Borrelia OspA

Borrelia Burgdorferi Outer Surface Protein A Recombinant
Shipped with Ice Packs
In Stock

Description

Recombinant Borrelia Burgdorferi (strain B31) OspA produced in E.coli is a non-glycosylated, full-length polypeptide chain having a calculated molecular mass of 27kDa.
Borrelia OspA is expressed with a -6x His tag at N-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Borrelia is a genus of bacteria belonging to the spirochete phylum. These bacteria are responsible for causing borreliosis, a zoonotic vector-borne disease primarily transmitted through ticks and, in some cases, lice, depending on the specific species. Among the 36 recognized species of Borrelia, 12 are known to cause Lyme disease or borreliosis and are spread by ticks. The primary Borrelia species responsible for Lyme disease include Borrelia burgdorferi, Borrelia afzelii, and Borrelia garinii. Members of the Borrelia genus possess a linear chromosome approximately 900 kbp in length, along with numerous linear and circular plasmids ranging in size from 5 to 220 kbp. Unlike typical bacterial plasmids, these plasmids are unique because they contain multiple paralogous sequences, a significant number of pseudogenes, and, in some instances, essential genes. Furthermore, certain plasmids exhibit characteristics suggesting their potential role as prophages.
Description
Recombinant Borrelia Burgdorferi (strain B31) OspA, produced in E.coli, is a full-length polypeptide chain that is not glycosylated and has a calculated molecular mass of 27kDa. This protein is expressed with a -6x His tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
Borrelia OspA is supplied in a buffer solution containing 20mM HEPES (pH 8.0), 200mM NaCl, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Minimize repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Source
Escherichia Coli.

Q&A

What is the molecular structure of OspA and how does it relate to its function?

OspA is a 31 kDa lipoprotein with a unique structure characterized by repetitive antiparallel β topology. X-ray crystallography at 1.9 Å resolution reveals that OspA has an unusual nonglobular region of "freestanding" sheet connecting globular N- and C-terminal domains . Arrays of residues with alternating charges are a predominant feature of the folding pattern in the nonglobular region . The C-terminal domain contains a hydrophobic cavity buried in a positively charged cleft that potentially serves as a binding site for an unknown ligand . This structural organization facilitates OspA's primary function: mediating Borrelia adherence to tick midgut epithelium, which is essential for bacterial colonization and survival within the arthropod vector.

How is OspA expression regulated during the Borrelia life cycle?

OspA exhibits distinct differential expression patterns throughout the Borrelia life cycle. It is abundantly expressed by B. burgdorferi within unfed ticks but is rapidly down-regulated as ticks take a blood meal and as borreliae are transmitted to the mammalian host . This differential regulation occurs through complex molecular mechanisms that respond to environmental cues associated with tick feeding, including temperature changes, pH shifts, and host-derived signals . The down-regulation of OspA during transmission to mammals suggests that its function is primarily required during the tick phase of the bacterial life cycle but may be unnecessary or potentially disadvantageous during early mammalian infection.

What experimental evidence demonstrates OspA's essential role in the tick phase of the Borrelia life cycle?

The essential role of OspA in the tick phase has been definitively demonstrated through genetic knockout studies. Researchers created an OspA/B-deficient mutant of an infectious human isolate of B. burgdorferi (strain 297) and found that while OspA/B function was not required for B. burgdorferi infection of mice or accompanying tissue pathology, it was absolutely essential for bacterial colonization of and survival within tick midguts . The OspA/B-deficient mutant showed dramatically reduced ability to colonize tick midguts compared to wild-type spirochetes . Supporting studies demonstrated that purified recombinant OspA bound to extracts of tick midgut cells, and that adherence of B. burgdorferi to tick midgut epithelium was markedly reduced in the presence of OspA antibodies . These findings collectively establish OspA as a critical molecular determinant for maintaining B. burgdorferi in its natural enzootic life cycle.

How do OspA serotypes vary across different Borrelia species, and what methodologies can characterize this diversity?

