3,088
Views
14
CrossRef citations to date
0
Altmetric
Editorial - Commissioned

Brucella Lipopolysaccharide and pathogenicity: The core of the matter

Pages 379-382 | Received 13 Oct 2017, Accepted 17 Oct 2017, Published online: 01 Mar 2018
This article refers to:
Immunomodulatory properties of Brucella melitensis lipopolysaccharide determinants on mouse dendritic cells in vitro and in vivo

Brucellosis is one of the most prevalent zoonotic infections worldwide.Citation1 The half million reported new cases per year likely represents a gross underestimate related to challenges in diagnosis and incomplete reporting. In humans, brucellosis initially manifests as undulating fever accompanied by flu-like myalgias and arthralgias. However, chronic infection may lead to peripheral arthritis, sacroiliitis, orchitis, endocarditis and neurobrucellosis.Citation1,2 Treatment involves prolonged courses of multiple antibiotics and relapses occur in up to 10% of patients.Citation3 The causative agents, Brucella species, are Gram-negative facultative intracellular bacteria which infect herd animals such as goats, cattle and swine. In animals, Brucella species cause abortion, resulting in tremendous economic losses. Although animal vaccine strains such as Rev1, RB51 and S19 have been effective at controlling disease in the herds of nations with sufficient infrastructure and regulation, Brucella infections remain intractable in many parts of the world:Citation4 Brucellosis is endemic in Central and South America, Sub-Saharan Africa, the Middle East and Asia.Citation5 Brucella are primarily transmitted to human hosts by ingestion of contaminated unpasteurized dairy products, but Brucella also poses a risk to herders and abattoir workers. Because of the low number needed for infection (10-100 organisms), and ready aerosolization, Brucella is considered a bioterror threat.Citation6 Current vaccine strains cause disease in humans.Citation7–9 Thus at present, no safe and effective human vaccine exists.

With the goal of improving therapeutic and vaccine strategies, research has focused on understanding the pathogenic determinants that allow Brucella to establish successful chronic infections and evade immune eradication. Ultimately, immune control of Brucella involves the development of effective Th1 cellular immunity.Citation10,11 However, mobilization of Brucella-responsive T cells first requires recognition by “sentinel” dendritic cells of the innate immune system, and activation of these cells to become effective antigen presenting cells.Citation12 Innate immune cells detect pathogens via repeated cellular patterns (PAMPs) such as the Lipopolysaccharide (LPS) coat on Gram-negative bacteria. Intriguingly, one of the major Brucella virulence factors identified to date is its non-canonical LPS.Citation13

LPS plays a vital role in the integrity of Gram-negative bacterial outer membranes. It comprises three regions: a Lipid A moiety composed of a disaccharide backbone linked to up to seven hydrophobic acyl chains that are embedded in the outer membrane, a species-conserved core polysaccharide linker, and the external most O-polysaccharide chain.Citation14 The inner core contains unusual sugars (e.g. 3-deoxy-D-manno-octulosonic acid (Kdo)), but the outer core has more common sugars such as hexoses and hexosamines. The O-polysaccharide, a long chain of repeating glycosyl subunits, exhibits the most variability, providing the means for strain differentiation in labs and antigenic stimulation of humoral immune responses. Innate immune cells recognize LPS via a heteromeric receptor composed of Toll-like receptor 4 (TLR4) and its binding partner myeloid differentiation-2 (MD2). MD2 contains a large hydrophobic pocket that accompanies 5 of the Lipid A acyl chains, whereas the LPS polysaccharide forms polar interactions with the rim of MD2 and TLR4.Citation15,16 Agonist activity (endotoxicity) is generally thought to reflect the acyl chain number, length, and chemical modifications.Citation14

Brucella LPS counters innate immune defenses on multiple levels: the Lipid A moiety contains overly long C16-18 fatty acids (including up to C28), rather than the optimal 12–14 carbons, correlating with poor MD2 binding and low endotoxicity.Citation13,17–19 The Lipid A component also kills neutrophils through an unclear mechanism.Citation20 The O-polysaccharide resists complement deposition and activation.Citation21 Inside cells, this outer polysaccharide is also important for evasion of lysosomal destruction. Indeed “rough” strains or mutants of Brucella lacking the external O-polysaccharide display significantly attenuated virulence.Citation22,23 More recently the core moiety of Brucella has come to the fore as a key modulator of virulence.Citation19 The Brucella core polysaccharide contains 2 Kdo sugars; one connects to the O-polysaccharide, and the other to an unusual branching side chain.Citation24,25 This side chain, with its positive charges, is thought to “shield” the more internal negative charges from effective interaction with the MD2/TLR4 receptor.Citation19,24 Brucella mutants deficient in the wadC mannosyltransferase enzyme required for this core saccharide branch display enhanced MD2 binding, and increased triggering of cytokine production. Interestingly, the wadC mutants are also more susceptible to complement and antibacterial cationic peptides – thus not all the serum resistance reflects O-polysaccharide or Lipid A composition.Citation21,26 Perhaps because of this increased susceptibility, plus the increased immune activation, the wadC mutant Brucella are attenuated in vivo in mice.Citation19 Conversely, the immune evasion properties of wild type Brucella LPS contribute to the prevailing conception of Brucella as a “stealthy pathogen”.Citation27,28

