B. abortus type IV secretion system (T4SS) effector protein VceC. Blockade of TNF-α reduced placental inflammation and improved fetal viability in mice. This work sheds light on a tissue-specific response of the placenta to B. abortus infection that may be important for bacterial transmission via abortion in the natural host species.
Collapse
|
32
|
Muñoz PM, Conde-Álvarez R, Andrés-Barranco S, de Miguel MJ, Zúñiga-Ripa A, Aragón-Aranda B, Salvador-Bescós M, Martínez-Gómez E, Iriarte M, Barberán M, Vizcaíno N, Moriyón I, Blasco JM. A Brucella melitensis H38ΔwbkF rough mutant protects against Brucella ovis in rams. Vet Res 2022; 53:16. [PMID: 35236406 PMCID: PMC8889640 DOI: 10.1186/s13567-022-01034-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/20/2022] [Indexed: 01/05/2023] Open
Abstract
Brucella melitensis and Brucella ovis are gram-negative pathogens of sheep that cause severe economic losses and, although B. ovis is non-zoonotic, B. melitensis is the main cause of human brucellosis. B. melitensis carries a smooth (S) lipopolysaccharide (LPS) with an N-formyl-perosamine O-polysaccharide (O-PS) that is absent in the rough LPS of B. ovis. Their control and eradication require vaccination, but B. melitensis Rev 1, the only vaccine available, triggers anti-O-PS antibodies that interfere in the S-brucellae serodiagnosis. Since eradication and serological surveillance of the zoonotic species are priorities, Rev 1 is banned once B. melitensis is eradicated or where it never existed, hampering B. ovis control and eradication. To develop a B. ovis specific vaccine, we investigated three Brucella live vaccine candidates lacking N-formyl-perosamine O-PS: Bov::CAΔwadB (CO2-independent B. ovis with truncated LPS core oligosaccharide); Rev1::wbdRΔwbkC (carrying N-acetylated O-PS); and H38ΔwbkF (B. melitensis rough mutant with intact LPS core). After confirming their attenuation and protection against B. ovis in mice, were tested in rams for efficacy. H38ΔwbkF yielded similar protection to Rev 1 against B. ovis but Bov::CAΔwadB and Rev1::wbdRΔwbkC conferred no or poor protection, respectively. All H38ΔwbkF vaccinated rams developed a protracted antibody response in ELISA and immunoprecipitation B. ovis diagnostic tests. In contrast, all remained negative in Rose Bengal and complement fixation tests used routinely for B. melitensis diagnosis, though some became positive in S-LPS ELISA owing to LPS core epitope reactivity. Thus, H38ΔwbkF is an interesting candidate for the immunoprophylaxis of B. ovis in B. melitensis-free areas.
Collapse
Affiliation(s)
- Pilar M Muñoz
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain.
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain.
| | - Raquel Conde-Álvarez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Sara Andrés-Barranco
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain
| | - María-Jesús de Miguel
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain
| | - Amaia Zúñiga-Ripa
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Beatriz Aragón-Aranda
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Miriam Salvador-Bescós
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - Estrella Martínez-Gómez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
- Otology and Neurotology Group CTS495, Department of Genomic Medicine, GENYO Centre for Genomics and Oncological Research, Pfizer-University of Granada-Junta de Andalucía, Granada, Spain
| | - Maite Iriarte
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | | | - Nieves Vizcaíno
- Departamento de Microbiología y Genética, Universidad de Salamanca, Salamanca, Spain
| | - Ignacio Moriyón
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra and Departamento de Microbiología y Parasitología, Universidad de Navarra, Pamplona, Spain
| | - José M Blasco
- Departamento de Ciencia Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain
- Instituto Agroalimentario de Aragón-IA2 (CITA-Universidad de Zaragoza), Zaragoza, Spain
| |
Collapse
|
33
|
Sancho E, Granados-Chinchilla F, Barquero-Calvo E. Determination of streptomycin and doxycycline using LC/MS towards an effective treatment against an experimental Brucella abortus infection in mice. J Microbiol Methods 2022; 194:106436. [DOI: 10.1016/j.mimet.2022.106436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/27/2022]
|
34
|
Poveda-Urkixo I, Ramírez GA, Grilló MJ. Kinetics of Placental Infection by Different Smooth Brucella Strains in Mice. Pathogens 2022; 11:pathogens11030279. [PMID: 35335603 PMCID: PMC8955611 DOI: 10.3390/pathogens11030279] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 01/18/2023] Open
Abstract
Abortion and reproductive failures induced by Brucella are the main symptoms of animal brucellosis. Laboratory animal models are essential tools of research to study the Brucella pathogenesis before experimentation in natural hosts. To extend the existing knowledge, we studied B. melitensis 16M (virulent) and Rev1 (attenuated) as well as B. suis bv2 infections in pregnant mice. Here, we report new information about kinetics of infection (in spleens, blood, placentas, vaginal shedding, and foetuses), serum cytokine profiles, and histopathological features in placentas and the litter throughout mice pregnancy. Both B. melitensis strains showed a marked placental tropism and reduced viability of pups (mainly in 16M infections), which was preceded by an intense Th1-immune response during placental development. In contrast, B. suis bv2 displayed lower placental tropism, mild proinflammatory immune response, and scarce bacterial transmission to the litter, thus allowing foetal viability. Overall, our studies revealed three different smooth Brucella patterns of placental and foetal pathogenesis in mice, providing a useful animal model for experimental brucellosis.