OspA exhibits significant diversity across Borrelia species, with multiple serotypes (ST) identified that correlate with different genospecies and geographical distributions. In Europe, four Borrelia species presenting six different OspA serotypes are responsible for the majority of human clinical cases: B. burgdorferi (OspA ST1), B. afzelii (OspA ST2), B. garinii (OspA ST3, OspA ST5, and OspA ST6), and B. bavariensis (OspA ST4) .

Traditional serotyping methods rely on reactivity with monoclonal antibodies, but modern approaches include sequence-based in silico typing. A recent methodology development has established an OspA in silico typing (IST) system that uses next-generation sequencing data to characterize OspA diversity . This approach has identified additional ISTs beyond the classical serotypes (ST1-8), including new OspA variants in B. bavariensis (IST9–10), B. garinii (IST11–12), and other Borrelia species (IST13–17) . The sequence-based approach provides greater resolution and objectivity compared to antibody-based methods.

OspA TypeAssociated Borrelia SpeciesGeographic Prevalence
ST1B. burgdorferi sensu strictoNorth America, Europe
ST2B. afzeliiEurope, Asia
ST3B. gariniiEurope, Asia
ST4B. bavariensisEurope, Asia
ST5B. gariniiEurope, Asia
ST6B. gariniiEurope, Asia
IST9-10B. bavariensisNewly characterized
IST11-12B. gariniiNewly characterized
IST13-17Other Borrelia speciesNewly characterized

What experimental approaches can researchers use to investigate the functional differences between OspA serotypes?

Researchers can employ multiple experimental approaches to investigate functional differences between OspA serotypes:

  • Recombinant protein expression and purification: Expressing different OspA serotypes as recombinant proteins for structural and functional studies .

  • Binding assays: Quantitatively comparing the ability of different OspA variants to bind to tick gut epithelial cells or extracts .

  • Mutagenesis studies: Creating chimeric OspA proteins that combine regions from different serotypes to map the determinants of specific functions .

  • Cross-species colonization experiments: Testing the ability of Borrelia strains expressing different OspA serotypes to colonize various tick species.

  • X-ray crystallography and structural analysis: Determining high-resolution structures of different OspA variants to identify structural differences that may impact function .

  • Surface plasmon resonance: Measuring binding kinetics between different OspA variants and potential ligands or receptors.

  • Tick feeding studies: Examining how different OspA serotypes affect Borrelia persistence during and after tick feeding.

These approaches collectively provide a comprehensive toolkit for understanding how sequence variations in OspA translate into functional differences relevant to Borrelia biology and pathogenesis.

How can researchers develop and validate a new OspA typing methodology?

Developing and validating a new OspA typing methodology requires a systematic approach:

  • Database compilation: Assemble a comprehensive database of known OspA sequences spanning different Borrelia species and geographical origins. For example, recent research compiled over 400 Borrelia genomes encompassing the 4 most common disease-causing genospecies .

  • Sequence analysis: Conduct detailed sequence alignments and phylogenetic analyses to understand the pattern of OspA diversity and identify natural clustering.

  • Boundary definition: Define quantitative boundaries for classification based on sequence similarity percentages that correlate with functional or antigenic differences .

  • Nomenclature development: Establish a clear nomenclature system that accommodates both existing serotypes and newly discovered variants, such as the OspA in silico type (IST) system .

  • Validation against reference strains: Test the new typing method against well-characterized reference strains with known OspA serotypes.

  • Cross-method comparison: Compare results with traditional serotyping methods to establish concordance and identify potential discrepancies.

  • Database integration: Make the typing scheme and associated variants publicly available through databases such as the PubMLST Borrelia spp. database .

  • Standardization of protocols: Develop standardized protocols that ensure reproducibility across different laboratories.

This methodical approach ensures that new typing systems are robust, comprehensive, and broadly applicable for characterization of OspA diversity in research and surveillance settings.

How can researchers generate and validate OspA-deficient mutants in virulent Borrelia strains?

Generating OspA-deficient mutants in virulent Borrelia strains presents significant technical challenges due to the low transformation efficiency of B. burgdorferi and potential essential nature of OspA in certain contexts. Researchers should follow this methodological framework:

  • Construct design: Design allelic exchange vectors containing antibiotic resistance markers flanked by sequences homologous to regions upstream and downstream of the ospA/B operon .

