Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Vaccines (Basel) ; 12(1)2024 Jan 12.
Article En | MEDLINE | ID: mdl-38250891

Lyme disease (LD) is the most common tick-borne illness in the United States (U.S.), Europe, and Asia. Borrelia burgdorferi, a spirochete bacterium transmitted by the tick vector Ixodes scapularis, causes LD in the U.S. If untreated, Lyme arthritis, heart block, and meningitis can occur. Given the absence of a human Lyme disease vaccine, we developed a vaccine using the rabies virus (RABV) vaccine vector BNSP333 and an outer surface borrelial protein, BBI39. BBI39 was previously utilized as a recombinant protein vaccine and was protective in challenge experiments; therefore, we decided to utilize this protective antigen in a rabies virus-vectored vaccine against Borrelia burgdorferi. To incorporate BBI39 into the RABV virion, we generated a chimeric BBI39 antigen, BBI39RVG, by fusing BBI39 with the final amino acids of the RABV glycoprotein by molecular cloning and viral recovery with reverse transcription genetics. Here, we have demonstrated that the BBI39RVG antigen was incorporated into the RABV virion via immunofluorescence and Western blot analysis. Mice vaccinated with our BPL inactivated RABV-BBI39RVG (BNSP333-BBI39RVG) vaccine induced high amounts of BBI39-specific antibodies, which were maintained long-term, up to eight months post-vaccination. The BBI39 antibodies neutralized Borrelia in vaccinated mice when challenged with Borrelia burgdorferi by either syringe injection or infected ticks and they reduced the Lyme disease pathology of arthritis in infected mouse joints. Overall, the RABV-based LD vaccine induced more and longer-term antibodies compared to the recombinant protein vaccine. This resulted in lower borrelial RNA in RABV-based vaccinated mice compared to recombinant protein vaccinated mice. The results of this study indicate the successful use of BBI39 as a vaccine antigen and RABV as a vaccine vector for LD.

2.
mSystems ; 9(1): e0108723, 2024 Jan 23.
Article En | MEDLINE | ID: mdl-38078774

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.


Borrelia burgdorferi , Lyme Disease , Mice , Animals , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Lipoproteins/genetics , Bacterial Vaccines/genetics , Antigens, Surface/genetics , Lyme Disease/diagnosis , Borrelia burgdorferi/genetics , Mammals
3.
mBio ; 14(5): e0213523, 2023 Oct 31.
Article En | MEDLINE | ID: mdl-37830812

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.


Borrelia burgdorferi , Borrelia , Ixodes , Lyme Disease , Ticks , Animals , Humans , Mice , Immune Evasion , Lyme Disease/microbiology , Borrelia burgdorferi/metabolism , Ticks/microbiology , Ixodes/microbiology
4.
Nat Genet ; 55(2): 301-311, 2023 02.
Article En | MEDLINE | ID: mdl-36658436

Ixodes spp. and related ticks transmit prevalent infections, although knowledge of their biology and development of anti-tick measures have been hindered by the lack of a high-quality genome. In the present study, we present the assembly of a 2.23-Gb Ixodes scapularis genome by sequencing two haplotypes within one individual, complemented by chromosome-level scaffolding and full-length RNA isoform sequencing, yielding a fully reannotated genome featuring thousands of new protein-coding genes and various RNA species. Analyses of the repetitive DNA identified transposable elements, whereas the examination of tick-associated bacterial sequences yielded an improved Rickettsia buchneri genome. We demonstrate how the Ixodes genome advances tick science by contributing to new annotations, gene models and epigenetic functions, expansion of gene families, development of in-depth proteome catalogs and deciphering of genetic variations in wild ticks. Overall, we report critical genetic resources and biological insights impacting our understanding of tick biology and future interventions against tick-transmitted infections.