Collapse
Affiliation(s)
- Irati Poveda-Urkixo
- Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Avda. Pamplona 123, 31192 Mutilva, Spain;
| | - Gustavo A. Ramírez
- Departamento de Sanidad Animal, Universidad de Lleida, 25198 Lleida, Spain;
| | - María-Jesús Grilló
- Instituto de Agrobiotecnología (IdAB, CSIC-Gobierno de Navarra), Avda. Pamplona 123, 31192 Mutilva, Spain;
- Correspondence:
| |
Collapse
|
35
|
Arias-Gómez B, Fonseca-Muñoz R, Alfaro-Alarcón A, Chacón-Díaz C, Moreno E, Rucavado A, Barquero-Calvo E. Platelet depletion does not alter the course of Brucella abortus infection in vivo. Microb Pathog 2022; 164:105458. [DOI: 10.1016/j.micpath.2022.105458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 01/18/2023]
|
36
|
Al-Mariri A, Al-Hallab L, Alabras R, Kherbik H, Khawajkiah M. Protection against virulent Brucella spp. by gamma-irradiated B. ovis in BALB/c mice model. Clin Exp Vaccine Res 2022; 11:53-62. [PMID: 35223665 PMCID: PMC8844668 DOI: 10.7774/cevr.2022.11.1.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/20/2021] [Indexed: 01/18/2023] Open
Abstract
Purpose Brucella spp. is a zoonosis that causes undulant fever in humans and abortion in livestock worldwide. Lately, it was conveyed that vaccines developed by irradiation have induced a strong cellular and humoral immune response which have made these types of vaccines highly effective. Materials and Methods In this study, we aimed to use the gamma-irradiated B. ovis as a vaccine and to study the humoral immune response and cytokines production in order to evaluate it for protecting mice against B. abortus 544, B. melitensis 16M, and B. ovis. Results The humoral immune response in immunized mice with gamma-irradiated B. ovis showed a lasting for 8 weeks after immunization. Moreover, immunoglobulin G (IgG), IgG1, IgG2a, and IgG2b isotypes antibodies against B. ovis were observed after 4 and 8 weeks of the last immunization. It was noticed that the production of tumor necrosis factor-α, interferon-γ, and interleukin (IL)-10 continued after 4 and 8 weeks by splenocytes from immunized BALB/c mice, while no production of IL-4 or IL-5 was observed. Conclusion Our results indicate that the protection of BALB/c mice against B. melitensis 16M, B. abortus 544, and B. ovis was induced and the developed vaccine at our laboratory could stimulate similar protection to those induced by the traditional vaccine.
Collapse
Affiliation(s)
- Ayman Al-Mariri
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria, Damascus, Syria
| | - Laila Al-Hallab
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria, Damascus, Syria
| | - Rasha Alabras
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria, Damascus, Syria
| | - Heba Kherbik
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria, Damascus, Syria
| | - Marwa Khawajkiah
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria, Damascus, Syria
| |
Collapse
|
37
|
Chacón-Díaz C, Zabalza-Baranguá A, San Román B, Blasco JM, Iriarte M, Salas-Alfaro D, Hernández-Mora G, Barquero-Calvo E, Guzmán-Verri C, Chaves-Olarte E, Grilló MJ, Moreno E. Brucella abortus S19 GFP-tagged vaccine allows the serological identification of vaccinated cattle. PLoS One 2021; 16:e0260288. [PMID: 34807952 PMCID: PMC8608319 DOI: 10.1371/journal.pone.0260288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/05/2021] [Indexed: 11/18/2022] Open
Abstract
Bovine brucellosis induces abortion in cows, produces important economic losses, and causes a widely distributed zoonosis. Its eradication was achieved in several countries after sustained vaccination with the live attenuated Brucella abortus S19 vaccine, in combination with the slaughtering of serologically positive animals. S19 induces antibodies against the smooth lipopolysaccharide (S-LPS), making difficult the differentiation of infected from vaccinated bovines. We developed an S19 strain constitutively expressing the green fluorescent protein (S19-GFP) coded in chromosome II. The S19-GFP displays similar biological characteristics and immunogenic and protective efficacies in mice to the parental S19 strain. S19-GFP can be distinguished from S19 and B. abortus field strains by fluorescence and multiplex PCR. Twenty-five heifers were vaccinated withS19-GFP (5×109 CFU) by the subcutaneous or conjunctival routes and some boosted with GFP seven weeks thereafter. Immunized animals were followed up for over three years and tested for anti-S-LPS antibodies by both the Rose Bengal test and a competitive ELISA. Anti-GFP antibodies were detected by an indirect ELISA and Western blotting. In most cases, anti-S-LPS antibodies preceded for several weeks those against GFP. The anti-GFP antibody response was higher in the GFP boosted than in the non-boosted animals. In all cases, the anti-GFP antibodies persisted longer, or at least as long, as those against S-LPS. The drawbacks and potential advantages of using the S19-GFP vaccine for identifying vaccinated animals in infected environments are discussed.