  • Transformation optimization: Optimize transformation conditions specifically for virulent Borrelia strains, which are typically more difficult to transform than laboratory-adapted strains.

  • Mutant verification: Confirm the absence of the ospA gene through both PCR and Southern blot analyses, and verify the absence of OspA protein expression by Western blot .

  • Complementation: Restore OspA expression in mutant strains using a shuttle vector containing the ospA gene under the control of its native promoter to confirm the specificity of observed phenotypes.

  • Phenotypic characterization: Comprehensively characterize the mutant by testing its ability to:

    • Infect mice (spirochete loads in skin, joints, and hearts can be measured by quantitative PCR)

    • Induce pathology (joint swelling and histopathology)

    • Colonize ticks (using both unfed and feeding ticks)

  • Stability assessment: Ensure the stability of the mutant phenotype through multiple passages both in vitro and in vivo.

This approach has been successfully used to create the first OspA/B-deficient mutant from a virulent strain of B. burgdorferi (strain 297), which provided definitive evidence for OspA's essential role in the tick phase of the Borrelia life cycle .

What are the methodological considerations for studying OspA expression during tick-mammal transmission?

Studying OspA expression during the complex process of tick-mammal transmission requires specialized methodological considerations:

  • Tick infection models: Establish laboratory colonies of relevant Ixodes species infected with B. burgdorferi strains of interest, ensuring consistent infection rates for reproducible experiments.

  • Temporal sampling: Collect samples at precise time points during tick feeding (unfed, partially fed at 24h, 48h, and 72h, and fully fed) to capture the dynamic changes in OspA expression.

  • RNA preservation and extraction: Develop protocols for rapid preservation of tick tissues to prevent RNA degradation, and optimize extraction methods to obtain high-quality RNA from small numbers of spirochetes within tick tissues.

  • Quantitative analysis: Employ quantitative RT-PCR with carefully validated reference genes to accurately measure ospA transcript levels during different stages of tick feeding .

  • Protein detection: Use immunofluorescence microscopy with specific anti-OspA antibodies to visualize OspA expression on individual spirochetes within tick tissues during feeding.

  • In situ localization: Combine immunohistochemistry with confocal microscopy to correlate OspA expression with spirochete location within tick tissues and migration patterns during feeding.

  • Single-cell analysis: Consider single-cell RNA sequencing approaches to capture the heterogeneity of OspA expression among individual spirochetes during transmission.

  • Controlled environment variables: Account for environmental factors that might influence expression patterns, including temperature, humidity, and host factors.

These methodological considerations help ensure that researchers capture the true dynamics of OspA expression during the critical transition between tick and mammalian environments.

How can researchers resolve contradictory data regarding OspA's role in late-stage Lyme disease pathology?

Resolving contradictory data regarding OspA's role in late-stage Lyme disease pathology requires a multifaceted methodological approach:

  • Standardized detection methods: Implement highly sensitive and specific methods for detecting OspA expression in tissues during chronic infection, using techniques such as RNAseq, digital droplet PCR, and mass spectrometry-based proteomics.

  • Strain considerations: Systematically compare multiple well-characterized Borrelia strains under identical experimental conditions, as strain-specific differences may explain contradictory findings.

  • Temporal analysis: Conduct longitudinal studies examining OspA expression and immune responses over the entire course of infection through late-stage disease.

  • Tissue-specific expression: Analyze potential differential expression of OspA in different tissues during late-stage infection, as expression may vary by microenvironment.

  • Immune response analysis: Characterize anti-OspA immune responses in depth, including antibody titers, antibody isotypes, T cell responses, and cytokine profiles in both animal models and human patients.

  • Animal models: Develop improved animal models that better recapitulate human chronic Lyme disease pathology, particularly treatment-resistant Lyme arthritis, which has been linked to OspA in some studies .

  • Molecular mimicry investigation: Rigorously test the hypothesis that molecular mimicry between OspA and human leukocyte function-associated antigen-1 may drive autoimmunity in chronic Lyme arthritis .

  • Clinical correlation: Analyze well-characterized patient cohorts with different clinical manifestations of late Lyme disease, correlating OspA expression and anti-OspA immune responses with specific pathologies.

This comprehensive approach can help reconcile conflicting data and develop a more coherent understanding of OspA's potential roles in late-stage Lyme disease pathology.