Ixodes , Animals , Ixodes/genetics , Ixodes/microbiology , Genome/genetics , Bacteria/genetics , Base Sequence , RNA
5.
Science ; 379(6628): eabl3837, 2023 01 13.
Article En | MEDLINE | ID: mdl-36634189

Ancestral signaling pathways serve critical roles in metazoan development, physiology, and immunity. We report an evolutionary interspecies communication pathway involving a central Ixodes scapularis tick receptor termed Dome1, which acquired a mammalian cytokine receptor motif exhibiting high affinity for interferon-gamma (IFN-γ). Host-derived IFN-γ facilitates Dome1-mediated activation of the Ixodes JAK-STAT pathway. This accelerates tick blood meal acquisition and development while upregulating antimicrobial components. The Dome1-JAK-STAT pathway, which exists in most Ixodid tick genomes, regulates the regeneration and proliferation of gut cells-including stem cells-and dictates metamorphosis through the Hedgehog and Notch-Delta networks, ultimately affecting Ixodes vectorial competence. We highlight the evolutionary dependence of I. scapularis on mammalian hosts through cross-species signaling mechanisms that dually influence arthropod immunity and development.


Arachnid Vectors , Host-Parasite Interactions , Ixodes , Janus Kinases , Receptors, Cytokine , STAT Transcription Factors , Animals , Interferon-gamma/metabolism , Ixodes/genetics , Ixodes/immunology , Janus Kinases/genetics , Janus Kinases/metabolism , Signal Transduction , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Host-Parasite Interactions/immunology , Receptors, Cytokine/metabolism , Arachnid Vectors/immunology
6.
Trends Microbiol ; 31(1): 62-75, 2023 01.
Article En | MEDLINE | ID: mdl-36055896

Tick-transmitted bacterial pathogens thrive in enzootic infection cycles, colonizing disparate vertebrate and arthropod tissues, often establishing persistent infections. Therefore, the evolution of robust immune evasion strategies is central to their successful persistence or transmission between hosts. To survive in nature, these pathogens must counteract a broad range of microbicidal host responses that can be localized, tissue-specific, or systemic, including a mix of these responses at the host-vector interface. Herein, we review microbial immune evasion strategies focusing on Lyme disease spirochetes and rickettsial or tularemia agents as models for extracellular and intracellular tick-borne pathogens, respectively. A better understanding of these adaptive strategies could enrich our knowledge of the infection biology of relevant tick-borne diseases, contributing to the development of future preventions.


Borrelia burgdorferi , Ixodes , Rickettsia , Tick-Borne Diseases , Animals , Humans , Ixodes/microbiology , Immune Evasion , Tick-Borne Diseases/microbiology
7.
Infect Immun ; 90(5): e0005922, 2022 05 19.
Article En | MEDLINE | ID: mdl-35416705

The Borrelia burgdorferi BB0323 protein undergoes a complex yet poorly defined proteolytic maturation event that generates N-terminal and C-terminal proteins with essential functions in cell growth and infection. Here, we report that a borrelial protease, B. burgdorferi high temperature requirement A protease (BbHtrA), cleaves BB0323 between asparagine (N) and leucine (L) at positions 236 and 237, while the replacement of these residues with alanine in the mutant protein prevents its cleavage, despite preserving its normal secondary structure. The N-terminal BB0323 protein binds BbHtrA, but its cleavage site mutant displays deficiency in such interaction. An isogenic borrelial mutant with NL-to-AA substitution in BB0323 (referred to as Bbbb0323NL) maintains normal growth yet is impaired for infection of mice or transmission from infected ticks. Notably, the BB0323 protein is still processed in Bbbb0323NL, albeit with lower levels of mature N-terminal BB0323 protein and multiple aberrantly processed polypeptides, which could result from nonspecific cleavages at other asparagine and leucine residues in the protein. The lack of infectivity of Bbbb0323NL is likely due to the impaired abundance or stoichiometry of a protein complex involving BB0238, another spirochete protein. Together, these studies highlight that a precise proteolytic event and a particular protein-protein interaction, involving multiple borrelial virulence determinants, are mutually inclusive and interconnected, playing essential roles in the infectivity of Lyme disease pathogens.