Collapse
Affiliation(s)
- Carlos Chacón-Díaz
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San Pedro, San José, Costa Rica
- * E-mail:
| | - Ana Zabalza-Baranguá
- Instituto de Agrobiotecnología, CSIC-Gobierno de Navarra, Mutilva, Navarra, Spain
| | - Beatriz San Román
- Instituto de Agrobiotecnología, CSIC-Gobierno de Navarra, Mutilva, Navarra, Spain
| | - José-María Blasco
- Unidad de Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria (CITA), Gobierno de Aragón, Aragón, Zaragoza, Spain
| | - Maite Iriarte
- Departamento de Microbiología y Parasitología, Instituto de Salud Tropical, Universidad de Navarra, Pamplona, Navarra, Spain
| | - Dariana Salas-Alfaro
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San Pedro, San José, Costa Rica
| | - Gabriela Hernández-Mora
- Servicio Nacional de Salud Animal, Ministerio de Agricultura y Ganadería, Lagunilla, Heredia, Costa Rica
| | - Elías Barquero-Calvo
- Programa de Investigación en Enfermedades Tropicales (PIET), Escuela de Medicina Veterinaria, Universidad Nacional, Lagunilla, Heredia, Costa Rica
| | - Caterina Guzmán-Verri
- Programa de Investigación en Enfermedades Tropicales (PIET), Escuela de Medicina Veterinaria, Universidad Nacional, Lagunilla, Heredia, Costa Rica
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, San Pedro, San José, Costa Rica
| | - María-Jesús Grilló
- Instituto de Agrobiotecnología, CSIC-Gobierno de Navarra, Mutilva, Navarra, Spain
| | - Edgardo Moreno
- Programa de Investigación en Enfermedades Tropicales (PIET), Escuela de Medicina Veterinaria, Universidad Nacional, Lagunilla, Heredia, Costa Rica
| |
Collapse
|
38
|
Aragón-Aranda B, Palacios-Chaves L, Salvador-Bescós M, de Miguel MJ, Muñoz PM, Vences-Guzmán MÁ, Zúñiga-Ripa A, Lázaro-Antón L, Sohlenkamp C, Moriyón I, Iriarte M, Conde-Álvarez R. The Phospholipid N-Methyltransferase and Phosphatidylcholine Synthase Pathways and the ChoXWV Choline Uptake System Involved in Phosphatidylcholine Synthesis Are Widely Conserved in Most, but Not All Brucella Species. Front Microbiol 2021; 12:614243. [PMID: 34421831 PMCID: PMC8371380 DOI: 10.3389/fmicb.2021.614243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/09/2021] [Indexed: 11/13/2022] Open
Abstract
The brucellae are facultative intracellular bacteria with a cell envelope rich in phosphatidylcholine (PC). PC is abundant in eukaryotes but rare in prokaryotes, and it has been proposed that Brucella uses PC to mimic eukaryotic-like features and avoid innate immune responses in the host. Two PC synthesis pathways are known in prokaryotes: the PmtA-catalyzed trimethylation of phosphatidylethanolamine and the direct linkage of choline to CDP-diacylglycerol catalyzed by the PC synthase Pcs. Previous studies have reported that B. abortus and B. melitensis possess non-functional PmtAs and that PC is synthesized exclusively via Pcs in these strains. A putative choline transporter ChoXWV has also been linked to PC synthesis in B. abortus. Here, we report that Pcs and Pmt pathways are active in B. suis biovar 2 and that a bioinformatics analysis of Brucella genomes suggests that PmtA is only inactivated in B. abortus and B. melitensis strains. We also show that ChoXWV is active in B. suis biovar 2 and conserved in all brucellae except B. canis and B. inopinata. Unexpectedly, the experimentally verified ChoXWV dysfunction in B. canis did not abrogate PC synthesis in a PmtA-deficient mutant, which suggests the presence of an unknown mechanism for obtaining choline for the Pcs pathway in Brucella. We also found that ChoXWV dysfunction did not cause attenuation in B. suis biovar 2. The results of these studies are discussed with respect to the proposed role of PC in Brucella virulence and how differential use of the Pmt and Pcs pathways may influence the interactions of these bacteria with their mammalian hosts.