What methodological approaches are used to design multivalent OspA vaccines targeting diverse Borrelia species?

Designing multivalent OspA vaccines requires sophisticated methodological approaches to address the challenge of OspA serotype diversity:

  • Epitope mapping: Identify protective epitopes within each OspA serotype through techniques such as monoclonal antibody binding studies, hydrogen-deuterium exchange mass spectrometry, and X-ray crystallography of antibody-antigen complexes .

  • Chimeric protein design: Engineer chimeric proteins that combine protective epitopes from multiple OspA serotypes. Recent research has successfully developed chimeric proteins connecting epitopes from OspA ST1, ST2, ST4, and ST5 .

  • Fusion protein strategies: Link chimeric proteins to form fusion constructs that provide broader protection. For example, researchers have created fusion proteins that provide protection against both OspA ST1 and ST2 .

  • Structural analysis: Use structural biology techniques to ensure that protective epitopes maintain their native conformation within chimeric constructs.

  • Immunogenicity optimization: Modify sequences to enhance immunogenicity while preserving protective epitopes, potentially through techniques such as glycosylation engineering or codon optimization.

  • Adjuvant formulation: Develop and test adjuvant formulations that enhance the breadth and durability of immune responses against multiple OspA variants.

  • Conservative design approach: Focus on developing a single recombinant antigen that can provide broad protection, rather than a mixture of multiple proteins, to reduce manufacturing complexity .

This approach has yielded promising results, with proof-of-concept studies demonstrating that multivalent OspA-based vaccines can provide significant protection against both in vitro-grown spirochetes and infected ticks expressing different OspA serotypes .

What experimental protocols are most effective for evaluating OspA vaccine efficacy in preclinical models?

Evaluating OspA vaccine efficacy in preclinical models requires rigorous experimental protocols:

  • Immunization protocols: Establish standardized immunization schedules, typically including primary immunization and one or more booster doses with carefully selected adjuvants.

  • Challenge methods: Employ two complementary challenge approaches:

    • Needle inoculation: Challenge with cultured Borrelia strains expressing relevant OspA serotypes

    • Tick challenge: Use laboratory-infected ticks for a more natural challenge that tests the unique mechanism of OspA vaccines (killing spirochetes in the tick midgut during feeding)

  • Protection assessment: Evaluate protection through multiple complementary methods:

    • Culture recovery: Attempt to culture Borrelia from tissues of challenged animals

    • Quantitative PCR: Measure Borrelia loads in tissues (skin, joints, heart) using qPCR targeting the flaB gene

    • Serological conversion: Test for antibodies against Borrelia antigens not included in the vaccine

  • Pathology evaluation: Assess prevention of disease manifestations:

    • Joint measurements: Measure joint swelling using calipers

    • Histopathology: Conduct histopathological examination of tissues

    • Functional assessments: Evaluate physiological parameters relevant to Lyme disease

  • Immunological analysis: Characterize vaccine-induced immune responses:

    • Antibody titers: Measure serotype-specific antibody responses by ELISA

    • Borreliacidal activity: Assess the ability of immune sera to kill spirochetes in vitro

    • Epitope specificity: Determine which epitopes are recognized by vaccine-induced antibodies

  • Cross-protection studies: Test vaccine efficacy against multiple Borrelia strains expressing different OspA serotypes to assess the breadth of protection.

These protocols collectively provide comprehensive assessment of vaccine immunogenicity, protective efficacy, and potential to prevent both infection and disease manifestations.

How can researchers address the challenges of OspA's down-regulation during transmission in vaccine development?

The down-regulation of OspA during tick feeding and transmission to mammals presents a unique challenge for vaccine development. Researchers can address this through several methodological approaches:

  • Transmission blocking mechanism: Design vaccines explicitly to target the pre-transmission phase by inducing high-titer antibodies that enter the tick midgut during blood feeding and kill spirochetes before they down-regulate OspA and migrate to salivary glands .

  • Antibody kinetics optimization: Develop vaccination protocols that maintain persistently high antibody titers to ensure sufficient antibodies enter the tick midgut during feeding.

