1
|
Fu S, Qian K, Tu X, Lu J, Yao T, Ye L, Ye J. Comparative analysis of intestinal structure, enzyme activity, intestinal microbiota and gene expression in different segments of pufferfish (Takifugu Obscurus). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101341. [PMID: 39427531 DOI: 10.1016/j.cbd.2024.101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
The structure of fish intestines does not have a clear regional division, while the function of the intestines may be related to their structure. Therefore, in this study, the delimitation of intestinal segments in pufferfish (Takifugu obscurus) was achieved by morphological analysis. Subsequently, enzyme activity, intestinal microbiota, and gene expression were examined to compare the differences among the pufferfish various segments. According to four morphological parameters: height of mucosa folds (HF), width of mucosa folds (WF), thickness of muscularis (TM), and cross-sectional area (CSA), the pufferfish's intestine was divided into anterior intestine (AI), middle intestine (MI), and posterior intestine (PI). The activity levels of amylase, lipase, and trypsin in the AI and MI were significantly higher than these in the PI. According to the analysis of 16S rDNA, the dominant microbiota at the phylum level in the different segments were Epsilonbacteraeota, Spirochaetes, and Proteobacteria. At the genus level, there were variations observed in the relative abundance of Brevinema, Mycobacterium, Bradyrhizobium, and Microvirga. α diversity analysis revealed that the richness indexes (Ace and Chao1) were the lowest in the MI, while β diversity analysis revealed significant difference in intestinal microbial community composition among the three intestinal segments. Furthermore, RNA-Seq was used to identify differential expression genes (DEGs) and biological pathways among the different intestinal segments. The DEGs between the AI and MI were enriched in pancreatic secretion and protein digestion and absorption, those between AI and PI were involved in ascorbate and aldarate metabolism and glutathione metabolism, and those between MI and PI were involved in steroid biosynthesis, fat digestion and absorption, vitamin digestion and absorption, and glycine, serine and threonine metabolism. In conclusion, the presented results compare and analyze the differences in various intestinal segments of pufferfish, which will be conductive to future exploration of the functions of these different segments.
Collapse
Affiliation(s)
- Shengli Fu
- Key Laboratory of Aquatic Product Processing, Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou 510300, PR China
| | - Kun Qian
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, School of Life Sciences, South China Normal University, Guangzhou 510631, PR China
| | - Xiao Tu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, School of Life Sciences, South China Normal University, Guangzhou 510631, PR China
| | - Jie Lu
- Key Laboratory of Aquatic Product Processing, Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou 510300, PR China
| | - Tuo Yao
- Key Laboratory of Aquatic Product Processing, Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou 510300, PR China
| | - Lingtong Ye
- Key Laboratory of Aquatic Product Processing, Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Science, Guangzhou 510300, PR China.
| | - Jianmin Ye
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, School of Life Sciences, South China Normal University, Guangzhou 510631, PR China.
| |
Collapse
|
2
|
Groshong AM, Gibbons NE, Moore BP, Bellamy WT, Blevins JS. The plasmid-encoded members of paralogous gene family 52 are dispensable to the enzootic cycle of Borrelia burgdorferi. Infect Immun 2024; 92:e0021424. [PMID: 39120148 PMCID: PMC11475691 DOI: 10.1128/iai.00214-24] [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: 05/15/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
Lyme disease, the leading vector-borne disease in the United States and Europe, develops after infection with Borrelia burgdorferi sensu lato bacteria. Transmission of the spirochete from the tick vector to a vertebrate host requires global changes in gene expression that are controlled, in part, by the Rrp2/RpoN/RpoS alternative sigma factor cascade. Transcriptional studies defining the B. burgdorferi RpoS regulon have suggested that RpoS activates the transcription of paralogous family 52 (PFam52) genes. In strain B31, PFam52 genes (bbi42, bbk53, and bbq03) encode a set of conserved hypothetical proteins with >89% amino acid identity that are predicted to be surface-localized. Extensive homology among members of paralogous families complicates studies of protein contributions to pathogenicity as the potential for functional redundancy will obfuscate findings. Using a sequential mutagenesis approach, we generated clones expressing a single PFam52 paralog, as well as a strain deficient in all three. The single paralog expressing strains were used to confirm BBI42, BBK53, and BBQ03 surface localization and RpoS regulation. Surprisingly, the PFam52-deficient strain was able to infect mice and complete the enzootic cycle similar to the wild-type parental strain. Indeed, the presence of numerous pseudogenes that contain frameshifts or internal stop codons among the PFam52 genes suggests that they may be subjected to gene loss in B. burgdorferi's reduced genome. Alternatively, the lack of phenotype might reflect the limitations of the experimental mouse infection model.
Collapse
Affiliation(s)
- Ashley M. Groshong
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Medicine, UConn Health, Farmington, Connecticut, USA
- Laboratory of Bacteriology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Nora E. Gibbons
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Brendan P. Moore
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - William T. Bellamy
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jon S. Blevins
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
3
|
Strnad M, Vancová M, Rego ROM. Borrelia (Borreliella) burgdorferi. Trends Microbiol 2024:S0966-842X(24)00228-2. [PMID: 39341692 DOI: 10.1016/j.tim.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Affiliation(s)
- Martin Strnad
- Institute of Parasitology, Biology Centre CAS, Branišovská 31, 37005, České Budějovice, Czech Republic; Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, České Budějovice, Czech Republic
| | - Marie Vancová
- Institute of Parasitology, Biology Centre CAS, Branišovská 31, 37005, České Budějovice, Czech Republic; Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, České Budějovice, Czech Republic
| | - Ryan O M Rego
- Institute of Parasitology, Biology Centre CAS, Branišovská 31, 37005, České Budějovice, Czech Republic; Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, České Budějovice, Czech Republic.
| |
Collapse
|
4
|
Bourgeois JS, Hu LT. Hitchhiker's Guide to Borrelia burgdorferi. J Bacteriol 2024; 206:e0011624. [PMID: 39140751 PMCID: PMC11411949 DOI: 10.1128/jb.00116-24] [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] [Indexed: 08/15/2024] Open
Abstract
Don't Panic. In the nearly 50 years since the discovery of Lyme disease, Borrelia burgdorferi has emerged as an unlikely workhorse of microbiology. Interest in studying host-pathogen interactions fueled significant progress in making the fastidious microbe approachable in laboratory settings, including the development of culture methods, animal models, and genetic tools. By developing these systems, insight has been gained into how the microbe is able to survive its enzootic cycle and cause human disease. Here, we discuss the discovery of B. burgdorferi and its development as a model organism before diving into the critical lessons we have learned about B. burgdorferi biology at pivotal stages of its lifecycle: gene expression changes during the tick blood meal, colonization of a new vertebrate host, and developing a long-lasting infection in that vertebrate until a new tick feeds. Our goal is to highlight the advancements that have facilitated B. burgdorferi research and identify gaps in our current understanding of the microbe.
Collapse
Affiliation(s)
- Jeffrey S Bourgeois
- Department of Molecular Biology and Microbiology, Tufts University Lyme Disease Initiative, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Linden T Hu
- Department of Molecular Biology and Microbiology, Tufts University Lyme Disease Initiative, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Raghunandanan S, Priya R, Lin G, Alanazi F, Zoss A, Warren E, Yang XF. Positive feedback regulation between RpoS and BosR in the Lyme disease pathogen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.14.613071. [PMID: 39314342 PMCID: PMC11419129 DOI: 10.1101/2024.09.14.613071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
In Borrelia burgdorferi, the Lyme disease pathogen, differential gene expression is primarily controlled by the alternative sigma factor RpoS (σS). Understanding how RpoS levels are regulated is crucial for elucidating how B. burgdorferi is maintained throughout its enzootic cycle. Our recent studies have shown that a homolog of Fur/PerR repressor/activator, BosR, functions as an RNA-binding protein that controls the rpoS mRNA stability. However, the mechanisms of regulation of BosR, particularly in response to host signals and environmental cues, remain largely unclear. In this study, we revealed a positive feedback loop between RpoS and BosR, where RpoS post-transcriptionally regulates BosR levels. Specifically, mutation or deletion of rpoS significantly reduced BosR levels, while artificial induction of rpoS resulted in a dose-dependent increase in BosR levels. Notably, RpoS does not affect bosR mRNA levels but instead modulates the turnover rate of the BosR protein. Furthermore, we demonstrated that environmental cues do not directly influence bosR expression but instead induce rpoS transcription and RpoS production, thereby enhancing BosR protein levels. This discovery adds a new layer of complexity to the RpoN-RpoS pathway and suggests the need to re-evaluate the factors and signals previously believed to regulate RpoS levels through BosR.
Collapse
Affiliation(s)
- Sajith Raghunandanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Gaofeng Lin
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Fuad Alanazi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Andrew Zoss
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Elise Warren
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
6
|
Zafar K, Azuama OC, Parveen N. Current and emerging approaches for eliminating Borrelia burgdorferi and alleviating persistent Lyme disease symptoms. Front Microbiol 2024; 15:1459202. [PMID: 39345262 PMCID: PMC11427371 DOI: 10.3389/fmicb.2024.1459202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Lyme disease is the most prevalent tick-borne infection caused by Borrelia burgdorferi bacteria in North America. Other Borrelia species are predominately the cause of this disease in Eurasia with some distinct and various overlapping manifestations. Consequently, caution must be exercised when comparing the disease and its manifestations and treatment regimens in North America and Europe. Diagnosis of the early Lyme disease remains difficult using the currently FDA approved serological tests in the absence of a reported tick bite or of erythema migrans in many individuals, non-specific initial symptoms, and the absence of detectable anti-Borrelia antibodies in the prepatent period of infection. Furthermore, it is difficult to distinguish persistence of infection and disease versus reinfection in the endemic regions of Lyme disease by serological assays. If early infection remains untreated, spirochetes can disseminate and could affect various organs in the body with a variety of disease manifestations including arthralgias and musculoskeletal pain, neurologic symptoms and anomalies, and acrodermatitis chronicum atrophicans (ACA) in Europe. Although most patients recover after antibiotic treatment, an estimated ∼10-20% patients in the United States show persistence of symptoms known as post-treatment Lyme disease syndrome (PTLDS). The causes and biomarkers of PTLDS are not well-defined; however, several contributing factors with inconsistent degree of supporting evidence have been suggested. These include antigenic debris, dysregulation of immunological response, bacterial persisters, or combination of these features. This review highlights currently employed treatment approaches describing different antimicrobials used, and vaccine candidates tried to prevent B. burgdorferi infection.
Collapse
Affiliation(s)
| | | | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
7
|
Krusenstjerna AC, Jusufovic N, Saylor TC, Stevenson B. DnaA modulates the gene expression and morphology of the Lyme disease spirochete. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.08.598065. [PMID: 38895450 PMCID: PMC11185795 DOI: 10.1101/2024.06.08.598065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
All bacteria encode a multifunctional DNA-binding protein, DnaA, which initiates chromosomal replication. Despite having the most complex, segmented bacterial genome, little is known about Borrelia burgdorferi DnaA and its role in maintaining the spirochete's physiology. In this work we utilized inducible CRISPR-interference and overexpression to modulate cellular levels of DnaA to better understand this essential protein. Dysregulation of DnaA, either up or down, increased or decreased cell lengths, respectively, while also significantly slowing replication rates. Using fluorescent microscopy, we found the DnaA CRISPRi mutants had increased numbers of chromosomes with irregular spacing patterns. DnaA-depleted spirochetes also exhibited a significant defect in helical morphology. RNA-seq of the conditional mutants showed significant changes in the levels of transcripts involved with flagellar synthesis, elongation, cell division, virulence, and other functions. These findings demonstrate that the DnaA plays a commanding role in maintaining borrelial growth dynamics and protein expression, which are essential for the survival of the Lyme disease spirochete.
Collapse
Affiliation(s)
- Andrew C Krusenstjerna
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Nerina Jusufovic
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Timothy C Saylor
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Brian Stevenson
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Department of Entomology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
8
|
Danner R, Prochniak LM, Pereckas M, Rouse JR, Wahhab A, Hackner LG, Lochhead RB. Identification of Major Histocompatibility Complex Class II Epitopes From Lyme Autoantigen Apolipoprotein B-100 and Borrelia burgdorferi Mcp4 in Murine Lyme Arthritis. J Infect Dis 2024; 230:S27-S39. [PMID: 39140726 PMCID: PMC11322890 DOI: 10.1093/infdis/jiae324] [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] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND During infection with the Lyme arthritis (LA) pathogen Borrelia burgdorferi, T-cell responses to both host and pathogen are dysregulated, resulting in chronic infection and frequent development of autoimmunity. METHODS To assess CD4+ T-cell epitopes presented during development of LA, we used an unbiased, immunopeptidomics approach to characterize the major histocompatibility complex (MHC) class II immunopeptidome in B burgdorferi-infected C57BL/6 (B6) mice, which develop mild, self-limiting LA, and infected B6 Il10-/- mice, which develop severe, persistent LA at 0, 4, and 16 weeks postinfection (22-23 mice per group). RESULTS Peptides derived from proteins involved in adaptive T- and B-cell responses and cholesterol metabolism, including human Lyme autoantigen apolipoprotein B-100 (apoB-100), were enriched in infected Il10-/- mice; whereas peptides derived from proteins involved in neutrophil extracellular net formation were enriched in infected B6 mice. Presentation of apoB-100 peptides showed evidence of epitope expansion during infection. Of several identified B burgdorferi peptides, only 1, a methyl-accepting chemotaxis protein peptide Mcp4442-462, was immunogenic. CONCLUSIONS ApoB-100, a human Lyme autoantigen, undergoes marked epitope expansion during LA development. The paucity of immunogenic B burgdorferi epitopes supports previous findings suggesting CD4+ T-cell responses are suppressed in murine LA.