Collapse
Affiliation(s)
- Beatriz Aragón-Aranda
- Dpto. de Microbiología y Parasitología, Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Leyre Palacios-Chaves
- Dpto. de Microbiología y Parasitología, Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Miriam Salvador-Bescós
- Dpto. de Microbiología y Parasitología, Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain
| | - María Jesús de Miguel
- Unidad de Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón, Zaragoza, Spain.,Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Pilar M Muñoz
- Unidad de Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón, Zaragoza, Spain.,Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | | | - Amaia Zúñiga-Ripa
- Dpto. de Microbiología y Parasitología, Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Leticia Lázaro-Antón
- Dpto. de Microbiología y Parasitología, Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Christian Sohlenkamp
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Ignacio Moriyón
- Dpto. de Microbiología y Parasitología, Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Maite Iriarte
- Dpto. de Microbiología y Parasitología, Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain
| | - Raquel Conde-Álvarez
- Dpto. de Microbiología y Parasitología, Instituto de Salud Tropical (ISTUN), Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
39
|
Stranahan LW, Arenas-Gamboa AM. When the Going Gets Rough: The Significance of Brucella Lipopolysaccharide Phenotype in Host-Pathogen Interactions. Front Microbiol 2021; 12:713157. [PMID: 34335551 PMCID: PMC8319746 DOI: 10.3389/fmicb.2021.713157] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/22/2021] [Indexed: 01/18/2023] Open
Abstract
Brucella is a facultatively intracellular bacterial pathogen and the cause of worldwide zoonotic infections, infamous for its ability to evade the immune system and persist chronically within host cells. Despite the frequent association with attenuation in other Gram-negative bacteria, a rough lipopolysaccharide phenotype is retained by Brucella canis and Brucella ovis, which remain fully virulent in their natural canine and ovine hosts, respectively. While these natural rough strains lack the O-polysaccharide they, like their smooth counterparts, are able to evade and manipulate the host immune system by exhibiting low endotoxic activity, resisting destruction by complement and antimicrobial peptides, entering and trafficking within host cells along a similar pathway, and interfering with MHC-II antigen presentation. B. canis and B. ovis appear to have compensated for their roughness by alterations to their outer membrane, especially in regards to outer membrane proteins. B. canis, in particular, also shows evidence of being less proinflammatory in vivo, suggesting that the rough phenotype may be associated with an enhanced level of stealth that could allow these pathogens to persist for longer periods of time undetected. Nevertheless, much additional work is required to understand the correlates of immune protection against the natural rough Brucella spp., a critical step toward development of much-needed vaccines. This review will highlight the significance of rough lipopolysaccharide in the context of both natural disease and host–pathogen interactions with an emphasis on natural rough Brucella spp. and the implications for vaccine development.
Collapse
Affiliation(s)
- Lauren W Stranahan
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Angela M Arenas-Gamboa
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
40
|
Khatun MM, Islam MA, Baek BK. In Vitro and In Vivo IFN-γ and IL-10 Measurement in Experimental Brucella abortus Biotype 1 Infection in Sprague-Dawley Rats. Vector Borne Zoonotic Dis 2021; 21:579-585. [PMID: 34077683 DOI: 10.1089/vbz.2020.2738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The immune response to Brucella abortus mainly depends on antigen-specific T cell activation, CD4+ and CD8+ T cells, and Brucella-specific humoral response. Protective immune response against Brucella infection has not been performed in the Sprague-Dawley (SD) rat model. We measured bacterial kinetics in addition to in vivo and in vitro interferon gamma (IFN-γ) and interleukin-10 (IL-10) production against crude Brucella protein in the SD rats at different days of postinfection with B. abortus biotype 1 by indirect enzyme-linked immunosorbent assay. Forty SD rats were inoculated intraperitoneally with 0.1 mL sterile injectable pyrogen-free solution containing 1 × 1010 colony-forming units/mL of B. abortus biotype 1 obtained from cattle in Korea. Four rats were used as uninfected control. Serum IFN-γ level at 3 and 7 days postinfection were significantly higher (p > 0.001) compared with the IL-10 level. On the contrary, serum IL-10 levels were observed significantly higher at 21 and 28 days postinfection compared with the serum IFN-γ levels (p < 0.001). The production of IFN-γ by spleen cells was significantly higher at 7 and 14 days postinfection compared with IL-10 (p < 0.001). On the contrary, IL-10 productions were found to be significantly higher at 21, 28, 35, and 42 days postinfection compared with IFN-γ (p < 0.001). The presence of B. abortus in blood was marked till 5 weeks of infection, throughout the experiment in case of spleen, and no bacteria were isolated from the kidney and liver at 6 weeks postinfection. The in vivo and in vitro IFN-γ and IL-10 measurement in our study reported that B. abortus infection in rats primarily educe T helper (Th)1-dominant immune response in acute infection accompanied by Th2-dominant immune response in chronic infection.