Borrelia burgdorferi , Lyme Disease , Animals , Asparagine/metabolism , Bacterial Proteins/metabolism , Leucine/metabolism , Lyme Disease/metabolism , Mice , Peptide Hydrolases/metabolism , Proteolysis , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
8.
Methods Mol Biol ; 2411: 269-286, 2022.
Article En | MEDLINE | ID: mdl-34816411

Arthropod vectors account for a number of animal and human diseases, posing substantial threats to health and safety on a global scale. Ticks are considered as one of the most prominent vectors, as they can parasitize almost any vertebrate class and transmit a multitude of infectious diseases, particularly ones that affect humans and domestic animals. While various tick species elicit different tick-borne infections in specific geographic regions, single species can have widespread effects, such as blacklegged ticks, which are widely distributed across the eastern United States and can transmit a variety of infections, including Lyme borreliosis, anaplasmosis, relapsing fever disease, ehrlichiosis, babesiosis, and Powassan virus disease. Despite increasing awareness about ticks as serious disease vectors, effective vaccines against most tick-borne infections are not available. Previously, the successful development of an anti-tick vaccine for use in veterinary animals was based on an 86-kDa midgut antigen from Rhipicephalus (formerly Boophilus) microplus ticks. Herein we describe the fundamentals of vaccine development using protein antigens as model vaccinogen candidates, beginning with the cloning, expression, and purification of recombinant proteins, host immunization, and the assessment of protective efficacy in laboratory settings using a tick-borne murine model of Lyme borreliosis.


Lyme Disease , Tick-Borne Diseases , Vaccines , Animals , Antigens , Humans , Ixodes , Lyme Disease/prevention & control , Mice , Tick-Borne Diseases/prevention & control , United States , Vaccine Development
9.
In Vivo ; 35(6): 3203-3209, 2021.
Article En | MEDLINE | ID: mdl-34697151

BACKGROUND/AIM: Sperm cells are competent to integrate exogenous DNA into their genome. We sought to clarify Human Pappiloma Virus (HPV) internalization in spermatozoa and early preimplantation embryos. MATERIALS AND METHODS: Sperm was incubated with plasmid vectors containing the complete genome of human HPV 16 and HPV 18 tagged with the green fluorescent protein (GFP) gene, to investigate HPV 16 and HPV 18 integration in mouse spermatozoa. Oocytes were in vitro fertilized with preincubated spermatozoa to investigate HPV 16 and HPV 18 potential transfer to mouse embryos. RESULTS: Spermatozoa were able to internalize constructs of cloned high-risk HPV either as integrated or as episomal DNA. Constructs of cloned HPV can also be transferred to mouse embryos, through in vitro fertilization of the oocytes by mouse spermatozoa. CONCLUSION: Viral DNA transmission to the early mouse embryo via sperm, highlights the effect of HPV in reproductive cells and preimplantation development.


Human papillomavirus 18 , Spermatozoa , Animals , Blastocyst , DNA, Viral , Human papillomavirus 16/genetics , Human papillomavirus 18/genetics , Male , Mice
10.
Mol Microbiol ; 116(5): 1241-1248, 2021 11.
Article En | MEDLINE | ID: mdl-34570926

Ticks are regarded as one of the most ancient, unique, and highly evolved ectoparasites. They can parasitize diverse vertebrates and transmit a number of widespread infections. Once acquired from infected hosts, many tick-borne pathogens, like Borrelia burgdorferi, are confined within the tick gut lumen and are surrounded by discrete gut barriers. Such barriers include the peritrophic membrane (PM) and the dityrosine network (DTN), which are in close contact with resident microbiota and invading pathogens, influencing their survival within the vector. Herein, we review our current state of knowledge about tick-microbe interactions involving the PM and DTN structures. As a model, we will focus on Ixodes ticks, their microbiome, and the pathogen of Lyme disease. We will address the most salient findings on the structural and physiological roles of these Ixodes gut barriers on microbial interactions, with a comparison to analogous functions in other model vectors, such as mosquitoes. We will distill how this information could be leveraged towards a better understanding of the basic mechanisms of gut biology and tick-microbial interactions, which could contribute to potential therapeutic strategies in response to ticks and tick-borne infections.


Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiology , Host-Pathogen Interactions , Ixodes/microbiology , Ixodes/physiology , Lyme Disease/microbiology , Animals , Gastrointestinal Microbiome
11.
Trends Immunol ; 42(7): 554-574, 2021 07.
Article En | MEDLINE | ID: mdl-34074602

Ticks have an unparalleled ability to parasitize diverse land vertebrates. Their natural persistence and vector competence are supported by the evolution of sophisticated hematophagy and remarkable host immune-evasion activities. We analyze the immunomodulatory roles of tick saliva which facilitates their acquisition of a blood meal from natural hosts and allows pathogen transmission. We also discuss the contrasting immunological events of tick-host associations in non-reservoir or incidental hosts, in which the development of acquired tick resistance can deter tick attachment. A critical appraisal of the intricate immunobiology of tick-host associations can plant new seeds of innovative research and contribute to the development of novel preventive strategies against ticks and tick-transmitted infections.


Ticks , Animals , Humans , Immune Evasion , Immunity , Immunomodulation , Saliva
12.
Sci Rep ; 11(1): 6095, 2021 03 17.
Article En | MEDLINE | ID: mdl-33731754

Ixodes scapularis ticks transmit multiple pathogens, including Borrelia burgdorferi sensu stricto, and encode many proteins harboring epidermal growth factor (EGF)-like domains. We show that I. scapularis produces multiple orthologs for Bm86, a widely studied tick gut protein considered as a target of an anti-tick vaccine, herein termed as Is86. We show that Is86 antigens feature at least three identifiable regions harboring EGF-like domains (termed as EGF-1, EGF-2, and EGF-3) and are differentially upregulated during B. burgdorferi infection. Although the RNA interference-mediated knockdown of Is86 genes did not show any influences on tick engorgement or B. burgdorferi sensu stricto persistence, the immunization of murine hosts with specific recombinant EGF antigens marginally reduced spirochete loads in the skin, in addition to affecting tick blood meal engorgement and molting. However, given the borderline impact of EGF immunization on tick engorgement and pathogen survival in the vector, it is unlikely that these antigens, at least in their current forms, could be developed as potential vaccines. Further investigations of the biological significance of Is86 (and other tick antigens) would enrich our knowledge of the intricate biology of ticks, including their interactions with resident pathogens, and contribute to the development of anti-tick measures to combat tick-borne illnesses.


Antibodies/immunology , Arthropod Proteins/immunology , Borrelia burgdorferi/immunology , Feeding Behavior , Ixodes/immunology , Lyme Disease/immunology , Animals , Mice
13.
Cell Microbiol ; 23(2): e13275, 2021 02.
Article En | MEDLINE | ID: mdl-33006213

The peritrophic matrix (PM) is an acellular membrane that covers the gut epithelium in arthropods and physically separates it from the lumen. The structure is thought to play an important role in tick biology. The PM is also known to impact the persistence of tick-borne pathogens like Borrelia burgdorferi, although limited information is available about its molecular constituents or their biological significance. Herein, we characterise a novel PM-associated gut protein in Ixodes scapularis ticks, annotated as Peritrophic Membrane Chitin Binding Protein (PM_CBP), for its role in the integrity and function of the matrix. The PM_CBP displays homology to the chitin deacetylase metalloenzyme, shows upregulation during tick feeding, and is localized at the luminal surface of the gut epithelium. The structural integrity of the PM was impaired both by the knock down of PM_CBP expression via RNA interference and by treatment with anti-PM_CBP antibodies, as revealed by its electron microscopic appearance. Additionally, the duration of tick engorgement on mice and the passage of experimentally-inoculated fluorescent dextran molecules across the PM are affected by the knock down of PM_CBP expression. The transfer of anti-PM_CBP antibodies into the tick gut impacted the overall composition of the resident microbiome, and also influenced B. burgdorferi acquisition in ticks and its transmission to mice. Taken together, these data highlight the biological significance of the Ixodes PM and suggest that the targeting of its molecular constituents may contribute to the development of novel interventions against tick-borne infections.