Collapse
Affiliation(s)
- Rebecca Danner
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Lauren M Prochniak
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michaela Pereckas
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Joseph R Rouse
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Amanda Wahhab
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Lauren G Hackner
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Robert B Lochhead
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
9
|
Scott C, Dias AP, De Buck J. Adherence and metal-ion acquisition gene expression increases during infection with Treponema phagedenis strains from bovine digital dermatitis. Infect Immun 2024; 92:e0011724. [PMID: 38940601 PMCID: PMC11320908 DOI: 10.1128/iai.00117-24] [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: 03/13/2024] [Accepted: 05/16/2024] [Indexed: 06/29/2024] Open
Abstract
Digital dermatitis (DD) is an ulcerative foot lesion on the heel bulbs of dairy cattle. DD is a polymicrobial disease with no precise etiology, although Treponema spirochetes are found disproportionally abundant in diseased tissue. Within Treponema, several different species are found in DD; however, the species Treponema phagedenis is uniformly found in copious quantities and deep within the skin layers of the active, ulcerative stages of disease. The pathogenic mechanisms these bacteria use to persist in the skin and the precise role they play in the pathology of DD are widely unknown. To explore the pathogenesis and virulence of Treponema phagedenis, newly isolated strains of this species were investigated in a subcutaneous murine abscess model. In the first trial, a dosage study was conducted to compare the pathogenicity of different strains across three different treponemes per inoculum (TPI) doses based on abscess volumes. In the second trial, the expression levels of 11 putative virulence genes were obtained to gain insight into their involvement in pathogenesis. During the RT-qPCR analysis, it was determined that genes encoding for two metal-ion import lipoproteins and two adherence genes were found highly upregulated during infection. Conversely, two genes involved in motility and chemotaxis were found to not be significantly upregulated or utilized during infection. These results were supported by gene expression data from natural M2 lesions of dairy cattle. This gene expression analysis could highlight the preference in strategy for T. phagedenis to persist and adhere in the host rather than engage in motility and disseminate.
Collapse
Affiliation(s)
- Colton Scott
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Angelica P. Dias
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeroen De Buck
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Murphy BT, Wiepen JJ, Graham DE, Swanson SK, Kashipathy MM, Cooper A, Battaile KP, Johnson DK, Florens L, Blevins JS, Lovell S, Zückert WR. Borrelia burgdorferi BB0346 is an Essential, Structurally Variant LolA Homolog that is Primarily Required for Homeostatic Localization of Periplasmic Lipoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606844. [PMID: 39149330 PMCID: PMC11326224 DOI: 10.1101/2024.08.06.606844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
In diderm bacteria, the Lol pathway canonically mediates the periplasmic transport of lipoproteins from the inner membrane (IM) to the outer membrane (OM) and therefore plays an essential role in bacterial envelope homeostasis. After extrusion of modified lipoproteins from the IM via the LolCDE complex, the periplasmic chaperone LolA carries lipoproteins through the periplasm and transfers them to the OM lipoprotein insertase LolB, itself a lipoprotein with a LolA-like fold. Yet, LolB homologs appear restricted to γ-proteobacteria and are missing from spirochetes like the tick-borne Lyme disease pathogen Borrelia burgdorferi, suggesting a different hand-off mechanism at the OM. Here, we solved the crystal structure of the B. burgdorferi LolA homolog BB0346 (LolABb) at 1.9 Å resolution. We identified multiple structural deviations in comparative analyses to other solved LolA structures, particularly a unique LolB-like protruding loop domain. LolABb failed to complement an Escherichia coli lolA knockout, even after codon optimization, signal I peptide adaptation, and a C-terminal chimerization which had allowed for complementation with an α-proteobacterial LolA. Analysis of a conditional B. burgdorferi lolA knockout strain indicated that LolABb was essential for growth. Intriguingly, protein localization assays indicated that initial depletion of LolABb led to an emerging mislocalization of both IM and periplasmic OM lipoproteins, but not surface lipoproteins. Together, these findings further support the presence of two separate primary secretion pathways for periplasmic and surface OM lipoproteins in B. burgdorferi and suggest that the distinct structural features of LolABb allow it to function in a unique LolB-deficient lipoprotein sorting system.
Collapse
Affiliation(s)
- Bryan T. Murphy
- University of Kansas School of Medicine, Department of Microbiology, Molecular Genetics & Immunology, Kansas City, Kansas
| | - Jacob J. Wiepen
- University of Kansas School of Medicine, Department of Microbiology, Molecular Genetics & Immunology, Kansas City, Kansas
| | - Danielle E. Graham
- University of Arkansas for Medical Sciences, Department of Microbiology & Immunology, Little Rock, Arkansas
| | | | - Maithri M. Kashipathy
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, 98109, USA
| | - Anne Cooper
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, 98109, USA
- University of Kansas, Protein Structure and X-ray Crystallography Laboratory, Lawrence, Kansas
| | | | - David K. Johnson
- University of Kansas, Protein Structure and X-ray Crystallography Laboratory, Lawrence, Kansas
| | | | - Jon S. Blevins
- University of Arkansas for Medical Sciences, Department of Microbiology & Immunology, Little Rock, Arkansas
| | - Scott Lovell
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, 98109, USA
- University of Kansas, Protein Structure and X-ray Crystallography Laboratory, Lawrence, Kansas
| | - Wolfram R. Zückert
- University of Kansas School of Medicine, Department of Microbiology, Molecular Genetics & Immunology, Kansas City, Kansas
| |
Collapse
|
11
|
Olsen KJ, Sachan S, Baumgarth N. Mouse Models for the Study of Borrelia burgdorferi Infection. Curr Protoc 2024; 4:e1127. [PMID: 39193882 PMCID: PMC11361713 DOI: 10.1002/cpz1.1127] [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] [Indexed: 08/29/2024]
Abstract
Lyme disease, a tickborne illness caused by Borrelia burgdorferi, is an emerging, significant public health concern. B. burgdorferi infections are challenging to study because of their complex life cycle that requires adaptation to both ticks and mammalian hosts for long-term survival and transmission. Bacterial adaptation is accomplished through extensive gene expression alterations in response to environmental cues that remain to be more fully explored. Mouse models of infection serve as valuable tools for studying B. burgdorferi adaptation to the mammalian host and the spirochete's ability to cause persistent infections and thus to interact with and evade the immune system. This article details three mouse models that differ in their primary methods of infection: infestation with B. burgdorferi infected ticks, intradermal inoculation of culture-grown spirochetes, and infection via subcutaneous transplantation of infected tissue. Each method offers unique advantages and limitations. Tick infestation is the route of natural transmission but presents logistical challenges. Syringe inoculation is easy and provides precise control over the infectious dose, but infection is with culture-adapted bacteria. Transplantation of infected tissue introduces mammalian-host-adapted B. burgdorferi in precise anatomical locations, but misses the transfer of tick factors affecting immunity. Detailed protocols are provided for each of the three infection routes, and pros and cons of each method are outlined to help researchers identify the best approach for a research question to be addressed. A protocol is also provided for the treatment of mice with antibiotics that reliably eliminates detectable spirochetes from the animals. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Syringe inoculation of mice with cultured B. burgdorferi and collection of necropsy tissues Basic Protocol 2: Infection of mice with B. burgdorferi via tick infestation Basic Protocol 3: Infection of mice with host-adapted B. burgdorferi via tissue transplant Support Protocol: Clearance of B. burgdorferi by antibiotic treatment.
Collapse
Affiliation(s)
- Kimberly J. Olsen
- Dept. Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis
| | - Shilpa Sachan
- Dept. Molecular Microbiology and Immunology, Lyme and Tickborne Diseases Research and Education Institute, Bloomberg School of Public Health
| | - Nicole Baumgarth
- Dept. Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis
- Dept. Molecular Microbiology and Immunology, Lyme and Tickborne Diseases Research and Education Institute, Bloomberg School of Public Health
- Dept. Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University
| |
Collapse
|
12
|
Murphy B, Veras J, Kolli S, Formanowski B, Greenberg P, Kleinman L, Malhotra A, Bhise V. Seroprevalence of Lyme Disease in Children With Facial Nerve Palsy. J Child Neurol 2024; 39:310-316. [PMID: 39196299 DOI: 10.1177/08830738241272069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
This retrospective chart review examined children with documented Lyme disease serology in New Jersey aged <21 years presenting with facial nerve palsy. The presence of symptoms including tick bite, fever, headache, and arthritis was recorded. Data were categorized based on demographic factors, and multivariate regression was employed. We enrolled 122 children, 54% female (mean age of 11.4 ± 5.1 years); 22.1% had Lyme disease. Fever was a significant predictor of Lyme disease (P = .01), confirmed by multivariate regression (odds ratio [OR] = 16.11, 95% confidence interval [CI] = 2.04, 366.14), as was male gender (P = .01, OR = 3.68, 95% CI = 1.21, 12.89). This association held especially true in Lyme-endemic regions (prevalence ≥ 0.35). The combination of headache with fever was also significantly predictive (P = .01). We found no significant predictive value in the remaining symptoms. These findings suggest that clinical predictors may be useful in diagnosing Lyme disease and initiating early empiric treatment.
Collapse
Affiliation(s)
- Bethany Murphy
- Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Julissa Veras
- Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Sindhura Kolli
- Rutgers Robert Wood Johnson School of Biomedical Sciences, Piscataway, NJ, USA
| | - Brielle Formanowski
- Biostatistics and Epidemiology Services Center, Rutgers School of Public Health, Rutgers University, Piscataway, NJ, USA
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Rutgers University, Piscataway, NJ, USA
| | - Patricia Greenberg
- Biostatistics and Epidemiology Services Center, Rutgers School of Public Health, Rutgers University, Piscataway, NJ, USA
| | - Lawrence Kleinman
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Amisha Malhotra
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Vikram Bhise
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
13
|
Hart TM, Sonnert ND, Tang X, Chaurasia R, Allen PE, Hunt JR, Read CB, Johnson EE, Arora G, Dai Y, Cui Y, Chuang YM, Yu Q, Rahman MS, Mendes MT, Rolandelli A, Singh P, Tripathi AK, Ben Mamoun C, Caimano MJ, Radolf JD, Lin YP, Fingerle V, Margos G, Pal U, Johnson RM, Pedra JHF, Azad AF, Salje J, Dimopoulos G, Vinetz JM, Carlyon JA, Palm NW, Fikrig E, Ring AM. An atlas of human vector-borne microbe interactions reveals pathogenicity mechanisms. Cell 2024; 187:4113-4127.e13. [PMID: 38876107 DOI: 10.1016/j.cell.2024.05.023] [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: 02/18/2023] [Revised: 01/15/2024] [Accepted: 05/13/2024] [Indexed: 06/16/2024]
Abstract
Vector-borne diseases are a leading cause of death worldwide and pose a substantial unmet medical need. Pathogens binding to host extracellular proteins (the "exoproteome") represents a crucial interface in the etiology of vector-borne disease. Here, we used bacterial selection to elucidate host-microbe interactions in high throughput (BASEHIT)-a technique enabling interrogation of microbial interactions with 3,324 human exoproteins-to profile the interactomes of 82 human-pathogen samples, including 30 strains of arthropod-borne pathogens and 8 strains of related non-vector-borne pathogens. The resulting atlas revealed 1,303 putative interactions, including hundreds of pairings with potential roles in pathogenesis, including cell invasion, tissue colonization, immune evasion, and host sensing. Subsequent functional investigations uncovered that Lyme disease spirochetes recognize epidermal growth factor as an environmental cue of transcriptional regulation and that conserved interactions between intracellular pathogens and thioredoxins facilitate cell invasion. In summary, this interactome atlas provides molecular-level insights into microbial pathogenesis and reveals potential host-directed targets for next-generation therapeutics.
Collapse
Affiliation(s)
- Thomas M Hart
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nicole D Sonnert
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Xiaotian Tang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Reetika Chaurasia
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Paige E Allen
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Jason R Hunt
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Curtis B Read
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Emily E Johnson
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Epidemiology and Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
| | - Gunjan Arora
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yile Dai
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yingjun Cui
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yu-Min Chuang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Qian Yu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - M Sayeedur Rahman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pallavi Singh
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Abhai K Tripathi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Choukri Ben Mamoun
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Melissa J Caimano
- Department of Medicine, UConn Health, Farmington, CT 06030, USA; Department of Pediatrics, UConn Health, Farmington, CT 06030, USA; Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030, USA
| | - Justin D Radolf
- Department of Medicine, UConn Health, Farmington, CT 06030, USA; Department of Pediatrics, UConn Health, Farmington, CT 06030, USA; Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA; Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
| | - Volker Fingerle
- Bavarian Health and Food Safety Authority, Oberschleißheim, Munich 85764, Bavaria, Germany
| | - Gabriele Margos
- Bavarian Health and Food Safety Authority, Oberschleißheim, Munich 85764, Bavaria, Germany
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Raymond M Johnson
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Abdu F Azad
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeanne Salje
- Department of Pathology, University of Cambridge, Cambridge CB2 1TN, UK; Department of Biochemistry, University of Cambridge, Cambridge CB2 1TN, UK
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joseph M Vinetz
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Laboratorio ICEMR-Amazonia, Laboratorios de Investigación Y Desarrollo, Facultad de Ciencias Y Filosofia, Universidad Peruana Cayetano Heredia, Lima 15102, Peru; Instituto de Medicina Tropical Alexander Von Humboldt, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA.
| | - Noah W Palm
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Erol Fikrig
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Aaron M Ring
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98102, USA.
| |
Collapse
|
14
|
Bourgeois JS, You SS, Clendenen LH, Shrestha M, Petnicki-Ocwieja T, Telford SR, Hu LT. Comparative reservoir competence of Peromyscus leucopus, C57BL/6J, and C3H/HeN for Borrelia burgdorferi B31. Appl Environ Microbiol 2024; 90:e0082224. [PMID: 38899883 PMCID: PMC11267898 DOI: 10.1128/aem.00822-24] [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: 04/26/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Borrelia burgdorferi, a Lyme disease spirochete, causes a range of acute and chronic maladies in humans. However, a primary vertebrate reservoir in the United States, the white-footed deermouse Peromyscus leucopus, is reported not to have reduced fitness following infection. Although laboratory strains of Mus musculus mice have successfully been leveraged to model acute human Lyme disease, the ability of these rodents to model B. burgdorferi-P. leucopus interactions remains understudied. Here, we compared infection of P. leucopus with B. burgdorferi B31 with infection of the traditional B. burgdorferi murine models-C57BL/6J and C3H/HeN Mus musculus, which develop signs of inflammation akin to human disease. We find that B. burgdorferi was able to reach much higher burdens (10- to 30-times higher) in multiple M. musculus skin sites and that the overall dynamics of infection differed between the two rodent species. We also found that P. leucopus remained transmissive to larval Ixodes scapularis for a far shorter period than either M. musculus strain. In line with these observations, we found that P. leucopus does launch a modest but sustained inflammatory response against B. burgdorferi in the skin, which we hypothesize leads to reduced bacterial viability and rodent-to-tick transmission in these hosts. Similarly, we also observe evidence of inflammation in infected P. leucopus hearts. These observations provide new insight into reservoir species and the B. burgdorferi enzootic cycle.IMPORTANCEA Lyme disease-causing bacteria, Borrelia burgdorferi, must alternate between infecting a vertebrate host-usually rodents or birds-and ticks. In order to be successful in that endeavor, the bacteria must avoid being killed by the vertebrate host before it can infect a new larval tick. In this work, we examine how B. burgdorferi and one of its primary vertebrate reservoirs, Peromyscus leucopus, interact during an experimental infection. We find that B. burgdorferi appears to colonize its natural host less successfully than conventional laboratory mouse models, which aligns with a sustained seemingly anti-bacterial response by P. leucopus against the microbe. These data enhance our understanding of P. leucopus host-pathogen interactions and could potentially serve as a foundation to uncover ways to disrupt the spread of B. burgdorferi in nature.