Collapse
Affiliation(s)
- Mst Minara Khatun
- Department of Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Jeonju, Republic of Korea.,Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md Ariful Islam
- Department of Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Jeonju, Republic of Korea.,Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Byeong Kirl Baek
- Department of Veterinary Public Health, College of Veterinary Medicine, Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
41
|
Priyanka, Shringi BN, Choudhary OP, Kashyap SK. Expression profiling of cytokine-related genes in Brucella abortus infected cattle. BIOL RHYTHM RES 2021. [DOI: 10.1080/09291016.2019.1600263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Priyanka
- Department of Veterinary Microbiology and Biotechnology, College of Veterinary and Animal Sciences, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Brij Nandan Shringi
- Department of Veterinary Microbiology and Biotechnology, College of Veterinary and Animal Sciences, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy and Histology, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, India
| | - Sudhir Kumar Kashyap
- Department of Veterinary Microbiology and Biotechnology, College of Veterinary and Animal Sciences, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| |
Collapse
|
42
|
VU SH, KIM B, REYES AWB, HUY TXN, LEE JH, KIM S, KIM HJ. Global metabolomic analysis of blood from mice infected with Brucella abortus. J Vet Med Sci 2021; 83:482-486. [PMID: 33473061 PMCID: PMC8025415 DOI: 10.1292/jvms.20-0630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/06/2021] [Indexed: 11/22/2022] Open
Abstract
To better understanding Brucella abortus infection, serum metabolites of B. abortus-infected and -uninfected mice were analyzed and twenty-one metabolites were tentatively identified at 3 and 14 days post-infection (d.p.i.). Level of most lysophosphatidylcholines (LPCs) was found to increase in infected mice at 3 d.p.i., while it was decreased at 14 d.p.i. as compared to uninfected mice. In contrast, acylcarnitines were initially reduced at 3 d.p.i then elevated after two-weeks of infection, while hydroxysanthine was increased at 14 d.p.i. in infected mice. Our findings suggest that the significant changes in LPCs and other identified metabolites may serve as potential biomarkers in acute phase of B. abortus infection.
Collapse
Affiliation(s)
- Son Hai VU
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of
Korea
- Institute of Applied Sciences, Ho Chi Minh City University of Technology–HUTECH, 475A Dien Bien Phu St., Ward 25, Binh Thanh
District, Ho Chi Minh City, Vietnam
| | - Bomin KIM
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of
Korea
| | | | - Tran Xuan Ngoc HUY
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of
Korea
| | - John Hwa LEE
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Suk KIM
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of
Korea
| | - Hyun-Jin KIM
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of
Korea
| |
Collapse
|
43
|
Quantification of Brucella abortus population structure in a natural host. Proc Natl Acad Sci U S A 2021; 118:2023500118. [PMID: 33688053 DOI: 10.1073/pnas.2023500118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cattle are natural hosts of the intracellular pathogen Brucella abortus, which inflicts a significant burden on the health and reproduction of these important livestock. The primary routes of infection in field settings have been described, but it is not known how the bovine host shapes the structure of B. abortus populations during infection. We utilized a library of uniquely barcoded B. abortus strains to temporally and spatially quantify population structure during colonization of cattle through a natural route of infection. Introducing 108 bacteria from this barcoded library to the conjunctival mucosa resulted in expected levels of local lymph node colonization at a 1-wk time point. We leveraged variance in strain abundance in the library to demonstrate that only 1 in 10,000 brucellae introduced at the site of infection reached a parotid lymph node. Thus, cattle restrict the overwhelming majority of B. abortus introduced via the ocular conjunctiva at this dose. Individual strains were spatially restricted within the host tissue, and the total B. abortus census was dominated by a small number of distinct strains in each lymph node. These results define a bottleneck that B. abortus must traverse to colonize local lymph nodes from the conjunctival mucosa. The data further support a model in which a small number of spatially isolated granulomas founded by unique strains are present at 1 wk postinfection. These experiments demonstrate the power of barcoded transposon tools to quantify infection bottlenecks and to define pathogen population structure in host tissues.
Collapse
|
44
|
Reyes AWB, Huy TXN, Vu SH, Kang CK, Min W, Lee HJ, Lee JH, Kim S. Formyl peptide receptor 2 (FPR2) antagonism is a potential target for the prevention of Brucella abortus 544 infection. Immunobiology 2021; 226:152073. [PMID: 33657463 DOI: 10.1016/j.imbio.2021.152073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 10/22/2022]
Abstract
Here, we explore the potential role of formyl peptide receptor 2 (FPR2) during Brucella abortus infection. FPR2 manipulation affected B. abortus internalization but not its growth within macrophages. During the activation of FPR2 induced by its agonist AGP-8694, a high level of Brucella uptake was accompanied by an increase in ERK phosphorylation, while intracellular survival at 24 h postincubation was observed to be associated with slightly reduced nitrite accumulation but augmented superoxide anion production. Attenuated secretion of IL-6 and IL-10 were observed 48 h postincubation in the bone marrow-derived macrophages (BMDMs) treated with the FPR2 antagonist WRW4. An opposite pattern of bacterial uptake was observed upon treatment with the FPR2 antagonist, but no significant changes in the activation of MAPKs or the production of nitrite or superoxide anion were observed. Interestingly, AGP-8694 treatment of mice did not lead to differences in spleen or liver weight but slightly enhanced bacterial proliferation was observed in the spleen. Although the weights of the spleen or liver did not differ, WRW4 treatment led to reduced bacterial proliferation in the spleen. Furthermore, FPR2 antagonist treatment was associated with high serum levels of the proinflammatory cytokines IL-12, TNF-α, IFN-γ and MCP-1, while the production of TNF-α was inhibited in AGP-8694-treated mice. IL-6 and IL-10 levels were slightly increased in AGP-8694-treated mice at 24 h postinfection. Our findings demonstrated the contribution of FPR2 via manipulating this receptor using its reported agonist AGP-8694 and antagonist WRW4 in both in vitro and in vivo systems. Although activation of the receptor did not consistently induced Brucella infection, FPR2 inhibition may be a promising strategy to treat brucellosis in animals which encourages further investigation.