  • Combination approaches: Consider combining OspA with antigens expressed during mammalian infection to provide protection through multiple mechanisms.

  • Tick feeding studies: Conduct detailed studies examining the kinetics of OspA down-regulation during tick feeding and the window of opportunity for antibody-mediated killing.

  • Epitope selection: Focus on epitopes that remain accessible for the longest period during the early phases of tick feeding.

  • Adjuvant optimization: Test adjuvants specifically for their ability to induce durable antibody responses of appropriate isotypes for complement-mediated killing in the tick midgut environment.

  • Formulation development: Explore formulations that might enhance antibody entry into the tick midgut during feeding, potentially through interactions with tick midgut proteins.

The fundamental principle of OspA-based vaccines is that anti-OspA antibodies ingested by ticks during blood feeding kill spirochetes in the tick midgut before transmission to the host can occur . Understanding and optimizing this unique mechanism is crucial for effective vaccine development.

How can structural biology approaches advance our understanding of OspA function and vaccine design?

Advanced structural biology approaches offer powerful tools for understanding OspA function and improving vaccine design:

  • High-resolution structural determination: While the crystal structure of OspA has been determined at 1.9 Å resolution , applying newer techniques such as cryo-electron microscopy could provide insights into dynamic aspects of OspA structure and interactions.

  • Structure-function correlation: Systematically map how specific structural features of OspA contribute to its functions by combining structural information with functional assays:

    • The N-terminal domain contains a well-conserved surface that overlaps with certain antibody epitopes

    • The C-terminal domain features a hydrophobic cavity buried in a positively charged cleft that could serve as a binding site for an unknown ligand

  • Molecular dynamics simulations: Employ computational approaches to model the dynamic behavior of OspA in different environments, such as tick midgut versus mammalian tissues.

  • Epitope-focused vaccine design: Use structural information to design immunogens that present key protective epitopes in optimal orientations while eliminating regions that might induce non-protective or potentially harmful immune responses.

  • Receptor-ligand interactions: Identify and characterize interactions between OspA and tick receptors at the molecular level, potentially using techniques such as:

    • Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

    • Single-particle cryo-EM to visualize complexes

    • Surface plasmon resonance to measure binding kinetics

  • Rational stabilization: Apply structure-based protein engineering to stabilize OspA in conformations that optimally present protective epitopes for vaccine purposes.

Understanding the exposed variable region on the C-terminal domain of OspA is particularly important for vaccine design, as this region is predicted to be a significant factor in the worldwide effectiveness of OspA-based vaccines .

What novel methodologies are emerging for studying OspA expression and function at the single-cell level?

Emerging single-cell methodologies offer unprecedented insights into OspA expression and function:

  • Single-cell RNA sequencing (scRNA-seq): Apply scRNA-seq to populations of Borrelia during different life cycle stages to capture the heterogeneity of ospA expression among individual spirochetes.

  • Single-molecule fluorescence in situ hybridization (smFISH): Detect and quantify ospA mRNA transcripts in individual spirochetes within tick tissues.

  • Single-cell proteomics: Develop methods to analyze protein expression in individual Borrelia cells, potentially using techniques such as mass cytometry (CyTOF) adapted for bacterial cells.

  • Live-cell imaging: Employ advanced microscopy techniques such as super-resolution microscopy to visualize OspA distribution and dynamics on the surface of individual live spirochetes.

  • Single-molecule tracking: Track the movement and interactions of individual OspA molecules labeled with quantum dots or fluorescent proteins on the Borrelia surface.

  • Microfluidic approaches: Use microfluidic devices to isolate and analyze individual spirochetes under controlled conditions that mimic different host environments.

  • CRISPR interference (CRISPRi): Apply CRISPRi systems adapted for Borrelia to modulate ospA expression in a controlled manner and observe effects on individual cells.

  • Spatial transcriptomics: Map ospA expression in the context of tick tissues to understand spatial regulation during tick feeding and spirochete migration.

These emerging methodologies promise to reveal previously unappreciated heterogeneity in OspA expression and function that may have important implications for understanding Borrelia pathogenesis and developing more effective interventions.

How can systems biology approaches integrate OspA research with broader understanding of Borrelia pathogenesis?