Collapse
Affiliation(s)
- Jeffrey S. Bourgeois
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, USA
- Tufts University, Tufts Lyme Disease Initiative, Boston, Massachusetts, USA
| | - Stephanie S. You
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, USA
- Tufts University, Tufts Lyme Disease Initiative, Boston, Massachusetts, USA
| | - Luke H. Clendenen
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, USA
- Tufts University, Tufts Lyme Disease Initiative, Boston, Massachusetts, USA
| | - Muskan Shrestha
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, USA
- Tufts University, Tufts Lyme Disease Initiative, Boston, Massachusetts, USA
| | - Tanja Petnicki-Ocwieja
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, USA
- Tufts University, Tufts Lyme Disease Initiative, Boston, Massachusetts, USA
| | - Sam R. Telford
- Tufts University, Tufts Lyme Disease Initiative, Boston, Massachusetts, USA
- Department of Infectious Disease and Global Health, Tufts University, North Grafton, Massachusetts, USA
| | - Linden T. Hu
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, USA
- Tufts University, Tufts Lyme Disease Initiative, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Pustijanac E, Buršić M, Millotti G, Paliaga P, Iveša N, Cvek M. Tick-Borne Bacterial Diseases in Europe: Threats to public health. Eur J Clin Microbiol Infect Dis 2024; 43:1261-1295. [PMID: 38676855 DOI: 10.1007/s10096-024-04836-5] [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: 02/16/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Tick-borne diseases, caused by bacterial pathogens, pose a growing threat to public health in Europe. This paper provides an overview of the historical context of the discovery of the most impactful pathogens transmitted by ticks, including Borrelia burgdorferi sensu lato, Rickettsia spp., Anaplasma spp., Francisella spp., Ehrlichia spp., and Neoehrlichia mikurensis. Understanding the historical context of their discovery provides insight into the evolution of our understanding of these pathogens. METHODS AND RESULTS Systematic investigation of the prevalence and transmission dynamics of these bacterial pathogens is provided, highlighting the intricate relationships among ticks, host organisms, and the environment. Epidemiology is explored, providing an in-depth analysis of clinical features associated with infections. Diagnostic methodologies undergo critical examination, with a spotlight on technological advancements that enhance detection capabilities. Additionally, the paper discusses available treatment options, addressing existing therapeutic strategies and considering future aspects. CONCLUSIONS By integrating various pieces of information on these bacterial species, the paper aims to provide a comprehensive resource for researchers and healthcare professionals addressing the impact of bacterial tick-borne diseases in Europe. This review underscores the importance of understanding the complex details influencing bacterial prevalence and transmission dynamics to better combat these emerging public health threats.
Collapse
Affiliation(s)
- Emina Pustijanac
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia.
| | - Moira Buršić
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Gioconda Millotti
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Paolo Paliaga
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Neven Iveša
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, Zagrebačka 30, 52100, Pula, Croatia
| | - Maja Cvek
- Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000, Rijeka, Croatia
- Teaching Institute of Public Health of the Region of Istria, Nazorova 23, 52100, Pula, Croatia
| |
Collapse
|
16
|
Thompson C, Waldron C, George S, Ouyang Z. Assessment of the hypothetical protein BB0616 in the murine infection of Borrelia burgdorferi. Infect Immun 2024; 92:e0009024. [PMID: 38700336 PMCID: PMC11237664 DOI: 10.1128/iai.00090-24] [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: 03/03/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
bb0616 of Borrelia burgdorferi, the Lyme disease pathogen, encodes a hypothetical protein of unknown function. In this study, we showed that BB0616 was not surface-exposed or associated with the membrane through localization analyses using proteinase K digestion and cell partitioning assays. The expression of bb0616 was influenced by a reduced pH but not by growth phases, elevated temperatures, or carbon sources during in vitro cultivation. A transcriptional start site for bb0616 was identified by using 5' rapid amplification of cDNA ends, which led to the identification of a functional promoter in the 5' regulatory region upstream of bb0616. By analyzing a bb0616-deficient mutant and its isogenic complemented counterparts, we found that the infectivity potential of the mutant was significantly attenuated. The inactivation of bb0616 displayed no effect on borrelial growth in the medium or resistance to oxidative stress, but the mutant was significantly more susceptible to osmotic stress. In addition, the production of global virulence regulators such as BosR and RpoS as well as virulence-associated outer surface lipoproteins OspC and DbpA was reduced in the mutant. These phenotypes were fully restored when gene mutation was complemented with a wild-type copy of bb0616. Based on these findings, we concluded that the hypothetical protein BB0616 is required for the optimal infectivity of B. burgdorferi, potentially by impacting B. burgdorferi virulence gene expression as well as survival of the spirochete under stressful conditions.
Collapse
Affiliation(s)
- Christina Thompson
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, USA
| | - Connor Waldron
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, USA
| | - Sierra George
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, USA
| | - Zhiming Ouyang
- Department of Molecular Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
17
|
Raghunandanan S, Zhang K, Zhang Y, Sze CW, Priya R, Luo Y, Lynch MJ, Crane BR, Li C, Yang XF. MCP5, a methyl-accepting chemotaxis protein regulated by both the Hk1-Rrp1 and Rrp2-RpoN-RpoS pathways, is required for the immune evasion of Borrelia burgdorferi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598185. [PMID: 38915556 PMCID: PMC11195095 DOI: 10.1101/2024.06.10.598185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Borrelia (or Borreliella) burgdorferi, the causative agent of Lyme disease, is a motile and invasive zoonotic pathogen, adept at navigating between its arthropod vector and mammalian host. While motility and chemotaxis are well established as essential for its enzootic cycle, the function of methyl-accepting chemotaxis proteins (MCPs) in the infectious cycle of B. burgdorferi remains unclear. In this study, we demonstrate that MCP5, one of the most abundant MCPs in B. burgdorferi, is differentially expressed in response to environmental signals as well as at different stages of the pathogen's enzootic cycle. Specifically, the expression of mcp5 is regulated by the Hk1-Rrp1 and Rrp2-RpoN-RpoS pathways, which are critical for the spirochete's colonization of the tick vector and mammalian host, respectively. Infection experiments with an mcp5 mutant revealed that spirochetes lacking MCP5 could not establish infections in either C3H/HeN mice or Severe Combined Immunodeficiency (SCID) mice, which are defective in adaptive immunity, indicating the essential role of MCP5 in mammalian infection. However, the mcp5 mutant could establish infection and disseminate in NOD SCID Gamma (NSG) mice, which are deficient in both adaptive and most innate immune responses, suggesting a crucial role of MCP5 in evading host innate immunity. In the tick vector, the mcp5 mutants survived feeding but failed to transmit to mice, highlighting the importance of MCP5 in transmission. Our findings reveal that MCP5, regulated by the Rrp1 and Rrp2 pathways, is critical for the establishment of infection in mammalian hosts by evading host innate immunity and is important for the transmission of spirochetes from ticks to mammalian hosts, underscoring its potential as a target for intervention strategies.
Collapse
Affiliation(s)
- Sajith Raghunandanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Kai Zhang
- Department of Oral Craniofacial Molecular Biology, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Yan Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China, 325035
| | - Ching Wooen Sze
- Department of Oral Craniofacial Molecular Biology, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Yongliang Luo
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China, 325035
| | - Michael J Lynch
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Brian R Crane
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Chunhao Li
- Department of Oral Craniofacial Molecular Biology, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
18
|
Nowak TA, Burke RL, Diuk-Wasser MA, Lin YP. Lizards and the enzootic cycle of Borrelia burgdorferi sensu lato. Mol Microbiol 2024; 121:1262-1272. [PMID: 38830767 DOI: 10.1111/mmi.15271] [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: 10/17/2023] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 06/05/2024]
Abstract
Emerging and re-emerging pathogens often stem from zoonotic origins, cycling between humans and animals, and are frequently vectored and maintained by hematophagous arthropod vectors. The efficiency by which these disease agents are successfully transmitted between vertebrate hosts is influenced by many factors, including the host on which a vector feeds. The Lyme disease bacterium Borrelia burgdorferi sensu lato has adapted to survive in complex host environments, vectored by Ixodes ticks, and maintained in multiple vertebrate hosts. The versatility of Lyme borreliae in disparate host milieus is a compelling platform to investigate mechanisms dictating pathogen transmission through complex networks of vertebrates and ticks. Squamata, one of the most diverse clade of extant reptiles, is comprised primarily of lizards, many of which are readily fed upon by Ixodes ticks. Yet, lizards are one of the least studied taxa at risk of contributing to the transmission and life cycle maintenance of Lyme borreliae. In this review, we summarize the current evidence, spanning from field surveillance to laboratory infection studies, supporting their contributions to Lyme borreliae circulation. We also summarize the current understanding of divergent lizard immune responses that may explain the underlying molecular mechanisms to confer Lyme spirochete survival in vertebrate hosts. This review offers a critical perspective on potential enzootic cycles existing between lizard-tick-Borrelia interactions and highlights the importance of an eco-immunology lens for zoonotic pathogen transmission studies.
Collapse
Affiliation(s)
- Tristan A Nowak
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Biomedical Sciences, State University of New York at Albany, Albany, New York, USA
| | - Russell L Burke
- Department of Biology, Hofstra University, Hempstead, New York, USA
| | - Maria A Diuk-Wasser
- Department of Ecology, Evolution and Environmental Biology, Columbia University, New York, New York, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Biomedical Sciences, State University of New York at Albany, Albany, New York, USA
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| |
Collapse
|
19
|
Raghunandanan S, Priya R, Alanazi F, Lybecker MC, Schlax P, Yang X. A Fur family protein BosR is a novel RNA-binding protein that controls rpoS RNA stability in the Lyme disease pathogen. Nucleic Acids Res 2024; 52:5320-5335. [PMID: 38366569 PMCID: PMC11109971 DOI: 10.1093/nar/gkae114] [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: 10/13/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
The σ54-σS sigma factor cascade plays a central role in regulating differential gene expression during the enzootic cycle of Borreliella burgdorferi, the Lyme disease pathogen. In this pathway, the primary transcription of rpoS (which encodes σS) is under the control of σ54 which is activated by a bacterial enhancer-binding protein (EBP), Rrp2. The σ54-dependent activation in B. burgdorferi has long been thought to be unique, requiring an additional factor, BosR, a homologue of classical Fur/PerR repressor/activator. However, how BosR is involved in this σ54-dependent activation remains unclear and perplexing. In this study, we demonstrate that BosR does not function as a regulator for rpoS transcriptional activation. Instead, it functions as a novel RNA-binding protein that governs the turnover rate of rpoS mRNA. We further show that BosR directly binds to the 5' untranslated region (UTR) of rpoS mRNA, and the binding region overlaps with a region required for rpoS mRNA degradation. Mutations within this 5'UTR region result in BosR-independent RpoS production. Collectively, these results uncover a novel role of Fur/PerR family regulators as RNA-binding proteins and redefine the paradigm of the σ54-σS pathway in B. burgdorferi.
Collapse
Affiliation(s)
- Sajith Raghunandanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Fuad Alanazi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Meghan C Lybecker
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Center for Disease Control and Prevention, Fort Collins, CO, USA
| | - Paula Jean Schlax
- Department of Chemistry and Biochemistry, Bates College, Lewiston, ME, USA
| | - X Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
20
|
Bourgeois JS, McCarthy JE, Turk SP, Bernard Q, Clendenen LH, Wormser GP, Marcos LA, Dardick K, Telford SR, Marques AR, Hu LT. Peromyscus leucopus , Mus musculus , and humans have distinct transcriptomic responses to larval Ixodes scapularis bites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592193. [PMID: 38746284 PMCID: PMC11092580 DOI: 10.1101/2024.05.02.592193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Ixodes scapularis ticks are an important vector for at least six tick-borne human pathogens, including the predominant North American Lyme disease spirochete Borrelia burgdorferi . The ability for these ticks to survive in nature is credited, in part, to their ability to feed on a variety of hosts without excessive activation of the proinflammatory branch of the vertebrate immune system. While the ability for nymphal ticks to feed on a variety of hosts has been well-documented, the host-parasite interactions between larval I. scapularis and different vertebrate hosts is relatively unexplored. Here we report on the changes in the vertebrate transcriptome present at the larval tick bite site using the natural I. scapularis host Peromyscus leucopus deermouse, a non-natural rodent host Mus musculus (BALB/c), and humans. We note substantially less evidence of activation of canonical proinflammatory pathways in P. leucopus compared to BALB/c mice and pronounced evidence of inflammation in humans. Pathway enrichment analyses revealed a particularly strong signature of interferon gamma, tumor necrosis factor, and interleukin 1 signaling at the BALB/c and human tick bite site. We also note that bite sites on BALB/c mice and humans, but not deermice, show activation of wound-healing pathways. These data provide molecular evidence of the coevolution between larval I. scapularis and P. leucopus as well as expand our overall understanding of I. scapularis feeding. Significance Ixodes scapularis tick bites expose humans to numerous diseases in North America. While larval tick feeding enables pathogens to enter the tick population and eventually spread to humans, how larval ticks interact with mammals has been understudied compared to other tick stages. Here we examined the transcriptomic response of a natural I. scapularis rodent host ( Peromyscus leucopus ), a non-native I. scapularis rodent host ( Mus musculus ), and an incidental host (humans). We find that there are differences in how all three species respond to larval I. scapularis , with the natural host producing the smallest transcriptomic signature of a canonical proinflammatory immune response and the incidental human host producing the most robust signature of inflammation in response to the larval tick. These data expand our understanding of the pressures on ticks in the wild and inform our ability to model these interactions in laboratory settings.