Collapse
Affiliation(s)
- Alisha Wehdnesday Bernard Reyes
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Tran Xuan Ngoc Huy
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; Institute of Applied Sciences, Ho Chi Minh City University of Technology - HUTECH, 475A Dien Bien Phu St., Ward 25, Binh Thanh District, Ho Chi Minh City, Viet Nam
| | - Son Hai Vu
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; Institute of Applied Sciences, Ho Chi Minh City University of Technology - HUTECH, 475A Dien Bien Phu St., Ward 25, Binh Thanh District, Ho Chi Minh City, Viet Nam
| | - Chang Keun Kang
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Wongi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hu Jang Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Suk Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea.
| |
Collapse
|
45
|
Development of Brucella melitensis Rev.1 ΔOmp19 mutants with DIVA feature and comparison of their efficacy against three commercial vaccines in a mouse model. Mol Immunol 2021; 133:44-52. [PMID: 33631554 DOI: 10.1016/j.molimm.2021.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/30/2020] [Accepted: 02/07/2021] [Indexed: 01/18/2023]
Abstract
Brucella is an intracellular zoonotic pathogen that can affect many hosts. Brucella melitensis Rev.1 is a live attenuated, is one of the most effective vaccine strain against brucellosis. It can be used safely in sheep, goats, and even cattle. Although many studies are available on this topic, there is no effective vaccine strain for sheep and goats that distinguishes the antibody titer produced between the field infections and vaccinations. Outer membrane protein 19 (Omp 19) is both virulent and a protective antigen found on the cell-wall of the Brucella strain. In this study, used the suicide plasmid pJQ200KS, which contained homologous region without Omp19 Open Reading Frame (ORF) that was transferred to B. melitensis Rev.1 and further transformed into spheroplasts along with penicillin, ampicillin, and glycine by electroporation. To obtain a mutant vector from Escherichia coli, we used the heat shock transformation method along with the blue-white colony screening using X-gal media, whereas for the gene transfer in Brucella, we used electroporation. A scanning electron microscope (S.E.M) was used to observe the spheroplast transformation while the mutant vector and deletion mutants were confirmed through PCR and sequence analysis. In the mouse model efficacy trials, three commercial vaccines were found to comply with the OIE standards. Although the deletion mutants 19 and 44/10 had similar efficiency as the commercial vaccines in terms of stimulation power, the ELISA test with Omp19 protein showed the same results as the negative control. The Rev.1 Omp19 deletion mutants obtained in this study contained sufficient residual virulence, and their protective immunity was similar to the commercial vaccines. The study showed that a vaccine prepared using a B. melitensis Rev.1 ΔOmp19 can act as a marker vaccine or differentiate infected from vaccinated animals (DIVA) through the ELISA test that detects the Omp19 protein.
Collapse
|
46
|
Roop RM, Barton IS, Hopersberger D, Martin DW. Uncovering the Hidden Credentials of Brucella Virulence. Microbiol Mol Biol Rev 2021; 85:e00021-19. [PMID: 33568459 PMCID: PMC8549849 DOI: 10.1128/mmbr.00021-19] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Bacteria in the genus Brucella are important human and veterinary pathogens. The abortion and infertility they cause in food animals produce economic hardships in areas where the disease has not been controlled, and human brucellosis is one of the world's most common zoonoses. Brucella strains have also been isolated from wildlife, but we know much less about the pathobiology and epidemiology of these infections than we do about brucellosis in domestic animals. The brucellae maintain predominantly an intracellular lifestyle in their mammalian hosts, and their ability to subvert the host immune response and survive and replicate in macrophages and placental trophoblasts underlies their success as pathogens. We are just beginning to understand how these bacteria evolved from a progenitor alphaproteobacterium with an environmental niche and diverged to become highly host-adapted and host-specific pathogens. Two important virulence determinants played critical roles in this evolution: (i) a type IV secretion system that secretes effector molecules into the host cell cytoplasm that direct the intracellular trafficking of the brucellae and modulate host immune responses and (ii) a lipopolysaccharide moiety which poorly stimulates host inflammatory responses. This review highlights what we presently know about how these and other virulence determinants contribute to Brucella pathogenesis. Gaining a better understanding of how the brucellae produce disease will provide us with information that can be used to design better strategies for preventing brucellosis in animals and for preventing and treating this disease in humans.