Systems biology approaches can integrate OspA research into a comprehensive understanding of Borrelia pathogenesis:

  • Multi-omics integration: Combine transcriptomics, proteomics, metabolomics, and genomics data to understand how OspA functions within the broader context of Borrelia biology:

    • Correlate ospA expression with global gene expression patterns

    • Identify metabolic pathways that may be linked to OspA function

    • Map protein-protein interaction networks involving OspA

  • Regulatory network analysis: Elucidate the regulatory networks controlling ospA expression, including transcription factors, small RNAs, and post-transcriptional mechanisms.

  • Host-pathogen interaction modeling: Develop computational models that integrate data on OspA function with host immune responses to predict outcomes of infection under different conditions.

  • Ecological modeling: Incorporate OspA diversity and function into broader models of Borrelia ecology, including tick-host-pathogen interactions across different geographic regions.

  • Comparative genomics: Analyze OspA sequence variation across the Borrelia genus in the context of whole-genome evolution to understand selective pressures and functional constraints.

  • Machine learning approaches: Apply machine learning algorithms to large datasets to identify previously unrecognized patterns linking OspA variation to specific aspects of Borrelia biology or pathogenesis.

  • Network pharmacology: Use network-based approaches to identify potential intervention points that might disrupt OspA function or expression for therapeutic purposes.

These integrative approaches can place OspA research in its broader biological context, potentially revealing emergent properties and unexpected connections that would not be apparent from more focused studies.

Product Science Overview

Introduction

Borrelia burgdorferi is the causative agent of Lyme disease, a tick-borne illness that affects thousands of people annually, particularly in the northern hemisphere. One of the key proteins associated with Borrelia burgdorferi is the Outer Surface Protein A (OspA). This protein has been extensively studied for its role in the pathogenesis of Lyme disease and its potential as a vaccine target.

Structure and Function of OspA

Outer Surface Protein A (OspA) is a lipoprotein found on the surface of Borrelia burgdorferi. It plays a crucial role in the bacterium’s ability to survive and infect its host. OspA is highly expressed when Borrelia burgdorferi resides in the tick vector, Ixodes spp., and is downregulated once the bacterium enters the mammalian host. This stage-specific expression is essential for the bacterium’s adaptation to different environments.

Recombinant OspA

Recombinant OspA refers to the protein produced through recombinant DNA technology. This involves inserting the gene encoding OspA into an expression vector, which is then introduced into a host cell, such as Escherichia coli, to produce the protein. The recombinant OspA can be purified and used for various applications, including vaccine development.

Vaccine Development

The potential of OspA as a vaccine target was first demonstrated in the early 1990s. Studies showed that immunization with recombinant OspA could induce protective antibodies in mice, preventing infection with Borrelia burgdorferi . This led to the development of a human Lyme disease vaccine based on recombinant OspA, known as LYMErix. LYMErix was approved by the FDA in 1998 and was shown to be safe and effective in preventing Lyme disease in adults and adolescents .

However, LYMErix was withdrawn from the market in 2002 due to poor sales and concerns about potential side effects, despite multiple organizations concluding that it was safe . Research on OspA-based vaccines continues, with newer formulations being developed to improve efficacy and safety.

Mechanism of Protection

The protective mechanism of OspA-based vaccines involves the induction of antibodies that target the protein on the surface of Borrelia burgdorferi. These antibodies can neutralize the bacterium, preventing it from establishing an infection in the host. Additionally, passive transfer of OspA-specific antibodies has been shown to protect immunodeficient mice from infection, further supporting the potential of OspA as a vaccine target .

Current Research and Future Directions

Recent research has focused on improving the immunogenicity and safety of OspA-based vaccines. This includes the development of lipidated forms of recombinant OspA, which have been shown to induce stronger immune responses . Additionally, there is a growing interest in developing vaccines that target the tick vector itself, rather than the pathogen, to prevent the transmission of Lyme disease .

In conclusion, Borrelia burgdorferi Outer Surface Protein A (OspA) remains a promising target for Lyme disease vaccine development. Advances in recombinant DNA technology and a better understanding of the protein’s role in the bacterium’s life cycle have paved the way for new and improved vaccines that could significantly reduce the incidence of Lyme disease.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.