Collapse
|
21
|
Wülbern J, Windorfer L, Sato K, Nakao M, Hepner S, Margos G, Fingerle V, Kawabata H, Becker NS, Kraiczy P, Rollins RE. Unprecedented genetic variability of PFam54 paralogs among Eurasian Lyme borreliosis-causing spirochetes. Ecol Evol 2024; 14:e11397. [PMID: 38779535 PMCID: PMC11109050 DOI: 10.1002/ece3.11397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 04/17/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Lyme borreliosis (LB) is the most common vector-borne disease in the Northern Hemisphere caused by spirochetes belonging to the Borrelia burgdorferi sensu lato (Bbsl) complex. Borrelia spirochetes circulate in obligatory transmission cycles between tick vectors and different vertebrate hosts. To successfully complete this complex transmission cycle, Bbsl encodes for an arsenal of proteins including the PFam54 protein family with known, or proposed, influences to reservoir host and/or vector adaptation. Even so, only fragmentary information is available regarding the naturally occurring level of variation in the PFam54 gene array especially in relation to Eurasian-distributed species. Utilizing whole genome data from isolates (n = 141) originated from three major LB-causing Borrelia species across Eurasia (B. afzelii, B. bavariensis, and B. garinii), we aimed to characterize the diversity of the PFam54 gene array in these isolates to facilitate understanding the evolution of PFam54 paralogs on an intra- and interspecies level. We found an extraordinarily high level of variation in the PFam54 gene array with 39 PFam54 paralogs belonging to 23 orthologous groups including five novel paralogs. Even so, the gene array appears to have remained fairly stable over the evolutionary history of the studied Borrelia species. Interestingly, genes outside Clade IV, which contains genes encoding for proteins associated with Borrelia pathogenesis, more frequently displayed signatures of diversifying selection between clades that differ in hypothesized vector or host species. This could suggest that non-Clade IV paralogs play a more important role in host and/or vector adaptation than previously expected, which would require future lab-based studies to validate.
Collapse
Affiliation(s)
- Janna Wülbern
- Evolutionary Ecology and Genetics, Faculty of BiologyChristian‐Albrechts‐Universität Zu KielKielGermany
| | - Laura Windorfer
- Terrestrial Ecology Research Group, Department of Life Science Systems, School of Life SciencesTechnical University of MunichFreisingGermany
- Disrupt.Design Lab, Faculty of Architecture and Town Planning, Segoe BuildingTechnion – Israel Institute of TechnologyTechnion CityIsrael
| | - Kozue Sato
- Department of Bacteriology INational Institute for Infectious DiseaseTokyoJapan
| | - Minoru Nakao
- Department of ParasitologyAsahikawa Medical UniversityAsahikawaJapan
| | - Sabrina Hepner
- German National Reference Center for Borrelia, Bavarian Health and Food Safety AuthorityOberschleissheimGermany
| | - Gabriele Margos
- German National Reference Center for Borrelia, Bavarian Health and Food Safety AuthorityOberschleissheimGermany
| | - Volker Fingerle
- German National Reference Center for Borrelia, Bavarian Health and Food Safety AuthorityOberschleissheimGermany
| | - Hiroki Kawabata
- Department of Bacteriology INational Institute for Infectious DiseaseTokyoJapan
| | - Noémie S. Becker
- Division of Evolutionary Biology, Faculty of BiologyLMU MunichPlanegg‐MartinsriedGermany
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital of FrankfurtGoethe University FrankfurtFrankfurtGermany
| | - Robert E. Rollins
- Institute of Avian Research “Vogelwarte Helgoland”WilhelmshavenGermany
| |
Collapse
|
22
|
Gunčar G, Kukar M, Smole T, Moškon S, Vovko T, Podnar S, Černelč P, Brvar M, Notar M, Köster M, Jelenc MT, Osterc Ž, Notar M. Differentiating viral and bacterial infections: A machine learning model based on routine blood test values. Heliyon 2024; 10:e29372. [PMID: 38644832 PMCID: PMC11033127 DOI: 10.1016/j.heliyon.2024.e29372] [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: 09/18/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/23/2024] Open
Abstract
The growing threat of antibiotic resistance necessitates accurate differentiation between bacterial and viral infections for proper antibiotic administration. In this study, a Virus vs. Bacteria machine learning model was developed to distinguish between these infection types using 16 routine blood test results, C-reactive protein concentration (CRP), biological sex, and age. With a dataset of 44,120 cases from a single medical center, the model achieved an accuracy of 82.2 %, a sensitivity of 79.7 %, a specificity of 84.5 %, a Brier score of 0.129, and an area under the ROC curve (AUC) of 0.905, outperforming a CRP-based decision rule. Notably, the machine learning model enhanced accuracy within the CRP range of 10-40 mg/L, a range where CRP alone is less informative. These results highlight the advantage of integrating multiple blood parameters in diagnostics. The "Virus vs. Bacteria" model paves the way for advanced diagnostic tools, leveraging machine learning to optimize infection management.
Collapse
Affiliation(s)
- Gregor Gunčar
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Slovenia
| | - Matjaž Kukar
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
- Faculty of Computer and Information Science, University of Ljubljana, Slovenia
| | - Tim Smole
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
| | - Sašo Moškon
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
| | - Tomaž Vovko
- Department of Infectious Diseases, University Medical Centre Ljubljana, Slovenia
| | - Simon Podnar
- Division of Neurology, University Medical Centre Ljubljana, Slovenia
| | - Peter Černelč
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
| | - Miran Brvar
- Centre for Clinical Toxicology and Pharmacology, University Medical Centre Ljubljana, Slovenia
| | - Mateja Notar
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
| | - Manca Köster
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
| | | | - Žiga Osterc
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
| | - Marko Notar
- Smart Blood Analytics Swiss SA, CH-8008, Zürich, Switzerland
| |
Collapse
|
23
|
Khogali R, Bastos A, Bargul JL, Getange D, Kabii J, Masiga D, Villinger J. Tissue-specific localization of tick-borne pathogens in ticks collected from camels in Kenya: insights into vector competence. Front Cell Infect Microbiol 2024; 14:1382228. [PMID: 38698904 PMCID: PMC11063324 DOI: 10.3389/fcimb.2024.1382228] [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: 02/05/2024] [Accepted: 03/15/2024] [Indexed: 05/05/2024] Open
Abstract
Background Tick-borne pathogen (TBP) surveillance studies often use whole-tick homogenates when inferring tick-pathogen associations. However, localized TBP infections within tick tissues (saliva, hemolymph, salivary glands, and midgut) can inform pathogen transmission mechanisms and are key to disentangling pathogen detection from vector competence. Methods We screened 278 camel blood samples and 504 tick tissue samples derived from 126 camel ticks sampled in two Kenyan counties (Laikipia and Marsabit) for Anaplasma, Ehrlichia, Coxiella, Rickettsia, Theileria, and Babesia by PCR-HRM analysis. Results Candidatus Anaplasma camelii infections were common in camels (91%), but absent in all samples from Rhipicephalus pulchellus, Amblyomma gemma, Hyalomma dromedarii, and Hyalomma rufipes ticks. We detected Ehrlichia ruminantium in all tissues of the four tick species, but Rickettsia aeschlimannii was only found in Hy. rufipes (all tissues). Rickettsia africae was highest in Am. gemma (62.5%), mainly in the hemolymph (45%) and less frequently in the midgut (27.5%) and lowest in Rh. pulchellus (29.4%), where midgut and hemolymph detection rates were 17.6% and 11.8%, respectively. Similarly, in Hy. dromedarii, R. africae was mainly detected in the midgut (41.7%) but was absent in the hemolymph. Rickettsia africae was not detected in Hy. rufipes. No Coxiella, Theileria, or Babesia spp. were detected in this study. Conclusions The tissue-specific localization of R. africae, found mainly in the hemolymph of Am. gemma, is congruent with the role of this tick species as its transmission vector. Thus, occurrence of TBPs in the hemolymph could serve as a predictor of vector competence of TBP transmission, especially in comparison to detection rates in the midgut, from which they must cross tissue barriers to effectively replicate and disseminate across tick tissues. Further studies should focus on exploring the distribution of TBPs within tick tissues to enhance knowledge of TBP epidemiology and to distinguish competent vectors from dead-end hosts.
Collapse
Affiliation(s)
- Rua Khogali
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
- Department of Parasitology, Faculty of Veterinary Medicine, University of Khartoum, Khartoum North, Sudan
| | - Armanda Bastos
- Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
- Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
| | - Joel L. Bargul
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya
| | - Dennis Getange
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - James Kabii
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Daniel Masiga
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Jandouwe Villinger
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| |
Collapse
|
24
|
Dulipati V, Kotimaa J, Rezola M, Kontiainen M, Jarva H, Nyman D, Meri S. Antibody responses to immunoevasion proteins BBK32 and OspE constitute part of the serological footprint in neuroborreliosis but are insufficient to prevent the disease. Scand J Immunol 2024; 99:e13353. [PMID: 39007994 DOI: 10.1111/sji.13353] [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: 07/01/2023] [Revised: 11/14/2023] [Accepted: 12/21/2023] [Indexed: 07/16/2024]
Abstract
Lyme borreliosis, caused by Borrelia burgdorferi sensu lato, is the most common tickborne disease. Its neuronal form, neuroborreliosis, comprises 3 to 38% of borreliosis cases in Europe. Borrelia outer surface proteins and virulence factors, OspE and BBK32, have been previously reported to help cause infection by promoting attachment to human host epithelial cells and evading complement attack. We assessed the serological responses to BBK32 and OspE in 19 individuals diagnosed with neuroborreliosis to see whether antibodies that could both target the bacteria and neutralize the virulence mechanisms on the microbial surface emerge. Results evaluate levels of total protein, IgG and the chemokine CXCL13, a determinant for B-cell recruitment during neuroinflammation, in patients' cerebrospinal fluid samples. Antibody levels against BBK32 and OspE correlated with those against VlsE, a well-characterized diagnostic serological marker of the disease. A dual serological profile of the patients was observed. K-means clustering split the cohort into two discrete groups presenting distinct serological and CNS responses. One group contained young patients with low levels of anti-BBK32 and OspE antibodies. The other group showed stronger responses, possibly following prolonged infections or reinfections. Additionally, we assessed anti-ganglioside antibodies that could cause autoimmunity or complement dysregulation but observed that they did not correlate with neuroborreliosis in our patient cohort. The dual nature of antibody responses against the virulence factors BBK32 and OspE in neuroborreliosis patients may suggest the necessity of repeated exposures for efficient immune responses. Better protection could be achieved if the virulence factors were formulated into vaccines.
Collapse
Affiliation(s)
- Vinaya Dulipati
- Translational Immunology Research Program, Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juha Kotimaa
- Translational Immunology Research Program, Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- VTT Technical Research Center of Finland, Espoo, Finland
| | - Mikel Rezola
- Translational Immunology Research Program, Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, Inflammation, Complement and Cancer team, Paris, France
| | - Mikko Kontiainen
- Translational Immunology Research Program, Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hanna Jarva
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Dag Nyman
- Åland Group for Borreliosis Research, Mariehamn, Finland
| | - Seppo Meri
- Translational Immunology Research Program, Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
25
|
Zhang JJ, Raghunandanan S, Wang Q, Priya R, Alanazi F, Lou Y, Yang XF. BadR directly represses the expression of the glycerol utilization operon in the Lyme disease pathogen. J Bacteriol 2024; 206:e0034023. [PMID: 38214528 PMCID: PMC10882987 DOI: 10.1128/jb.00340-23] [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: 10/16/2023] [Accepted: 12/14/2023] [Indexed: 01/13/2024] Open
Abstract
Glycerol utilization as a carbohydrate source by Borreliella burgdorferi, the Lyme disease spirochete, is critical for its successful colonization and persistence in the tick vector. The expression of the glpFKD (glp) operon, which encodes proteins for glycerol uptake/utilization, must be tightly regulated during the enzootic cycle of B. burgdorferi. Previous studies have established that the second messenger cyclic di-GMP (c-di-GMP) is required for the activation of glp expression, while an alternative sigma factor RpoS acts as a negative regulator for glp expression. In the present study, we report identification of a cis element within the 5´ untranslated region of glp that exerts negative regulation of glp expression. Further genetic screen of known and predicted DNA-binding proteins encoded in the genome of B. burgdorferi uncovered that overexpressing Borrelia host adaptation regulator (BadR), a known global regulator, dramatically reduced glp expression. Similarly, the badR mutant significantly increased glp expression. Subsequent electrophoretic mobility shift assay analyses demonstrated that BadR directly binds to this cis element, thereby repressing glp independent of RpoS-mediated repression. The efficiency of BadR binding was further assessed in the presence of c-di-GMP and various carbohydrates. This finding highlights multi-layered positive and negative regulatory mechanisms employed by B. burgdorferi to synchronize glp expression throughout its enzootic cycle.IMPORTANCEBorreliella burgdorferi, the Lyme disease pathogen, must modulate its gene expression differentially to adapt successfully to its two disparate hosts. Previous studies have demonstrated that the glycerol uptake and utilization operon, glpFKD, plays a crucial role in spirochetal survival within ticks. However, the glpFKD expression must be repressed when B. burgdorferi transitions to the mammalian host. In this study, we identified a specific cis element responsible for the repression of glpFKD. We further pinpointed Borrelia host adaptation regulator as the direct binding protein to this cis element, thereby repressing glpFKD expression. This discovery paves the way for a deeper exploration of how zoonotic pathogens sense distinct hosts and switch their carbon source utilization during transmission.
Collapse
Affiliation(s)
- Jun-Jie Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sajith Raghunandanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Qian Wang
- Department of Clinical Laboratory, Wenzhou People’s Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Fuad Alanazi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Yongliang Lou
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
26
|
Sloupenska K, Koubkova B, Horak P, Dolezilkova J, Hutyrova B, Racansky M, Miklusova M, Mares J, Raska M, Krupka M. Antigenicity and immunogenicity of different morphological forms of Borrelia burgdorferi sensu lato spirochetes. Sci Rep 2024; 14:4014. [PMID: 38369537 PMCID: PMC10874929 DOI: 10.1038/s41598-024-54505-y] [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: 10/02/2023] [Accepted: 02/13/2024] [Indexed: 02/20/2024] Open
Abstract
Borrelia burgdorferi sensu lato is a species complex of pleomorphic spirochetes, including species that cause Lyme disease (LD) in humans. In addition to classic spiral forms, these bacteria are capable of creating morphological forms referred to as round bodies and aggregates. The subject of discussion is their possible contribution to the persistence of infection or post-infection symptoms in LD. This study investigates the immunological properties of these forms by monitoring reactivity with early (n = 30) and late stage (n = 30) LD patient sera and evaluating the immune response induced by vaccination of mice. In patient sera, we found a quantitative difference in reactivity with individual morphotypes, when aggregates were recognized most intensively, but the difference was statistically significant in only half of the tested strains. In post-vaccination mouse sera, we observed a statistically significant higher reactivity with antigens p83 and p25 (OspC) in mice vaccinated with aggregates compared to mice vaccinated with spiral forms. The importance of the particulate nature of the antigen for the induction of a Th1-directed response has also been demonstrated. In any of morphological forms, the possibility of inducing antibodies cross-reacting with human nuclear and myositis specific/associated autoantigens was not confirmed by vaccination of mice.