Nevertheless, there are some challenges to the notion that Brucella completely evades TLR4 detection. Although results are conflicting, some studies have documented a role for TLR4 in Brucella resistance.Citation29–31 Adjuvant properties of Brucella LPS have been reported.Citation32,33 Most of the studies documenting poor TLR4 stimulation of dendritic cells by Brucella LPS have used GM-CSF derived dendritic cells in vitro. However, dendritic cell heterogeneity has been increasingly recognized. Just within the spleen, subtypes include plasmacytoid dendritic cells, CD11b+ and CD8a+ conventional dendritic cells, and monocyte-derived dendritic cells.Citation34 These subtypes vary in TLR expression, capacity for viral-induced IFN responses, T-cell costimulation and cross-presentation of external antigen to CD8 T cells. The roles of these different subsets in response to Brucella infection have not been clearly defined. The study by Zhao et al. entitled “Immunomodulatory properties of Brucella melitensis lipopolysaccharide determinants in mouse dendritic cells in vitro and in vivo” supports the importance of evaluating different dendritic subsets when assessing LPS stimulatory capacity.Citation35

In this study Zhao et al employed a “mix and match” approach, examining the effect of various purified LPS preparations sharing different features with Brucella LPS. For instance Ochrobactrum anthropi 3331 LPS contains a Lipid A moiety similar to Brucella, whereas Yersinia enterocolitica O:9 shares a similar O-chain polysaccharide.Citation36,37 E. coli LPS served as the gold standard TLR4 agonist. The ability of these LPS preps to activate different types of dendritic cells, as assessed by cytokine production and cell surface markers (B7 molecules, CD40, MHC class II, PDL-1) were compared in vitro and in vivo. Several notable findings challenge current paradigms:

1)

This study emphasized the importance of the Brucella core polysaccharide branch: surprisingly, Brucella-type lipid A and the O-polysaccharide were not in themselves problematic for TLR4 stimulation, as the O. anthropi, Y enterocolitica and the wadC Brucella mutant induced co-stimulator expression and cytokine production comparable to E. coli LPS in GM-CSF derived dendritic cells. Interestingly, only IL-10 production correlated with Lipid A structure. Thus a bulky Lipid A may be less of an issue for pro-inflammatory cytokine induction than previously thought. Indeed, despite the Brucella-type Lipid A, O. anthropi 3331 LPS induced greater production of IL-12p70, TNF-α, IL-6 and IL-1β than equimolar E. coli LPS.

2)

Brucella LPS is not completely inert: In Fms related tyrosine kinase 3 (Flt3) derived dendritic cells, Brucella LPS induces comparable TNF-α secretion and upregulation of MHC class II to other forms of LPS, and significant (though reduced) increases in co-stimulatory molecules. These findings challenge the notion that Brucella LPS is a universally poor TLR4 stimulus.

Their results also provide greater insight into how Brucella avoids potent activation of adaptive response: In vitro, Brucella LPS was deficient in stimulating Flt3 dendritic cell production of IL-12p70 and IFN-γ, 2 cytokines critical for the Th1 responses that control disease.Citation10,11 Brucella LPS also failed to activate Flt3 dendritic cells sufficiently to support CD4 or CD8 T cell proliferation in vitro. In vivo, unlike E. coli or the wadC mutant LPS, Brucella LPS induced very little splenic expansion of CD64+ DC-sign+ monocyte-derived dendritic cells. Interestingly, one of the few activation markers significantly up-regulated on CD11chi spleen cells was programmed death ligand (PDL-1), which would undermine adaptive responses by causing T cell exhaustion.Citation35 Consistent with this observation, CD8 T cell exhaustion has been reported in mouse models of brucellosis.Citation38,39

Together, the results reported by Zhou et al. raise compelling questions regarding the nature of TLR4/MD2 recognition of LPS. It is not clear how TLR4/MD2 distinguishes between the individual types of LPS to induce a different cytokine profile. Are there subtle differences in oligomerization or structural conformation that result in altered recruitment of cytosolic adaptors and other signaling molecules? The exact mechanism by which the Brucella core polysaccharide branch “shields” recognition, as well as how these different bacterial forms of LPS bind the TLR4 receptor complex may require crystallographic definition.