Arthropod Proteins/metabolism , Borrelia burgdorferi/physiology , Gastrointestinal Microbiome , Host-Pathogen Interactions , Ixodes/metabolism , Ixodes/microbiology , Lyme Disease/microbiology , Animals , Borrelia burgdorferi/pathogenicity , Carrier Proteins/metabolism , Chitin/metabolism , DNA, Bacterial , Female , Gene Knockdown Techniques , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C3H , Protein Binding , RNA Interference , RNA, Ribosomal, 16S
14.
Trends Genet ; 37(1): 8-11, 2021 01.
Article En | MEDLINE | ID: mdl-33020021

Ticks exist across diverse environments and transmit numerous pathogens. Due to their long and unique life cycles, these arthropods likely evolved robust epigenetic mechanisms that provide sustainable responses and buffers against extreme environmental conditions. Herein, we highlight how the study of the epigenetic basis of tick biology and vectorial capacity will enrich our knowledge of tick-borne infections.


Arthropod Vectors/growth & development , Disease Transmission, Infectious , Epigenesis, Genetic , Host-Pathogen Interactions , Tick-Borne Diseases/epidemiology , Ticks/physiology , Animals , Arthropod Vectors/classification , Arthropod Vectors/genetics , Humans , Tick-Borne Diseases/genetics , Tick-Borne Diseases/transmission , Ticks/microbiology , Ticks/parasitology , Ticks/virology
15.
Curr Issues Mol Biol ; 42: 113-144, 2021.
Article En | MEDLINE | ID: mdl-33289683

Borrelia burgdorferi sensu lato causes Lyme borreliosis in a variety of animals and humans. These atypical bacterial pathogens are maintained in a complex enzootic life cycle that primarily involves a vertebrate host and Ixodes spp. ticks. In the Northeastern United States, I. scapularis is the main vector, while wild rodents serve as the mammalian reservoir host. As B. burgdorferi is transmitted only by I. scapularis and closely related ticks, the spirochete-tick interactions are thought to be highly specific. Various borrelial and arthropod proteins that directly or indirectly contribute to the natural cycle of B. burgdorferi infection have been identified. Discrete molecular interactions between spirochetes and tick components also have been discovered, which often play critical roles in pathogen persistence and transmission by the arthropod vector. This review will focus on the past discoveries and future challenges that are relevant to our understanding of the molecular interactions between B. burgdorferi and Ixodes ticks. This information will not only impact scientific advancements in the research of tick- transmitted infections but will also contribute to the development of novel preventive measures that interfere with the B. burgdorferi life cycle.


Arachnid Vectors/microbiology , Borrelia burgdorferi , Host-Pathogen Interactions , Lyme Disease/microbiology , Lyme Disease/transmission , Ticks/microbiology , Animals , Arachnid Vectors/growth & development , Humans , Ixodes/microbiology , Life Cycle Stages , Lyme Disease/epidemiology , Lyme Disease/prevention & control , Northwestern United States/epidemiology , Ticks/growth & development
16.
Mol Biol Rep ; 47(11): 8711-8726, 2020 Nov.
Article En | MEDLINE | ID: mdl-33079326