Collapse
Affiliation(s)
- Kristyna Sloupenska
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00, Olomouc, Czech Republic
| | - Barbora Koubkova
- Department of Allergology and Clinical Immunology, University Hospital Olomouc, Zdravotniku 248/7, 779 00, Olomouc, Czech Republic
| | - Pavel Horak
- Third Department of Internal Medicine-Nephrology, Rheumatology and Endocrinology, University Hospital Olomouc, Zdravotniku 248/7, 779 00, Olomouc, Czech Republic
- Third Department of Internal Medicine-Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00, Olomouc, Czech Republic
| | - Jana Dolezilkova
- Laboratory of Medical Parasitology and Zoology, Public Health Institute Ostrava, Partyzanske Namesti 2633/7, Moravska Ostrava, 702 00, Ostrava, Czech Republic
| | - Beata Hutyrova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00, Olomouc, Czech Republic
- Department of Allergology and Clinical Immunology, University Hospital Olomouc, Zdravotniku 248/7, 779 00, Olomouc, Czech Republic
| | - Mojmir Racansky
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00, Olomouc, Czech Republic
- Department of Allergology and Clinical Immunology, University Hospital Olomouc, Zdravotniku 248/7, 779 00, Olomouc, Czech Republic
| | - Martina Miklusova
- Department of Neurology, University Hospital Olomouc, Zdravotniku 248/7, 779 00, Olomouc, Czech Republic
| | - Jan Mares
- Department of Neurology, University Hospital Olomouc, Zdravotniku 248/7, 779 00, Olomouc, Czech Republic
- Department of Neurology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00, Olomouc, Czech Republic
| | - Milan Raska
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00, Olomouc, Czech Republic
- Department of Immunology, University Hospital Olomouc, Zdravotniku 248/7, 779 00, Olomouc, Czech Republic
| | - Michal Krupka
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 779 00, Olomouc, Czech Republic.
| |
Collapse
|
27
|
Chen YL, Lee J, Liu Z, Strych U, Bottazzi ME, Lin YP, Chen WH. Biophysical and biochemical characterization of a recombinant Lyme disease vaccine antigen, CspZ-YA. Int J Biol Macromol 2024; 259:129295. [PMID: 38211914 DOI: 10.1016/j.ijbiomac.2024.129295] [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: 10/19/2023] [Revised: 12/19/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
Lyme disease, caused by Lyme Borrelia spirochetes, is the most common vector-borne illness in the United States. Despite its global significance, with an estimated 14.5 % seroprevalence, there is currently no licensed vaccine. Previously, we demonstrated that CspZ-YA protein conferred protection against Lyme Borrelia infection, making it a promising vaccine candidate. However, such a protein was tagged with hexahistidine, and thus not preferred for vaccine development; furthermore, the formulation to stabilize the protein was understudied. In this work, we developed a two-step purification process for tag-free E. coli-expressed recombinant CspZ-YA. We further utilized various bioassays to analyze the protein and determine the suitable buffer system for long-term storage and formulation as a vaccine immunogen. The results indicated that a buffer with a pH between 6.5 and 8.5 stabilized CspZ-YA by reducing its surface hydrophobicity and colloidal interactions. Additionally, low pH values induced a change in local spatial conformation and resulted in a decrease in α-helix content. Lastly, an optimal salinity of 22-400 mM at pH 7.5 was found to be important for its stability. Collectively, this study provides a fundamental biochemical and biophysical understanding and insights into the ideal stabilizing conditions to produce CspZ-YA recombinant protein for use in vaccine formulation and development.
Collapse
Affiliation(s)
- Yi-Lin Chen
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Jungsoon Lee
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Zhuyun Liu
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Ulrich Strych
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Maria Elena Bottazzi
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Biology, Baylor University, Waco, TX, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA; Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA.
| | - Wen-Hsiang Chen
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA.
| |
Collapse
|
28
|
Yaş OB, Coleman AS, Lipman RM, Sharma K, Raghunandanan S, Alanazi F, Rana VS, Kitsou C, Yang X, Pal U. A systemic approach to identify non-abundant immunogenic proteins in Lyme disease pathogens. mSystems 2024; 9:e0108723. [PMID: 38078774 PMCID: PMC10805064 DOI: 10.1128/msystems.01087-23] [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: 10/09/2023] [Accepted: 11/06/2023] [Indexed: 01/24/2024] Open
Abstract
Borrelia burgdorferi, the pathogen of Lyme disease, differentially produces many outer surface proteins (Osp), some of which represent the most abundant membrane proteins, such as OspA, OspB, and OspC. In cultured bacteria, these proteins can account for a substantial fraction of the total cellular or membrane proteins, posing challenges to the identification and analysis of non-abundant proteins, which could serve as novel pathogen detection markers or as vaccine candidates. Herein, we introduced serial mutations to remove these abundant Osps and generated a B. burgdorferi mutant deficient in OspA, OspB, and OspC in an infectious 297-isolate background, designated as OspABC- mutant. Compared to parental isolate, the mutant did not reflect growth defects in the cultured medium but showed differential mRNA expression of representative tested genes, in addition to gross changes in cellular and membrane protein profiles. The analysis of differentially detectable protein contents of the OspABC- mutant, as compared to the wild type, by two-dimensional gel electrophoresis followed by liquid chromatography-mass spectrometry, identified several spirochete proteins that are dominated by proteins of unknown functions, as well as membrane transporters, chaperons, and metabolic enzymes. We produced recombinant forms of two of these represented proteins, BBA34 and BB0238, and showed that these proteins are detectable during spirochete infection in the tick-borne murine model of Lyme borreliosis and thus serve as potential antigenic markers of the infection.IMPORTANCEThe present manuscript employed a systemic approach to identify non-abundant proteins in cultured Borrelia burgdorferi that are otherwise masked or hidden due to the overwhelming presence of abundant Osps like OspA, OspB, and OspC. As these Osps are either absent or transiently expressed in mammals, we performed a proof-of-concept study in which their removal allowed the analysis of otherwise less abundant antigens in OspABC-deficient mutants and identified several immunogenic proteins, including BBA34 and BB0238. These antigens could serve as novel vaccine candidates and/or genetic markers of Lyme borreliosis, promoting new research in the clinical diagnosis and prevention of Lyme disease.
Collapse
Affiliation(s)
- Ozlem Buyuktanir Yaş
- Department of Microbiology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Adam S. Coleman
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Rachel M. Lipman
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Kavita Sharma
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Sajith Raghunandanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Fuad Alanazi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Vipin S. Rana
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Xiuli Yang
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
- Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
29
|
Zamba-Campero M, Soliman D, Yu H, Lasseter AG, Chang YY, Liu J, Aravind L, Jewett MW, Storz G, Adams PP. Broadly conserved FlgV controls flagellar assembly and Borrelia burgdorferi dissemination in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574855. [PMID: 38260563 PMCID: PMC10802407 DOI: 10.1101/2024.01.09.574855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Flagella propel pathogens through their environments yet are expensive to synthesize and are immunogenic. Thus, complex hierarchical regulatory networks control flagellar gene expression. Spirochetes are highly motile bacteria, but peculiarly in the Lyme spirochete Borrelia burgdorferi, the archetypal flagellar regulator σ28 is absent. We rediscovered gene bb0268 in B. burgdorferi as flgV, a broadly-conserved gene in the flagellar superoperon alongside σ28 in many Spirochaetes, Firmicutes and other phyla, with distant homologs in Epsilonproteobacteria. We found that B. burgdorferi FlgV is localized within flagellar motors. B. burgdorferi lacking flgV construct fewer and shorter flagellar filaments and are defective in cell division and motility. During the enzootic cycle, B. burgdorferi lacking flgV survive and replicate in Ixodes ticks but are attenuated for dissemination and infection in mice. Our work defines infection timepoints when spirochete motility is most crucial and implicates FlgV as a broadly distributed structural flagellar component that modulates flagellar assembly.
Collapse
Affiliation(s)
- Maxime Zamba-Campero
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Soliman
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaxin Yu
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06536, USA
- Microbial Sciences Institute, Yale University, West Haven, CT 06516, USA
| | - Amanda G. Lasseter
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, 32827, USA
| | - Yuen-Yan Chang
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun Liu
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06536, USA
- Microbial Sciences Institute, Yale University, West Haven, CT 06516, USA
| | - L. Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Mollie W. Jewett
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, 32827, USA
| | - Gisela Storz
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philip P. Adams
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, 32827, USA
- Postdoctoral Research Associate Program, National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD 20892, USA
- Independent Research Scholar Program, Intramural Research Program, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
30
|
Brangulis K, Akopjana I, Bogans J, Kazaks A, Tars K. Structural studies of chromosomally encoded outer surface lipoprotein BB0158 from Borrelia burgdorferi sensu stricto. Ticks Tick Borne Dis 2024; 15:102287. [PMID: 38016210 DOI: 10.1016/j.ttbdis.2023.102287] [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: 09/24/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
Lyme disease, or also known as Lyme borreliosis, is caused by the spirochetes belonging to the Borrelia burgdorferi sensu lato complex, which can enter the human body following the bite of an infected tick. Many membrane lipid-bound proteins, also known as lipoproteins, are located on the surface of B. burgdorferi sensu lato and play a crucial role in the spirochete to interact with its environment, whether in ticks or mammals. Since the spirochete needs to perform various tasks, such as resisting the host's immune system or spreading throughout the organism, it is not surprising that numerous surface proteins have been found to be essential for B. burgdorferi sensu lato complex bacteria in causing Lyme disease. In this study, we have determined (at 2.4 Å resolution) and characterized the 3D structure of BB0158, one of the few chromosomally encoded outer surface proteins from B. burgdorferi sensu stricto. BB0158 belongs to the paralogous gene family 44 (PFam44), consisting of four other members (BB0159, BBA04, BBE09 and BBK52). The characterization of BB0158, which appears to form a domain-swapped dimer, in conjunction with the characterization of the corresponding PFam44 members, certainly contribute to our understanding of B. burgdorferi sensu stricto proteins.
Collapse
Affiliation(s)
- Kalvis Brangulis
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia.
| | - Inara Akopjana
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| | - Janis Bogans
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| | - Kaspars Tars
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| |
Collapse
|
31
|
Brangulis K, Drunka L, Akopjana I, Tars K. Structure of the Borrelia burgdorferi ATP-dependent metalloprotease FtsH in its functionally relevant hexameric form. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1872:140969. [PMID: 37852516 DOI: 10.1016/j.bbapap.2023.140969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/20/2023]
Abstract
ATP-dependent proteases FtsH are conserved in bacteria, mitochondria, and chloroplasts, where they play an essential role in degradation of misfolded/unneeded membrane and cytosolic proteins. It has also been demonstrated that the FtsH homologous protein BB0789 is crucial for mouse and tick infectivity and in vitro growth of the Lyme disease-causing agent Borrelia burgdorferi. This is not surprising, considering B. burgdorferi complex life cycle, residing in both in mammals and ticks, which requires a wide range of membrane proteins and short-lived cytosolic regulatory proteins to invade and persist in the host organism. In the current study, we have solved the crystal structure of the cytosolic BB0789166-614, lacking both N-terminal transmembrane α-helices and the small periplasmic domain. The structure revealed the arrangement of the AAA+ ATPase and the zinc-dependent metalloprotease domains in a hexamer ring, which is essential for ATPase and proteolytic activity. The AAA+ domain was found in an ADP-bound state, while the protease domain showed coordination of a zinc ion by two histidine residues and one aspartic acid residue. The loop region that forms the central pore in the oligomer was poorly defined in the crystal structure and therefore predicted by AlphaFold to complement the missing structural details, providing a complete picture of the functionally relevant hexameric form of BB0789. We confirmed that BB0789 is functionally active, possessing both protease and ATPase activities, thus providing novel structural-functional insights into the protein, which is known to be absolutely necessary for B. burgdorferi to survive and cause Lyme disease.
Collapse
Affiliation(s)
- Kalvis Brangulis
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, LV-1067 Riga, Latvia; Department of Human Physiology and Biochemistry, Riga Stradins University, Dzirciema 16, LV-1007 Riga, Latvia.
| | - Laura Drunka
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, LV-1067 Riga, Latvia
| | - Inara Akopjana
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, LV-1067 Riga, Latvia
| | - Kaspars Tars
- Latvian Biomedical Research and Study Centre, Ratsupites 1 k-1, LV-1067 Riga, Latvia
| |
Collapse
|
32
|
Kumaresan V, Ingle TM, Kilgore N, Zhang G, Hermann BP, Seshu J. Cellular and transcriptome signatures unveiled by single-cell RNA-Seq following ex vivo infection of murine splenocytes with Borrelia burgdorferi. Front Immunol 2023; 14:1296580. [PMID: 38149246 PMCID: PMC10749944 DOI: 10.3389/fimmu.2023.1296580] [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: 09/18/2023] [Accepted: 11/06/2023] [Indexed: 12/28/2023] Open
Abstract
Introduction Lyme disease, the most common tick-borne infectious disease in the US, is caused by a spirochetal pathogen Borrelia burgdorferi (Bb). Distinct host responses are observed in susceptible and resistant strains of inbred of mice following infection with Bb reflecting a subset of inflammatory responses observed in human Lyme disease. The advent of post-genomic methodologies and genomic data sets enables dissecting the host responses to advance therapeutic options for limiting the pathogen transmission and/or treatment of Lyme disease. Methods In this study, we used single-cell RNA-Seq analysis in conjunction with mouse genomics exploiting GFP-expressing Bb to sort GFP+ splenocytes and GFP- bystander cells to uncover novel molecular and cellular signatures that contribute to early stages of immune responses against Bb. Results These data decoded the heterogeneity of splenic neutrophils, macrophages, NK cells, B cells, and T cells in C3H/HeN mice in response to Bb infection. Increased mRNA abundance of apoptosis-related genes was observed in neutrophils and macrophages clustered from GFP+ splenocytes. Moreover, complement-mediated phagocytosis-related genes such as C1q and Ficolin were elevated in an inflammatory macrophage subset, suggesting upregulation of these genes during the interaction of macrophages with Bb-infected neutrophils. In addition, the role of DUSP1 in regulating the expression of Casp3 and pro-inflammatory cytokines Cxcl1, Cxcl2, Il1b, and Ccl5 in Bb-infected neutrophils were identified. Discussion These findings serve as a growing catalog of cell phenotypes/biomarkers among murine splenocytes that can be exploited for limiting spirochetal burden to limit the transmission of the agent of Lyme disease to humans via reservoir hosts.