One of the more intriguing issues raised by this study is the potential to use the wadC Brucella mutant as a vaccine. The wadC mutant LPS induces similar mobilization of CD11b BST-2+ monocyte derived dendritic cells as E. coli LPS and comparable induction of co-stimulatory marker expression, but displays attenuation in vivo. These authors have provided initial data that the wadC mutant Brucella may be at least as protective as the S19 vaccine strain.Citation40 However development of this mutant for vaccine purposes will require greater analysis of the effect of the whole bacteria in vivo (not just the LPS) and further exploration of the organism's protective capacity. Safety, particularly as it affects reproduction, will also need to be established.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Additional information

Funding

Funding Source: NIH-NIAID ID: R01 AI116453 Funding Source: NIH-NIAID ID: R01 AI073558-08

References

  • Pappas G, Akritidis N, Bosilkovski M, Tsianos E. Brucellosis. N Engl J Med 2005;352:2325–36.
  • Dean AS, Crump L, Greter H, Hattendorf J, Schelling E, Zinsstag J. Clinical manifestations of human brucellosis: a systematic review and meta-analysis. PLoS Negl Trop Dis 2012;6:e1929.
  • Franco MP, Mulder M, Gilman RH, Smits HL. Human brucellosis. Lancet Infect Dis 2007;7:775–86.
  • Avila-Calderon ED, Lopez-Merino A, Sriranganathan N, Boyle SM, Contreras-Rodriguez A. A history of the development of Brucella vaccines. Biomed Res Int 2013;2013:743509.
  • Pappas G, Papadimitriou P, Akritidis N, Christou L, Tsianos EV. The new global map of human brucellosis. Lancet Infect Dis 2006;6:91–9.
  • Pappas G, Panagopoulou P, Christou L, Akritidis N. Brucella as a biological weapon. Cell Mol Life Sci 2006;63:2229–36.
  • Wallach JC, Ferrero MC, Victoria Delpino M, Fossati CA, Baldi PC. Occupational infection due to Brucella abortus S19 among workers involved in vaccine production in Argentina. Clin Microbiol Infect 2008;14:805–7.
  • Montes J, Rodriguez MA, Martin T, Martin F. Laboratory-acquired meningitis caused by Brucella abortus strain 19. J Infect Dis 1986;154:915–6.
  • Centers for Disease C, Prevention. Human exposure to Brucella abortus strain RB51–Kansas, 1997. MMWR Morb Mortal Wkly Rep 1998;47:172–5.
  • Clapp B, Skyberg JA, Yang X, Thornburg T, Walters N, Pascual DW. Protective live oral brucellosis vaccines stimulate Th1 and th17 cell responses. Infect Immun 2011;79:4165–74.
  • Vitry MA, De Trez C, Goriely S, et al. Crucial role of gamma interferon-producing CD4+ Th1 cells but dispensable function of CD8+ T cell, B cell, Th2, and Th17 responses in the control of Brucella melitensis infection in mice. Infect Immun 2012;80:4271–80.
  • Maldonado-Lopez R, Moser M. Dendritic cell subsets and the regulation of Th1/Th2 responses. Semin Immunol 2001;13:275–82.
  • Lapaque N, Moriyon I, Moreno E, Gorvel JP. Brucella lipopolysaccharide acts as a virulence factor. Curr Opin Microbiol 2005;8:60–6.
  • Erridge C, Bennett-Guerrero E, Poxton IR. Structure and function of lipopolysaccharides. Microbes Infect 2002;4:837–51.
  • Meng J, Lien E, Golenbock DT. MD-2-mediated ionic interactions between lipid A and TLR4 are essential for receptor activation. J Biol Chem 2010;285:8695–702.
  • Klett J, Reeves J, Oberhauser N, Perez-Regidor L, Martin-Santamaria S. Modulation of toll-like receptor 4. Insights from x-ray crystallography and molecular modeling. Curr Top Med Chem 2014;14:2672–83.
  • Moreno E, Berman DT, Boettcher LA. Biological activities of Brucella abortus lipopolysaccharides. Infect Immun 1981;31:362–70.
  • Goldstein J, Hoffman T, Frasch C, et al. Lipopolysaccharide (LPS) from Brucella abortus is less toxic than that from Escherichia coli, suggesting the possible use of B. abortus or LPS from B. abortus as a carrier in vaccines. Infect Immun 1992;60:1385–9.
  • Conde-Alvarez R, Arce-Gorvel V, Iriarte M, et al. The lipopolysaccharide core of Brucella abortus acts as a shield against innate immunity recognition. PLoS Pathog 2012;8:e1002675.