The aim of the present study was to test whether inhibition of ovarian primordial follicles and subsequent activation can be achieved by transient mTOR inhibition. In this preclinical investigation, forty-five female immature Wistar rats were randomized in 5 groups. The control group received subcutaneous saline injections. The other groups received Everolimus, Everolimus plus Verapamil, Everolimus plus Fisetin, and Fisetin alone. Primary and secondary outcomes were measured in the left ovary after a treatment period of 8 weeks. Ten days later, animals received 35 IU FSH for 4 days and 35 IU of hCG on the 5th day. The same parameters were examined in the right ovary. AMH, estradiol, and progesterone levels were assessed at the end of both interventions. Significantly, more primordial and less atretic follicles were observed in the Everolimus plus Verapamil group. AMH and progesterone levels were substantially lower in the Everolimus group. Interestingly, after ovarian stimulation higher levels of AMH and progesterone were observed in the Everolimus plus Verapamil group. Immunoblot analysis of ovarian extracts revealed that the administration of Everolimus led to a significant reduction in the mTORC1-mediated phosphorylation of the 70-kDa ribosomal protein S6 kinase 1. This decrease was reversed in the presence of FSH after stopping drug administration. The expression of the anti-apoptotic molecule Bcl2 as well as of LC3-II and ATG12 was increased after removal of the Everolimus plus Verapamil combination, indicating reduced apoptosis and increased autophagy, whereas the levels of the proliferation marker PCNA in the granulosa cells were elevated, consistent with initiation of follicular growth.Thus, the combination of Everolimus plus Verapamil is capable of increasing the number of competent primordial follicles while reducing atresia.


Cell Differentiation/drug effects , Everolimus/pharmacology , Fertility Preservation/methods , Ovarian Follicle/drug effects , Verapamil/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Drug Evaluation, Preclinical , Female , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Ovarian Follicle/cytology , Rats , Rats, Wistar
17.
In Vivo ; 34(5): 2269-2276, 2020.
Article En | MEDLINE | ID: mdl-32871749

BACKGROUND/AIM: The expression of reverse transcriptase (RT) in ovaries, testes, gametes and embryos highlights its critical role in cell growth and differentiation. We sought to investigate the effects of the potent RT inhibitor lamivudine in gametogenesis and mouse embryo preimplantation development. MATERIALS AND METHODS: Male and female FVB/N mice were treated with the reverse transcriptase inhibitor Lamivudine for seven consecutive weeks. Following treatment, mouse sperm parameters, testicular and ovarian morphology as well as post-IVF embryo development were evaluated. RESULTS: Lamivudine impaired the sperm parameters and the testicular structure in male mice, the number of primordial germ cells and primary oocytes in ovaries of female mice, and the embryos' morphology and development up to the blastocyst stage during in vitro culture. CONCLUSION: The administration of lamivudine affected the processes of spermatogenesis and oogenesis as well as the in vitro preimplantation development of mouse embryos.


Oocytes , RNA-Directed DNA Polymerase , Animals , Blastocyst , Embryonic Development , Female , Male , Mice , Oogenesis , RNA-Directed DNA Polymerase/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
J Infect Dis ; 221(9): 1438-1447, 2020 04 07.
Article En | MEDLINE | ID: mdl-31758693

Borrelia burgdorferi conserved gene products BB0406 and BB0405, members of a common B. burgdorferi paralogous gene family, share 59% similarity. Although both gene products can function as potential porins, only BB0405 is essential for infection. Here we show that, despite sequence homology and coexpression from the same operon, both proteins differ in their membrane localization attributes, antibody accessibility, and immunogenicity in mice. BB0406 is required for spirochete survival in mammalian hosts, particularly for the disseminated infection in distant organs. We identified that BB0406 interacts with laminin, one of the major constituents of the vascular basement membrane, and facilitates spirochete transmigration across host endothelial cell barriers. A better understanding of how B. burgdorferi transmigrates through dermal and tissue vascular barriers and establishes disseminated infections will contribute to the development of novel therapeutics to combat early infection.