Collapse
Affiliation(s)
- Venkatesh Kumaresan
- Department of Molecular Microbiology and Immunology, The University of Texas at San Antonio, San Antonio, TX, United States
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Taylor MacMackin Ingle
- Department of Molecular Microbiology and Immunology, The University of Texas at San Antonio, San Antonio, TX, United States
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Nathan Kilgore
- Department of Molecular Microbiology and Immunology, The University of Texas at San Antonio, San Antonio, TX, United States
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Guoquan Zhang
- Department of Molecular Microbiology and Immunology, The University of Texas at San Antonio, San Antonio, TX, United States
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Brian P. Hermann
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Janakiram Seshu
- Department of Molecular Microbiology and Immunology, The University of Texas at San Antonio, San Antonio, TX, United States
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
33
|
Strnad M, Rudenko N, Rego RO. Pathogenicity and virulence of Borrelia burgdorferi. Virulence 2023; 14:2265015. [PMID: 37814488 PMCID: PMC10566445 DOI: 10.1080/21505594.2023.2265015] [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: 11/09/2022] [Accepted: 09/25/2023] [Indexed: 10/11/2023] Open
Abstract
Infection with Borrelia burgdorferi often triggers pathophysiologic perturbations that are further augmented by the inflammatory responses of the host, resulting in the severe clinical conditions of Lyme disease. While our apprehension of the spatial and temporal integration of the virulence determinants during the enzootic cycle of B. burgdorferi is constantly being improved, there is still much to be discovered. Many of the novel virulence strategies discussed in this review are undetermined. Lyme disease spirochaetes must surmount numerous molecular and mechanical obstacles in order to establish a disseminated infection in a vertebrate host. These barriers include borrelial relocation from the midgut of the feeding tick to its body cavity and further to the salivary glands, deposition to the skin, haematogenous dissemination, extravasation from blood circulation system, evasion of the host immune responses, localization to protective niches, and establishment of local as well as distal infection in multiple tissues and organs. Here, the various well-defined but also possible novel strategies and virulence mechanisms used by B. burgdorferi to evade obstacles laid out by the tick vector and usually the mammalian host during colonization and infection are reviewed.
Collapse
Affiliation(s)
- Martin Strnad
- Biology Centre CAS, Institute of Parasitology, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská, Czech Republic
| | - Natalie Rudenko
- Biology Centre CAS, Institute of Parasitology, České Budějovice, Czech Republic
| | - Ryan O.M. Rego
- Biology Centre CAS, Institute of Parasitology, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská, Czech Republic
| |
Collapse
|
34
|
Helble JD, Walsh MJ, McCarthy JE, Smith NP, Tirard AJ, Arnold BY, Villani AC, Hu LT. Single-cell RNA sequencing of murine ankle joints over time reveals distinct transcriptional changes following Borrelia burgdorferi infection. iScience 2023; 26:108217. [PMID: 37953958 PMCID: PMC10632114 DOI: 10.1016/j.isci.2023.108217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/06/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
Lyme disease is caused by the bacterial pathogen Borrelia burgdorferi, which can be readily modeled in laboratory mice. In order to understand the cellular and transcriptional changes that occur during B. burgdorferi infection, we conducted single-cell RNA sequencing (scRNA-seq) of ankle joints of infected C57BL/6 mice over time. We found that macrophages/monocytes, T cells, synoviocytes and fibroblasts all showed significant differences in gene expression of both inflammatory and non-inflammatory genes that peaked early and returned to baseline before the typical resolution of arthritis. Predictions of cellular interactions showed that macrophages appear to communicate extensively between different clusters of macrophages as well as with fibroblasts and synoviocytes. Our data give unique insights into the interactions between B. burgdorferi and the murine immune system over time and allow for a better understanding of mechanisms by which the dysregulation of the immune response may lead to prolonged symptoms in some patients.
Collapse
Affiliation(s)
- Jennifer D. Helble
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Michael J. Walsh
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Julie E. McCarthy
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Neal P. Smith
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alice J. Tirard
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Benjamin Y. Arnold
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Linden T. Hu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
35
|
Alanazi F, Raghunandanan S, Priya R, Yang XF. The Rrp2-RpoN-RpoS pathway plays an important role in the blood-brain barrier transmigration of the Lyme disease pathogen. Infect Immun 2023; 91:e0022723. [PMID: 37874144 PMCID: PMC10652863 DOI: 10.1128/iai.00227-23] [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: 06/13/2023] [Accepted: 09/14/2023] [Indexed: 10/25/2023] Open
Abstract
Lyme disease, caused by Borrelia (or Borreliella) burgdorferi, is a complex multisystemic disorder that includes Lyme neuroborreliosis resulting from the invasion of both the central and peripheral nervous systems. However, factors that enable the pathogen to cross the blood-brain barrier (BBB) and invade the central nervous system (CNS) are still not well understood. The objective of this study was to identify the B. burgdorferi factors required for BBB transmigration. We utilized a transwell BBB model based on human brain-microvascular endothelial cells and focused on investigating the Rrp2-RpoN-RpoS pathway, a central regulatory pathway that is essential for mammalian infection by B. burgdorferi. Our results demonstrated that the Rrp2-RpoN-RpoS pathway is crucial for BBB transmigration. Furthermore, we identified OspC, a major surface lipoprotein controlled by the Rrp2-RpoN-RpoS pathway, as a significant contributor to BBB transmigration. Constitutive production of OspC in a mutant defective in the Rrp2-RpoN-RpoS pathway did not rescue the impairment in BBB transmigration, indicating that this pathway controls additional factors for this process. Two other major surface lipoproteins controlled by this pathway, DbpA/B and BBK32, appeared to be dispensable for BBB transmigration. In addition, both the surface lipoprotein OspA and the Rrp1 pathway, which are required B. burgdorferi colonization in the tick vector, were found not required for BBB transmigration. Collectively, our findings using in vitro transwell assays uncover another potential role of the Rrp2-RpoN-RpoS pathway in BBB transmigration of B. burgdorferi and invasion into the CNS.
Collapse
Affiliation(s)
- Fuad Alanazi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Sajith Raghunandanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
36
|
Kozelková T, Dyčka F, Lu S, Urbanová V, Frantová H, Sojka D, Šíma R, Horn M, Perner J, Kopáček P. Insight Into the Dynamics of the Ixodes ricinus Nymphal Midgut Proteome. Mol Cell Proteomics 2023; 22:100663. [PMID: 37832788 PMCID: PMC10665701 DOI: 10.1016/j.mcpro.2023.100663] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/06/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Ticks are ectoparasites that feed on blood and have an impressive ability to consume and process enormous amounts of host blood, allowing extremely long periods of starvation between blood meals. The central role in the parasitic lifestyle of ticks is played by the midgut. This organ efficiently stores and digests ingested blood and serves as the primary interface for the transmission of tick-borne pathogens. In this study, we used a label-free quantitative approach to perform a novel dynamic proteomic analysis of the midgut of Ixodesricinus nymphs, covering their development from unfed to pre-molt stages. We identified 1534 I. ricinus-specific proteins with a relatively low proportion of host proteins. This proteome dataset, which was carefully examined by manual scrutiny, allowed precise annotation of proteins important for blood meal processing and their dynamic changes during nymphal ontogeny. We focused on midgut molecules related to lipid hydrolysis, storage, and transport, opening a yet unexplored avenue for studying lipid metabolism in ticks. Further dynamic profiling of the tick's multi-enzyme digestive network, protease inhibitors, enzymes involved in redox homeostasis and detoxification, antimicrobial peptides, and proteins responsible for midgut colonization by Borrelia spirochetes promises to uncover new targets for targeting tick nymphs, the most critical life stage for transmission the pathogens that cause tick-borne diseases.
Collapse
Affiliation(s)
- Tereza Kozelková
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Faculty of Sciences, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Filip Dyčka
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Faculty of Sciences, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Stephen Lu
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Veronika Urbanová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Helena Frantová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Daniel Sojka
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Radek Šíma
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Bioptic Laboratory, Ltd, Plzen, Czech Republic
| | - Martin Horn
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Perner
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic.
| |
Collapse
|
37
|
Sze CW, Zhang K, Lynch MJ, Iyer R, Crane BR, Schwartz I, Li C. A chemosensory-like histidine kinase is dispensable for chemotaxis in vitro but regulates the virulence of Borrelia burgdorferi through modulating the stability of RpoS. PLoS Pathog 2023; 19:e1011752. [PMID: 38011206 PMCID: PMC10703414 DOI: 10.1371/journal.ppat.1011752] [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: 08/10/2023] [Revised: 12/07/2023] [Accepted: 10/14/2023] [Indexed: 11/29/2023] Open
Abstract
As an enzootic pathogen, the Lyme disease bacterium Borrelia burgdorferi possesses multiple copies of chemotaxis proteins, including two chemotaxis histidine kinases (CHK), CheA1 and CheA2. Our previous study showed that CheA2 is a genuine CHK that is required for chemotaxis; however, the role of CheA1 remains mysterious. This report first compares the structural features that differentiate CheA1 and CheA2 and then provides evidence to show that CheA1 is an atypical CHK that controls the virulence of B. burgdorferi through modulating the stability of RpoS, a key transcriptional regulator of the spirochete. First, microscopic analyses using green-fluorescence-protein (GFP) tags reveal that CheA1 has a unique and dynamic cellular localization. Second, loss-of-function studies indicate that CheA1 is not required for chemotaxis in vitro despite sharing a high sequence and structural similarity to its counterparts from other bacteria. Third, mouse infection studies using needle inoculations show that a deletion mutant of CheA1 (cheA1mut) is able to establish systemic infection in immune-deficient mice but fails to do so in immune-competent mice albeit the mutant can survive at the inoculation site for up to 28 days. Tick and mouse infection studies further demonstrate that CheA1 is dispensable for tick colonization and acquisition but essential for tick transmission. Lastly, mechanistic studies combining immunoblotting, protein turnover, mutagenesis, and RNA-seq analyses reveal that depletion of CheA1 affects RpoS stability, leading to reduced expression of several RpoS-regulated virulence factors (i.e., OspC, BBK32, and DbpA), likely due to dysregulated clpX and lon protease expression. Bulk RNA-seq analysis of infected mouse skin tissues further show that cheA1mut fails to elicit mouse tnf-α, il-10, il-1β, and ccl2 expression, four important cytokines for Lyme disease development and B. burgdorferi transmigration. Collectively, these results reveal a unique role and regulatory mechanism of CheA1 in modulating virulence factor expression and add new insights into understanding the regulatory network of B. burgdorferi.
Collapse
Affiliation(s)
- Ching Wooen Sze
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Kai Zhang
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Michael J. Lynch
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Radha Iyer
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Brian R. Crane
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Ira Schwartz
- Department of Pathology, Microbiology, and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Chunhao Li
- Department of Oral Craniofacial Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
38
|
Foor SD, Brangulis K, Shakya AK, Rana VS, Bista S, Kitsou C, Ronzetti M, Alreja AB, Linden SB, Altieri AS, Baljinnyam B, Akopjana I, Nelson DC, Simeonov A, Herzberg O, Caimano MJ, Pal U. A unique borrelial protein facilitates microbial immune evasion. mBio 2023; 14:e0213523. [PMID: 37830812 PMCID: PMC10653885 DOI: 10.1128/mbio.02135-23] [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: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 10/14/2023] Open
Abstract
IMPORTANCE Lyme disease is a major tick-borne infection caused by a bacterial pathogen called Borrelia burgdorferi, which is transmitted by ticks and affects hundreds of thousands of people every year. These bacterial pathogens are distinct from other genera of microbes because of their distinct features and ability to transmit a multi-system infection to a range of vertebrates, including humans. Progress in understanding the infection biology of Lyme disease, and thus advancements towards its prevention, are hindered by an incomplete understanding of the microbiology of B. burgdorferi, partly due to the occurrence of many unique borrelial proteins that are structurally unrelated to proteins of known functions yet are indispensable for pathogen survival. We herein report the use of diverse technologies to examine the structure and function of a unique B. burgdorferi protein, annotated as BB0238-an essential virulence determinant. We show that the protein is structurally organized into two distinct domains, is involved in multiplex protein-protein interactions, and facilitates tick-to-mouse pathogen transmission by aiding microbial evasion of early host cellular immunity. We believe that our findings will further enrich our understanding of the microbiology of B. burgdorferi, potentially impacting the future development of novel prevention strategies against a widespread tick-transmitted infection.
Collapse
Affiliation(s)
- Shelby D. Foor
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Kalvis Brangulis
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Department of Human Physiology and Biochemistry, Riga Stradins University, Riga, Latvia
| | - Anil K. Shakya
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, USA
| | - Vipin S. Rana
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Sandhya Bista
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Michael Ronzetti
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Adit B. Alreja
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, USA
| | - Sara B. Linden
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, USA
| | - Amanda S. Altieri
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, USA
| | - Bolormaa Baljinnyam
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Inara Akopjana
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Daniel C. Nelson
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Osnat Herzberg
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, USA
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Melissa J. Caimano
- Departments of Medicine, Pediatrics, and Molecular Biology and Biophysics, UConn Health, Farmington, Connecticut, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
- Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
39
|
Baquer F, Jaulhac B, Barthel C, Paz M, Wolfgramm J, Müller A, Boulanger N, Grillon A. Skin microbiota secretomes modulate cutaneous innate immunity against Borrelia burgdorferi s.s. Sci Rep 2023; 13:16393. [PMID: 37773515 PMCID: PMC10541882 DOI: 10.1038/s41598-023-43566-0] [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: 06/13/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023] Open
Abstract
In Lyme borreliosis, the skin constitutes a major interface for the host, the bacteria and the tick. Skin immunity is provided by specialized immune cells but also by the resident cells: the keratinocytes and the fibroblasts. Discoveries on the role of the microbiome in the modulation of skin inflammation and immunity have reinforced the potential importance of the skin in vector-borne diseases. In this study, we analyzed in vitro the interaction of human primary keratinocytes and fibroblasts with Borrelia burgdorferi sensu stricto N40 in presence or absence of bacterial commensal supernatants. We aimed to highlight the role of resident skin cells and skin microbiome on the inflammation induced by B. burgdorferi s.s.. The secretomes of Staphylococcus epidermidis, Corynebacterium striatum and Cutibacterium acnes showed an overall increase in the expression of IL-8, CXCL1, MCP-1 and SOD-2 by fibroblasts, and of IL-8, CXCL1, MCP-1 and hBD-2 in the undifferentiated keratinocytes. Commensal bacteria showed a repressive effect on the expression of IL-8, CXCL1 and MCP-1 by differentiated keratinocytes. Besides the inflammatory effect observed in the presence of Borrelia on all cell types, the cutaneous microbiome appears to promote a rapid innate response of resident skin cells during the onset of Borrelia infection.