  • Barquero-Calvo E, Mora-Cartin R, Arce-Gorvel V, et al. Brucella abortus Induces the Premature Death of Human Neutrophils through the Action of Its Lipopolysaccharide. PLoS Pathog 2015;11:e1004853.
  • Eisenschenk FC, Houle JJ, Hoffmann EM. Mechanism of serum resistance among Brucella abortus isolates. Vet Microbiol 1999;68:235–44.
  • Jimenez de Bagues MP, Terraza A, Gross A, Dornand J. Different responses of macrophages to smooth and rough Brucella spp.: relationship to virulence. Infect Immun 2004;72:2429–33.
  • Mancilla M. Smooth to Rough Dissociation in Brucella: The Missing Link to Virulence. Front Cell Infect Microbiol 2015;5:98.
  • Fontana C, Conde-Alvarez R, Stahle J, et al. Structural Studies of Lipopolysaccharide-defective Mutants from Brucella melitensis Identify a Core Oligosaccharide Critical in Virulence. J Biol Chem 2016;291:7727–41.
  • Gil-Ramirez Y, Conde-Alvarez R, Palacios-Chaves L, et al. The identification of wadB, a new glycosyltransferase gene, confirms the branched structure and the role in virulence of the lipopolysaccharide core of Brucella abortus. Microb Pathogen 2014;73:53–9.
  • Martinez de Tejada G, Pizarro-Cerda J, Moreno E, Moriyon I. The outer membranes of Brucella spp. are resistant to bactericidal cationic peptides. Infect Immun 1995;63:3054–61.
  • Martirosyan A, Moreno E, Gorvel JP. An evolutionary strategy for a stealthy intracellular Brucella pathogen. Immunol Rev 2011;240:211–34.
  • Byndloss MX, Tsolis RM. Brucella spp. Virulence Factors and Immunity. Annu Rev Anim Biosci 2016;4:111–27.
  • Campos MA, Rosinha GM, Almeida IC, et al. Role of Toll-like receptor 4 in induction of cell-mediated immunity and resistance to Brucella abortus infection in mice. Infect Immun 2004;72:176–86.
  • Copin R, De Baetselier P, Carlier Y, Letesson JJ, Muraille E. MyD88-dependent activation of B220-CD11b+LY-6C+ dendritic cells during Brucella melitensis infection. J Immunol 2007;178:5182–91.
  • Weiss DS, Takeda K, Akira S, Zychlinsky A, Moreno E. MyD88, but not toll-like receptors 4 and 2, is required for efficient clearance of Brucella abortus. Infect Immun 2005;73:5137–43.
  • Mohammadi M, Kianmehr Z, Kaboudanian Ardestani S, Gharegozlou B. Improved immunogenicity of tetanus toxoid by Brucella abortus S19 LPS adjuvant. Iran J Immunol 2014;11:189–99.
  • Kianmehr Z, Soleimanjahi H, Ardestani SK, Fotouhi F, Abdoli A. Influence of Brucella abortus lipopolysaccharide as an adjuvant on the immunogenicity of HPV-16 L1VLP vaccine in mice. Med Microbiol Immunol 2015;204:205–13.
  • Hey YY, O'Neill HC. Murine spleen contains a diversity of myeloid and dendritic cells distinct in antigen presenting function. J Cell Mol Med 2012;16:2611–9.
  • Zhao Y, Hanniffy S, Arce-Gorvel V, et al. Immunomodulatory properties of Brucella melitensis lipopolysaccharide determinants on mouse dendritic cells in vitro and in vivo. Virulence 2017:0 doi. 10.1080/21505594.2017.1386831.
  • Velasco J, Bengoechea JA, Brandenburg K, et al. Brucella abortus and its closest phylogenetic relative, Ochrobactrum spp., differ in outer membrane permeability and cationic peptide resistance. Infect Immun 2000;68:3210–8.
  • Munoz PM, Marin CM, Monreal D, et al. Efficacy of several serological tests and antigens for diagnosis of bovine brucellosis in the presence of false-positive serological results due to Yersinia enterocolitica O:9. Clin Diagn Lab Immunol 2005;12:141–51.
  • Durward-Diioia M, Harms J, Khan M, Hall C, Smith JA, Splitter GA. CD8+ T cell exhaustion, suppressed gamma interferon production, and delayed memory response induced by chronic Brucella melitensis infection. Infect Immun 2015;83:4759–71.
  • Durward M, Radhakrishnan G, Harms J, Bareiss C, Magnani D, Splitter GA. Active evasion of CTL mediated killing and low quality responding CD8+ T cells contribute to persistence of brucellosis. PLoS One 2012;7:e34925.
  • Conde-Alvarez R, Arce-Gorvel V, Gil-Ramirez Y, et al. Lipopolysaccharide as a target for brucellosis vaccine design. Microbial pathogenesis 2013;58:29–34.