Bacterial Outer Membrane Proteins/immunology , Endothelial Cells/microbiology , Host-Pathogen Interactions , Laminin/metabolism , Lyme Disease/microbiology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/genetics , Borrelia burgdorferi/drug effects , Borrelia burgdorferi/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Gene Expression , Gene Targeting , Genetic Complementation Test , Humans , Mice , Mice, Inbred C3H , Mutation , Protein Binding
19.
Cell Microbiol ; 21(2): e12885, 2019 02.
Article En | MEDLINE | ID: mdl-29934966

Borrelia burgdorferi is the causative agent of Lyme disease that persists in a complex enzootic life cycle, involving Ixodes ticks and vertebrate hosts. The microbe invades ticks and vertebrate hosts in spite of active immune surveillance and potent microbicidal responses, and establishes long-term infection utilising mechanisms that are yet to be unravelled. The pathogen can cause multi-system disorders when transmitted to susceptible mammalian hosts, including in humans. In the past decades, several studies identified a limited number of B. burgdorferi gene-products critical for pathogen persistence, transmission between the vectors and the host, and host-pathogen interactions. This review will focus on the interactions between B. burgdorferi proteins, as well as between microbial proteins and host components, protein and non-protein components, highlighting their roles in pathogen persistence in the mammalian host. A better understanding of the contributions of protein interactions in the microbial virulence and persistence of B. burgdorferi would support development of novel therapeutics against the infection.


Bacterial Proteins/metabolism , Borrelia burgdorferi/pathogenicity , Host-Pathogen Interactions/physiology , Lyme Disease/pathology , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Borrelia burgdorferi/drug effects , Borrelia burgdorferi/genetics , Humans , Ixodes/microbiology , Lyme Disease/drug therapy , Lyme Disease/microbiology , Protein Binding/physiology , Protein Interaction Maps , Virulence , Virulence Factors/genetics
20.
Article En | MEDLINE | ID: mdl-29896452

Although Ixodes scapularis and other related tick species are considered prolific vectors for a number of important human diseases, many aspects of their biology, microbial interactions, and immunity are largely unknown; in particular, how these ancient vectors recognize invading pathogens like Borrelia burgdorferi and influence their persistence. The analysis of the Ixodes genome and a limited set of transcriptomic data have established that ticks encode many components of classical immune pathways; yet at the same time, they lack many key orthologs of these recognition networks. Therefore, whether a given immune pathway is active in Ixodes ticks and how precisely they exert its microbicidal functions are only incompletely delineated. A few recent studies have suggested that classical pathways like the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) as well as immunodeficiency (IMD) pathways are fully functional in I. scapularis, and upon challenge with microbes, generate potent microbicidal responses against diverse tick-borne pathogens including B. burgdorferi. These studies also highlight novel concepts of vector immunity that include both a direct and an indirect mode of recognition of pathogens, as well as the influence of the gut microbiome, which ultimately dictates the outcome of a robust microbicidal response. Further understanding of how Ixodes ticks recognize and suppress invading microbes like B. burgdorferi will enrich our fundamental knowledge of vector immunobiology, thereby contributing to the development of future interventions to better control the tick-borne pathogen.


Borrelia burgdorferi/immunology , Immunity , Ixodes/immunology , Lyme Disease/immunology , Animals , Anti-Infective Agents , Arachnid Vectors/genetics , Arachnid Vectors/immunology , Arachnid Vectors/microbiology , Borrelia burgdorferi/pathogenicity , Gastrointestinal Microbiome/immunology , Genome, Insect , Host-Pathogen Interactions/immunology , Ixodes/genetics , Ixodes/microbiology , Lyme Disease/microbiology , Signal Transduction , Transcriptome , Tyrosine/analogs & derivatives , Tyrosine/genetics , Tyrosine/immunology , Tyrosine/metabolism
...