Collapse
Affiliation(s)
- F Baquer
- Institut de Bactériologie, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, UR7290, ITI InnoVec, 3 Rue Koeberlé, 67000, Strasbourg, France.
- Laboratory of Bacteriology, Strasbourg University Hospital, 67000, Strasbourg, France.
| | - B Jaulhac
- Institut de Bactériologie, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, UR7290, ITI InnoVec, 3 Rue Koeberlé, 67000, Strasbourg, France
- Laboratory of Bacteriology, Strasbourg University Hospital, 67000, Strasbourg, France
- French National Reference Center for Borrelia, Strasbourg University Hospital, 67000, Strasbourg, France
| | - C Barthel
- Institut de Bactériologie, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, UR7290, ITI InnoVec, 3 Rue Koeberlé, 67000, Strasbourg, France
| | - M Paz
- Institut de Bactériologie, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, UR7290, ITI InnoVec, 3 Rue Koeberlé, 67000, Strasbourg, France
| | - J Wolfgramm
- Institut de Bactériologie, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, UR7290, ITI InnoVec, 3 Rue Koeberlé, 67000, Strasbourg, France
| | - A Müller
- Institut de Bactériologie, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, UR7290, ITI InnoVec, 3 Rue Koeberlé, 67000, Strasbourg, France
| | - N Boulanger
- Institut de Bactériologie, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, UR7290, ITI InnoVec, 3 Rue Koeberlé, 67000, Strasbourg, France
- French National Reference Center for Borrelia, Strasbourg University Hospital, 67000, Strasbourg, France
| | - A Grillon
- Institut de Bactériologie, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, UR7290, ITI InnoVec, 3 Rue Koeberlé, 67000, Strasbourg, France
- Laboratory of Bacteriology, Strasbourg University Hospital, 67000, Strasbourg, France
- French National Reference Center for Borrelia, Strasbourg University Hospital, 67000, Strasbourg, France
| |
Collapse
|
40
|
Hammond EM, Olsen KJ, Ram S, Tran GVV, Hall LS, Bradley JE, Lund FE, Samuels DS, Baumgarth N. Antigen-Specific CD4 T Cell and B Cell Responses to Borrelia burgdorferi. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:994-1005. [PMID: 37556156 PMCID: PMC10530202 DOI: 10.4049/jimmunol.2200890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/18/2023] [Indexed: 08/10/2023]
Abstract
Long-lived T-dependent B cell responses fail to develop during persistent infection of mice with Borrelia burgdorferi, the causative agent of Lyme disease, raising questions about the induction and/or functionality of anti-B. burgdorferi adaptive immune responses. Yet, a lack of reagents has limited investigations into B. burgdorferi-specific T and B cells. We attempted two approaches to track B. burgdorferi-induced CD4 T cells. First, a B. burgdorferi mutant was generated with an influenza hemagglutinin (HA) peptide, HA111-119, inserted into the B. burgdorferi arthritis-related protein (Arp) locus. Although this B. burgdorferi arp::HA strain remained infectious, peptide-specific TCR transgenic CD4 T cells in vitro, or adoptively transferred into B. burgdorferi arp::HA-infected BALB/c mice, did not clonally expand above those of recipients infected with the parental B. burgdorferi strain or a B. burgdorferi mutant containing an irrelevant peptide. Some expansion, however, occurred in B. burgdorferi arp::HA-infected BALB/c SCID mice. Second, a (to our knowledge) newly identified I-Ab-restricted CD4 T cell epitope, Arp152-166, was used to generate Arp MHC class II tetramers. Flow cytometry showed small numbers of Arp-specific CD4 T cells emerging in mice infected with B. burgdorferi but not with Arp-deficient Borrelia afzelii. Although up to 30% of Arp-specific CD4 T cells were ICOS+PD-1+CXCR5+BCL6+ T follicular helper cells, their numbers declined after day 12, before germinal centers (GCs) are prominent. Although some Arp-specific B cells, identified using fluorochrome-labeled rArp proteins, had the phenotype of GC B cells, their frequencies did not correlate with anti-Arp serum IgG. The data suggest a failure not in the induction, but in the maintenance of GC T follicular helper and/or B cells to B. burgdorferi.
Collapse
Affiliation(s)
- Elizabeth M. Hammond
- Graduate Group in Immunology, University of California Davis
- Center for Immunology and Infectious Diseases, University of California Davis
- Department of Pathology, Microbiology, and Immunology, University of California Davis
| | - Kimberly J. Olsen
- Center for Immunology and Infectious Diseases, University of California Davis
- Department of Pathology, Microbiology, and Immunology, University of California Davis
| | - Shivneel Ram
- Center for Immunology and Infectious Diseases, University of California Davis
| | - Giang Vu Vi Tran
- Center for Immunology and Infectious Diseases, University of California Davis
- Department of Pathology, Microbiology, and Immunology, University of California Davis
| | - Laura S. Hall
- Division of Biological Sciences, University of Montana
| | - John E. Bradley
- Department of Microbiology, University of Alabama, Birmingham
| | - Frances E. Lund
- Department of Microbiology, University of Alabama, Birmingham
| | | | - Nicole Baumgarth
- Graduate Group in Immunology, University of California Davis
- Center for Immunology and Infectious Diseases, University of California Davis
- Department of Pathology, Microbiology, and Immunology, University of California Davis
- Department of Molecular Microbiology and Immunology and Department of Molecular and Comparative Pathobiology, Johns Hopkins University
| |
Collapse
|
41
|
Pine M, Arora G, Hart TM, Bettini E, Gaudette BT, Muramatsu H, Tombácz I, Kambayashi T, Tam YK, Brisson D, Allman D, Locci M, Weissman D, Fikrig E, Pardi N. Development of an mRNA-lipid nanoparticle vaccine against Lyme disease. Mol Ther 2023; 31:2702-2714. [PMID: 37533256 PMCID: PMC10492027 DOI: 10.1016/j.ymthe.2023.07.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/19/2023] [Accepted: 07/28/2023] [Indexed: 08/04/2023] Open
Abstract
Lyme disease is the most common vector-borne infectious disease in the United States, in part because a vaccine against it is not currently available for humans. We propose utilizing the lipid nanoparticle-encapsulated nucleoside-modified mRNA (mRNA-LNP) platform to generate a Lyme disease vaccine like the successful clinical vaccines against SARS-CoV-2. Of the antigens expressed by Borrelia burgdorferi, the causative agent of Lyme disease, outer surface protein A (OspA) is the most promising candidate for vaccine development. We have designed and synthesized an OspA-encoding mRNA-LNP vaccine and compared its immunogenicity and protective efficacy to an alum-adjuvanted OspA protein subunit vaccine. OspA mRNA-LNP induced superior humoral and cell-mediated immune responses in mice after a single immunization. These potent immune responses resulted in protection against bacterial infection. Our study demonstrates that highly efficient mRNA vaccines can be developed against bacterial targets.
Collapse
Affiliation(s)
- Matthew Pine
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Thomas M Hart
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Emily Bettini
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brian T Gaudette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - István Tombácz
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Dustin Brisson
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Allman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michela Locci
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
42
|
Stevenson B. The Lyme disease spirochete, Borrelia burgdorferi, as a model vector-borne pathogen: insights on regulation of gene and protein expression. Curr Opin Microbiol 2023; 74:102332. [PMID: 37279610 PMCID: PMC10524203 DOI: 10.1016/j.mib.2023.102332] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 06/08/2023]
Abstract
The Lyme disease spirochete persists in nature through cycles between ticks and vertebrates. Although the spirochete interacts with numerous, distinct tissues and environmental conditions during its infectious cycle, Borrelia burgdorferi appears to possess a limited ability to sense its external environment. This apparent paradox is being resolved through detailed investigations of the molecular mechanisms through which B. burgdorferi controls production of virulence-associated factors such as the Erp outer surface proteins. The results have led to development of a model for how B. burgdorferi controls expression of its diverse proteins, wherein physiological and metabolic states that are unique to specific points in the infectious cycle trigger changes in gene and protein expression levels.
Collapse
Affiliation(s)
- Brian Stevenson
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA; Department of Entomology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
43
|
Lemieux JE, Huang W, Hill N, Cerar T, Freimark L, Hernandez S, Luban M, Maraspin V, Bogovič P, Ogrinc K, Ruzič-Sabljič E, Lapierre P, Lasek-Nesselquist E, Singh N, Iyer R, Liveris D, Reed KD, Leong JM, Branda JA, Steere AC, Wormser GP, Strle F, Sabeti PC, Schwartz I, Strle K. Whole genome sequencing of human Borrelia burgdorferi isolates reveals linked blocks of accessory genome elements located on plasmids and associated with human dissemination. PLoS Pathog 2023; 19:e1011243. [PMID: 37651316 PMCID: PMC10470944 DOI: 10.1371/journal.ppat.1011243] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/13/2023] [Indexed: 09/02/2023] Open
Abstract
Lyme disease is the most common vector-borne disease in North America and Europe. The clinical manifestations of Lyme disease vary based on the genospecies of the infecting Borrelia burgdorferi spirochete, but the microbial genetic elements underlying these associations are not known. Here, we report the whole genome sequence (WGS) and analysis of 299 B. burgdorferi (Bb) isolates derived from patients in the Eastern and Midwestern US and Central Europe. We develop a WGS-based classification of Bb isolates, confirm and extend the findings of previous single- and multi-locus typing systems, define the plasmid profiles of human-infectious Bb isolates, annotate the core and strain-variable surface lipoproteome, and identify loci associated with disseminated infection. A core genome consisting of ~900 open reading frames and a core set of plasmids consisting of lp17, lp25, lp36, lp28-3, lp28-4, lp54, and cp26 are found in nearly all isolates. Strain-variable (accessory) plasmids and genes correlate strongly with phylogeny. Using genetic association study methods, we identify an accessory genome signature associated with dissemination in humans and define the individual plasmids and genes that make up this signature. Strains within the RST1/WGS A subgroup, particularly a subset marked by the OspC type A genotype, have increased rates of dissemination in humans. OspC type A strains possess a unique set of strongly linked genetic elements including the presence of lp56 and lp28-1 plasmids and a cluster of genes that may contribute to their enhanced virulence compared to other genotypes. These features of OspC type A strains reflect a broader paradigm across Bb isolates, in which near-clonal genotypes are defined by strain-specific clusters of linked genetic elements, particularly those encoding surface-exposed lipoproteins. These clusters of genes are maintained by strain-specific patterns of plasmid occupancy and are associated with the probability of invasive infection.
Collapse
Affiliation(s)
- Jacob E. Lemieux
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Weihua Huang
- New York Medical College, Valhalla, New York, United States of America
- East Carolina University, Greenville, North Carolina, United States of America
| | - Nathan Hill
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Tjasa Cerar
- University of Ljubljana, Ljubljana, Slovenia
| | - Lisa Freimark
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Sergio Hernandez
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Matteo Luban
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Vera Maraspin
- University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Petra Bogovič
- University Medical Center Ljubljana, Ljubljana, Slovenia
| | | | | | - Pascal Lapierre
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Erica Lasek-Nesselquist
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Navjot Singh
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Radha Iyer
- New York Medical College, Valhalla, New York, United States of America
| | - Dionysios Liveris
- New York Medical College, Valhalla, New York, United States of America
| | - Kurt D. Reed
- University of Wisconsin, Madison, Wisconsin, United States of America
| | - John M. Leong
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - John A. Branda
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Allen C. Steere
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gary P. Wormser
- New York Medical College, Valhalla, New York, United States of America
| | - Franc Strle
- University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Pardis C. Sabeti
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Harvard University, Cambridge, Massachusetts, United States of America
- Harvard T.H.Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Ira Schwartz
- New York Medical College, Valhalla, New York, United States of America
| | - Klemen Strle
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
44
|
Wu-Chuang A, Mateos-Hernandez L, Maitre A, Rego ROM, Šíma R, Porcelli S, Rakotobe S, Foucault-Simonin A, Moutailler S, Palinauskas V, Aželytė J, Sǐmo L, Obregon D, Cabezas-Cruz A. Microbiota perturbation by anti-microbiota vaccine reduces the colonization of Borrelia afzelii in Ixodes ricinus. MICROBIOME 2023; 11:151. [PMID: 37482606 PMCID: PMC10364381 DOI: 10.1186/s40168-023-01599-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 06/14/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Ticks can transmit a broad variety of pathogens of medical importance, including Borrelia afzelii, the causative agent of Lyme borreliosis in Europe. Tick microbiota is an important factor modulating, not only vector physiology, but also the vector competence. Anti-microbiota vaccines targeting keystone taxa of tick microbiota can alter tick feeding and modulate the taxonomic and functional profiles of bacterial communities in the vector. However, the impact of anti-microbiota vaccine on tick-borne pathogen development within the vector has not been tested. RESULTS Here, we characterized the Ixodes ricinus microbiota modulation in response to B. afzelii infection and found that the pathogen induces changes in the microbiota composition, its beta diversity and structure of bacterial community assembly. Tick microbiota perturbation by anti-microbiota antibodies or addition of novel commensal bacteria into tick midguts causes departures from the B. afzelii-induced modulation of tick microbiota which resulted in a lower load of the pathogen in I. ricinus. Co-occurrence networks allowed the identification of emergent properties of the bacterial communities which better defined the Borrelia infection-refractory states of the tick microbiota. CONCLUSIONS These findings suggest that Borrelia is highly sensitive to tick microbiota perturbations and that departure from the modulation induced by the pathogen in the vector microbiota pose a high cost to the spirochete. Network analysis emerges as a suitable tool to identify emergent properties of the vector microbiota associated with infection-refractory states. Anti-microbiota vaccines can be used as a tool for microbiota perturbation and control of important vector-borne pathogens. Video Abstract.