Collapse
Affiliation(s)
- R Martin Roop
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Ian S Barton
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Dariel Hopersberger
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Daniel W Martin
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
47
|
Zhu X, Zhao Z, Ma S, Guo Z, Wang M, Li Z, Liu Z. Brucella melitensis, a latent "travel bacterium," continual spread and expansion from Northern to Southern China and its relationship to worldwide lineages. Emerg Microbes Infect 2021; 9:1618-1627. [PMID: 32594852 PMCID: PMC7473006 DOI: 10.1080/22221751.2020.1788995] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Brucellosis caused by Brucella melitensis is considered to
be one of the most important zoonotic diseases in China. In this study, Conventional
bio-typing, MLVA (multiple locus variable-number tandem repeat analysis), and WGS
(whole-genome sequencing)-SNP (single nucleotide polymorphism) were used to study the
genetic similarity of B. melitensis in northern and southern
China and analyze its relationship with worldwide lineages. Currently, the distribution of
species/biovars of B. melitensis has obviously changed, and
B. melitensis has become the dominant species in southern
regions of China. Strains from the southern had a common geographic origin with strains
from the northern. Many MLVA-16 events were shared in the genotypes of the southern and
northern strains, suggest that genotypic movement occurred from north to south. Based on
WGS-SNP analysis, strains from different provinces were closely related and may have
descended from one common ancestor, suggests that the southern strains originated from
northern China. These data indicate that B. melitensis is a
latent “travel bacterium” that spread and expanded from North China to South China.
Moreover, B. melitensis strains from China are also
genetically related to strains from other Asian regions (Kazakhstan, Russia, Mongolia, and
India). The movement of infected sheep and their products requires control.
Collapse
Affiliation(s)
- Xiong Zhu
- Sanya People's Hospital, Sanya, People's Republic of China
| | - Zhongzhi Zhao
- School of Medical Technology, Baotou Medical College, Baotou, People's Republic of China
| | - Shuyi Ma
- Qinghai Institute for Endemic Diseases Prevention and Control, Xining, People's Republic of China
| | - Zhiwei Guo
- Inner Mongolia Autonomous Region Center for Comprehensive Disease Control and Prevention, Huhhot, People's Republic of China
| | - Miao Wang
- Ulanqab Centre for Endemic Disease Prevention and Control, Jining, People's Republic of China
| | - Zhenjun Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Zhiguo Liu
- Sanya People's Hospital, Sanya, People's Republic of China.,State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| |
Collapse
|
48
|
Lázaro-Antón L, de Miguel MJ, Barbier T, Conde-Álvarez R, Muñoz PM, Letesson JJ, Iriarte M, Moriyón I, Zúñiga-Ripa A. Glucose Oxidation to Pyruvate Is Not Essential for Brucella suis Biovar 5 Virulence in the Mouse Model. Front Microbiol 2021; 11:620049. [PMID: 33519781 PMCID: PMC7840955 DOI: 10.3389/fmicb.2020.620049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/02/2020] [Indexed: 12/31/2022] Open
Abstract
Brucella species cause brucellosis, a worldwide extended zoonosis. The brucellae are related to free-living and plant-associated α2-Proteobacteria and, since they multiply within host cells, their metabolism probably reflects this adaptation. To investigate this, we used the rodent-associated Brucella suis biovar 5, which in contrast to the ruminant-associated Brucella abortus and Brucella melitensis and other B. suis biovars, is fast-growing and conserves the ancestral Entner-Doudoroff pathway (EDP) present in the plant-associated relatives. We constructed mutants in Edd (glucose-6-phosphate dehydratase; first EDP step), PpdK (pyruvate phosphate dikinase; phosphoenolpyruvate ⇌ pyruvate), and Pyk (pyruvate kinase; phosphoenolpyruvate → pyruvate). In a chemically defined medium with glucose as the only C source, the Edd mutant showed reduced growth rates and the triple Edd-PpdK-Pyk mutant did not grow. Moreover, the triple mutant was also unable to grow on ribose or xylose. Therefore, B. suis biovar 5 sugar catabolism proceeds through both the Pentose Phosphate shunt and EDP, and EDP absence and exclusive use of the shunt could explain at least in part the comparatively reduced growth rates of B. melitensis and B. abortus. The triple Edd-PpdK-Pyk mutant was not attenuated in mice. Thus, although an anabolic use is likely, this suggests that hexose/pentose catabolism to pyruvate is not essential for B. suis biovar 5 multiplication within host cells, a hypothesis consistent with the lack of classical glycolysis in all Brucella species and of EDP in B. melitensis and B. abortus. These results and those of previous works suggest that within cells, the brucellae use mostly 3 and 4 C substrates fed into anaplerotic pathways and only a limited supply of 5 and 6 C sugars, thus favoring the EDP loss observed in some species.