Collapse
Affiliation(s)
- Alejandra Wu-Chuang
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | - Lourdes Mateos-Hernandez
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | - Apolline Maitre
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | - Ryan O M Rego
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Radek Šíma
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
- Biopticka Laborator S.R.O, Plzen, Czech Republic
| | - Stefania Porcelli
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | - Sabine Rakotobe
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | - Angélique Foucault-Simonin
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | - Sara Moutailler
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | | | - Justė Aželytė
- Nature Research Centre, Akademijos 2, 09412, Vilnius, Lithuania
| | - Ladislav Sǐmo
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | - Dasiel Obregon
- School of Environmental Sciences, University of Guelph, Guelph, ON, Canada
| | - Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France.
| |
Collapse
|
45
|
Hepner S, Kuleshov K, Tooming-Kunderud A, Alig N, Gofton A, Casjens S, Rollins RE, Dangel A, Mourkas E, Sheppard SK, Wieser A, Hübner J, Sing A, Fingerle V, Margos G. A high fidelity approach to assembling the complex Borrelia genome. BMC Genomics 2023; 24:401. [PMID: 37460975 DOI: 10.1186/s12864-023-09500-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Bacteria of the Borrelia burgdorferi sensu lato (s.l.) complex can cause Lyme borreliosis. Different B. burgdorferi s.l. genospecies vary in their host and vector associations and human pathogenicity but the genetic basis for these adaptations is unresolved and requires completed and reliable genomes for comparative analyses. The de novo assembly of a complete Borrelia genome is challenging due to the high levels of complexity, represented by a high number of circular and linear plasmids that are dynamic, showing mosaic structure and sequence homology. Previous work demonstrated that even advanced approaches, such as a combination of short-read and long-read data, might lead to incomplete plasmid reconstruction. Here, using recently developed high-fidelity (HiFi) PacBio sequencing, we explored strategies to obtain gap-free, complete and high quality Borrelia genome assemblies. Optimizing genome assembly, quality control and refinement steps, we critically appraised existing techniques to create a workflow that lead to improved genome reconstruction. RESULTS Despite the latest available technologies, stand-alone sequencing and assembly methods are insufficient for the generation of complete and high quality Borrelia genome assemblies. We developed a workflow pipeline for the de novo genome assembly for Borrelia using several types of sequence data and incorporating multiple assemblers to recover the complete genome including both circular and linear plasmid sequences. CONCLUSION Our study demonstrates that, with HiFi data and an ensemble reconstruction pipeline with refinement steps, chromosomal and plasmid sequences can be fully resolved, even for complex genomes such as Borrelia. The presented pipeline may be of interest for the assembly of further complex microbial genomes.
Collapse
Affiliation(s)
- Sabrina Hepner
- German National Reference Centre for Borrelia, Oberschleissheim, Germany.
- Bavarian Health and Food Safety Authority, Oberschleissheim, Germany.
| | | | - Ave Tooming-Kunderud
- Department of Biosciences, Norwegian Sequencing Centre at Centre for Ecological and Evolutionary Synthesis, University of Oslo, Oslo, Norway
| | - Nikolas Alig
- German National Reference Centre for Borrelia, Oberschleissheim, Germany
- Bavarian Health and Food Safety Authority, Oberschleissheim, Germany
| | | | - Sherwood Casjens
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Robert E Rollins
- Institute of Avian Research "Vogelwarte Helgoland", Wilhelmshaven, Germany
| | - Alexandra Dangel
- Bavarian Health and Food Safety Authority, Oberschleissheim, Germany
| | | | | | - Andreas Wieser
- Medical Microbiology and Hospital Epidemiology, Max von Pettenkofer Institute, Faculty of Medicine, LMU Munich, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- Imunology, Infectious Disease and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Munich, Germany
| | - Johannes Hübner
- Dr. Von Hauner Children's Hospital, LMU Munich, Munich, Germany
| | - Andreas Sing
- German National Reference Centre for Borrelia, Oberschleissheim, Germany
- Bavarian Health and Food Safety Authority, Oberschleissheim, Germany
| | - Volker Fingerle
- German National Reference Centre for Borrelia, Oberschleissheim, Germany
- Bavarian Health and Food Safety Authority, Oberschleissheim, Germany
| | - Gabriele Margos
- German National Reference Centre for Borrelia, Oberschleissheim, Germany
- Bavarian Health and Food Safety Authority, Oberschleissheim, Germany
| |
Collapse
|
46
|
Sapiro AL, Hayes BM, Volk RF, Zhang JY, Brooks DM, Martyn C, Radkov A, Zhao Z, Kinnersley M, Secor PR, Zaro BW, Chou S. Longitudinal map of transcriptome changes in the Lyme pathogen Borrelia burgdorferi during tick-borne transmission. eLife 2023; 12:RP86636. [PMID: 37449477 PMCID: PMC10393048 DOI: 10.7554/elife.86636] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Borrelia burgdorferi (Bb), the causative agent of Lyme disease, adapts to vastly different environments as it cycles between tick vector and vertebrate host. During a tick bloodmeal, Bb alters its gene expression to prepare for vertebrate infection; however, the full range of transcriptional changes that occur over several days inside of the tick are technically challenging to capture. We developed an experimental approach to enrich Bb cells to longitudinally define their global transcriptomic landscape inside nymphal Ixodes scapularis ticks during a transmitting bloodmeal. We identified 192 Bb genes that substantially change expression over the course of the bloodmeal from 1 to 4 days after host attachment. The majority of upregulated genes encode proteins found at the cell envelope or proteins of unknown function, including 45 outer surface lipoproteins embedded in the unusual protein-rich coat of Bb. As these proteins may facilitate Bb interactions with the host, we utilized mass spectrometry to identify candidate tick proteins that physically associate with Bb. The Bb enrichment methodology along with the ex vivo Bb transcriptomes and candidate tick interacting proteins presented here provide a resource to facilitate investigations into key determinants of Bb priming and transmission during the tick stage of its unique transmission cycle.
Collapse
Affiliation(s)
- Anne L Sapiro
- Department of Biochemistry & Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Beth M Hayes
- Department of Biochemistry & Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Regan F Volk
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Jenny Y Zhang
- Department of Biochemistry & Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Diane M Brooks
- Division of Biological Sciences, University of MontanaMissoulaUnited States
| | - Calla Martyn
- Department of Biochemistry & Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Atanas Radkov
- Department of Biochemistry & Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Ziyi Zhao
- Department of Biochemistry & Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Margie Kinnersley
- Division of Biological Sciences, University of MontanaMissoulaUnited States
| | - Patrick R Secor
- Division of Biological Sciences, University of MontanaMissoulaUnited States
| | - Balyn W Zaro
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Seemay Chou
- Department of Biochemistry & Biophysics, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
47
|
Petroni E, Esnault C, Tetreault D, Dale RK, Storz G, Adams PP. Extensive diversity in RNA termination and regulation revealed by transcriptome mapping for the Lyme pathogen Borrelia burgdorferi. Nat Commun 2023; 14:3931. [PMID: 37402717 PMCID: PMC10319736 DOI: 10.1038/s41467-023-39576-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/16/2023] [Indexed: 07/06/2023] Open
Abstract
Transcription termination is an essential and dynamic process that can tune gene expression in response to diverse molecular signals. Yet, the genomic positions, molecular mechanisms, and regulatory consequences of termination have only been studied thoroughly in model bacteria. Here, we use several RNA-seq approaches to map RNA ends for the transcriptome of the spirochete Borrelia burgdorferi - the etiological agent of Lyme disease. We identify complex gene arrangements and operons, untranslated regions and small RNAs. We predict intrinsic terminators and experimentally test examples of Rho-dependent transcription termination. Remarkably, 63% of RNA 3' ends map upstream of or internal to open reading frames (ORFs), including genes involved in the unique infectious cycle of B. burgdorferi. We suggest these RNAs result from premature termination, processing and regulatory events such as cis-acting regulation. Furthermore, the polyamine spermidine globally influences the generation of truncated mRNAs. Collectively, our findings provide insights into transcription termination and uncover an abundance of potential RNA regulators in B. burgdorferi.
Collapse
Affiliation(s)
- Emily Petroni
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Caroline Esnault
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Daniel Tetreault
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Ryan K Dale
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Gisela Storz
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Philip P Adams
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA.
- Postdoctoral Research Associate Program, National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD, 20892, USA.
- Independent Research Scholar Program, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
48
|
Van Gundy T, Patel D, Bowler BE, Rothfuss MT, Hall AJ, Davies C, Hall LS, Drecktrah D, Marconi RT, Samuels DS, Lybecker MC. c-di-GMP regulates activity of the PlzA RNA chaperone from the Lyme disease spirochete. Mol Microbiol 2023; 119:711-727. [PMID: 37086029 PMCID: PMC10330241 DOI: 10.1111/mmi.15066] [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: 04/03/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/23/2023]
Abstract
PlzA is a c-di-GMP-binding protein crucial for adaptation of the Lyme disease spirochete Borrelia (Borreliella) burgdorferi during its enzootic life cycle. Unliganded apo-PlzA is important for vertebrate infection, while liganded holo-PlzA is important for survival in the tick; however, the biological function of PlzA has remained enigmatic. Here, we report that PlzA has RNA chaperone activity that is inhibited by c-di-GMP binding. Holo- and apo-PlzA bind RNA and accelerate RNA annealing, while only apo-PlzA can strand displace and unwind double-stranded RNA. Guided by the crystal structure of PlzA, we identified several key aromatic amino acids protruding from the N- and C-terminal domains that are required for RNA-binding and unwinding activity. Our findings illuminate c-di-GMP as a switch controlling the RNA chaperone activity of PlzA, and we propose that complex RNA-mediated modulatory mechanisms allow PlzA to regulate gene expression during both the vector and host phases of the B. burgdorferi life cycle.
Collapse
Affiliation(s)
- Taylor Van Gundy
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Center for Disease Control and Prevention, Fort Collins, CO 80521, USA
| | - Dhara Patel
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA
| | - Bruce E. Bowler
- Department of Chemistry and Biochemistry, University of Montana, Missoula, MT 59812, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Michael T. Rothfuss
- Department of Chemistry and Biochemistry, University of Montana, Missoula, MT 59812, USA
| | - Allie J. Hall
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Center for Disease Control and Prevention, Fort Collins, CO 80521, USA
| | - Christopher Davies
- Department of Biochemistry and Molecular Biology, University of Southern Alabama, Mobile, AL 36688, USA
| | - Laura S. Hall
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Dan Drecktrah
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Richard T. Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA
| | - D. Scott Samuels
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Meghan C. Lybecker
- Bacterial Diseases Branch, Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Center for Disease Control and Prevention, Fort Collins, CO 80521, USA
- Department of Biology, University of Colorado, 1420 Austin Bluffs Parkway, Colorado Springs CO 80917, USA
| |
Collapse
|
49
|
Farris LC, Torres-Odio S, Adams LG, West AP, Hyde JA. Borrelia burgdorferi Engages Mammalian Type I IFN Responses via the cGAS-STING Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1761-1770. [PMID: 37067290 PMCID: PMC10192154 DOI: 10.4049/jimmunol.2200354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 03/23/2023] [Indexed: 04/18/2023]
Abstract
Borrelia burgdorferi, the etiologic agent of Lyme disease, is a spirochete that modulates numerous host pathways to cause a chronic, multisystem inflammatory disease in humans. B. burgdorferi infection can lead to Lyme carditis, neurologic complications, and arthritis because of the ability of specific borrelial strains to disseminate, invade, and drive inflammation. B. burgdorferi elicits type I IFN (IFN-I) responses in mammalian cells and tissues that are associated with the development of severe arthritis or other Lyme-related complications. However, the innate immune sensors and signaling pathways controlling IFN-I induction remain unclear. In this study, we examined whether intracellular nucleic acid sensing is required for the induction of IFN-I to B. burgdorferi. Using fluorescence microscopy, we show that B. burgdorferi associates with mouse and human cells in culture, and we document that internalized spirochetes colocalize with the pattern recognition receptor cyclic GMP-AMP synthase (cGAS). Moreover, we report that IFN-I responses in mouse macrophages and murine embryonic fibroblasts are significantly attenuated in the absence of cGAS or its adaptor stimulator of IFN genes (STING), which function to sense and respond to intracellular DNA. Longitudinal in vivo tracking of bioluminescent B. burgdorferi revealed similar dissemination kinetics and borrelial load in C57BL/6J wild-type, cGAS-deficient, or STING-deficient mice. However, infection-associated tibiotarsal joint pathology and inflammation were modestly reduced in cGAS-deficient compared with wild-type mice. Collectively, these results indicate that the cGAS-STING pathway is a critical mediator of mammalian IFN-I signaling and innate immune responses to B. burgdorferi.
Collapse
Affiliation(s)
- Lauren C. Farris
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, USA
| | - Sylvia Torres-Odio
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, USA
| | - L. Garry Adams
- Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - A. Phillip West
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, USA
| | - Jenny A. Hyde
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, USA
| |
Collapse
|
50
|
Saylor TC, Savage CR, Krusenstjerna AC, Jusufovic N, Zückert WR, Brissette CA, Motaleb M, Schlax PJ, Stevenson B. Quantitative analyses of interactions between SpoVG and RNA/DNA. Biochem Biophys Res Commun 2023; 654:40-46. [PMID: 36889033 PMCID: PMC11086051 DOI: 10.1016/j.bbrc.2023.02.044] [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: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 03/02/2023]
Abstract
The Borrelia burgdorferi SpoVG protein has previously been found to be a DNA- and RNA-binding protein. To aid in the elucidation of ligand motifs, affinities for numerous RNAs, ssDNAs, and dsDNAs were measured and compared. The loci used in the study were spoVG, glpFKD, erpAB, bb0242, flaB, and ospAB, with particular focus on the untranslated 5' portion of the mRNAs. Performing binding and competition assays yielded that the 5' end of spoVG mRNA had the highest affinity while the lowest observed affinity was to the 5' end of flaB mRNA. Mutagenesis studies of spoVG RNA and ssDNA sequences suggested that the formation of SpoVG-nucleic acid complexes are not entirely dependent on either sequence or structure. Additionally, exchanging uracil for thymine in ssDNAs did not affect protein-nucleic acid complex formation.
Collapse
Affiliation(s)
- Timothy C Saylor
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Christina R Savage
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Andrew C Krusenstjerna
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Nerina Jusufovic
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Wolfram R Zückert
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Catherine A Brissette
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Md Motaleb
- Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA
| | - Paula J Schlax
- Department of Chemistry and Biochemistry, Bates College, Lewiston, ME, USA
| | - Brian Stevenson
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA; Department of Entomology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|