Collapse
Affiliation(s)
- Leticia Lázaro-Antón
- Department of Microbiology and Parasitology, Facultad de Medicina, ISTUN Instituto de Salud Tropical, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - María Jesús de Miguel
- Unidad de Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain.,Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Thibault Barbier
- Research Unit in Biology of Microorganisms (URBM), NARILIS, University of Namur, Namur, Belgium
| | - Raquel Conde-Álvarez
- Department of Microbiology and Parasitology, Facultad de Medicina, ISTUN Instituto de Salud Tropical, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Pilar M Muñoz
- Unidad de Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria de Aragón (CITA), Zaragoza, Spain.,Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Jean Jacques Letesson
- Research Unit in Biology of Microorganisms (URBM), NARILIS, University of Namur, Namur, Belgium
| | - Maite Iriarte
- Department of Microbiology and Parasitology, Facultad de Medicina, ISTUN Instituto de Salud Tropical, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ignacio Moriyón
- Department of Microbiology and Parasitology, Facultad de Medicina, ISTUN Instituto de Salud Tropical, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Amaia Zúñiga-Ripa
- Department of Microbiology and Parasitology, Facultad de Medicina, ISTUN Instituto de Salud Tropical, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| |
Collapse
|
49
|
Vu SH, Bernardo Reyes AW, Ngoc Huy TX, Min W, Lee HJ, Kim HJ, Lee JH, Kim S. Prostaglandin I2 (PGI 2) inhibits Brucella abortus internalization in macrophages via PGI 2 receptor signaling, and its analogue affects immune response and disease outcome in mice. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 115:103902. [PMID: 33091457 DOI: 10.1016/j.dci.2020.103902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/29/2020] [Accepted: 10/15/2020] [Indexed: 06/11/2023]
Abstract
To date, the implications of prostaglandin I2 (PGI2), a prominent lipid mediator for modulation of immune responses, has not been clearly understood in Brucella infection. In this study, we found that cyclooxygenase-2 (COX-2) was significantly expressed in both infected bone marrow-derived macrophages (BMMs) and RAW 264.7 cells. Prostaglandin I2 synthase (PTGIS) expression was not significantly changed, and PGI2receptor (PTGIR) expression was downregulated in BMMs but upregulated in RAW 264.7 macrophages at late infection. Here, we presented that PGI2, a COX-derived metabolite, was produced by macrophages during Brucella infection and its production was regulated by COX-2 and IL-10. We suggested that PGI2 and selexipag, a potent PGI2 analogue, inhibited Brucella internalization through IP signaling which led to down-regulation of F-actin polymerization and p38α MAPK activity. Administration with selexipag suppressed immune responses and resulted in a notable reduction in bacterial burden in spleen of Brucella-challenged mice. Taken together, our study is the first to characterize PGI2 synthesis and its effect in evasion strategy of macrophages against Brucella infection.
Collapse
Affiliation(s)
- Son Hai Vu
- Institute of Applied Sciences, Ho Chi Minh City University of Technology - HUTECH, 475A Dien Bien Phu St., Ward 25, Binh Thanh District, Ho Chi Minh City, Viet Nam; Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | | | - Tran Xuan Ngoc Huy
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Wongi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hu Jang Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hyun-Jin Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Suk Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
50
|
Development of Human Vectored Brucellosis Vaccine Formulation: Assessment of Safety and Protectiveness of Influenza Viral Vectors Expressing Brucella Immunodominant Proteins in Mice and Guinea Pigs. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1438928. [PMID: 33274194 PMCID: PMC7695499 DOI: 10.1155/2020/1438928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/10/2020] [Accepted: 10/15/2020] [Indexed: 01/18/2023]
Abstract
In this paper, we first used recombinant influenza viral vector (rIVV) subtype H5N1 expressing from the open reading frame of NS1 80 and NS1 124 amino acids of Brucella outer membrane proteins (Omp) 16 and 19, ribosomal L7/L12, and Cu-Zn superoxide dismutase (SOD) proteins to develop a human brucellosis vaccine. We made 18 combinations of IVVs in mono-, bi-, and tetravalent vaccine formulations and tested them on mice to select the safest and most effective vaccine samples. Then, the most effective vaccine candidates were further tested on guinea pigs. Safety of the rIVV-based vaccine candidate was evaluated by a mouse weight-gain test. Mice and guinea pigs were challenged with the virulent strain B. melitensis 16M. The protective effect of the rIVV-based vaccine candidate was assessed by quantitation of Brucella colonization in tissues and organs of challenged animals. All vaccine formulations were safe in mice. Tested vaccine formulations, as well as the commercial B. melitensis Rev.1 vaccine, have been found to protect mice from B. melitensis 16M infection within the range of 1.6 to 2.97 log10 units (P < 0.05). Tetravalent vaccine formulations from the position of NS1 80 amino acids (0.2 ± 0.4), as well as the commercial B. melitensis Rev.1 vaccine (1.2 ± 2.6), have been found to protect guinea pigs from B. melitensis 16M infection at a significant level (P < 0.05). Thus, tetravalent vaccine formulation Flu-NS1-80-Omp16+Flu-NS1-80-L7/L12+Flu-NS1-80-Omp19+Flu-NS1-80-SOD was chosen as a potential vaccine candidate for further development of an effective human vaccine against brucellosis. These results show a promising future for the development of a safe human vaccine against brucellosis based on rIVVs.
Collapse
|