RTN4R Human

Reticulon 4 Receptor Human Recombinant
Shipped with Ice Packs
In Stock

Description

Schizophrenia (SCZ) and Autism Spectrum Disorder (ASD)

Study TypeKey Findings
Common variantsWeak sex-specific association with SCZ in Afrikaner populations
Rare variantsMissense mutations (R292H, D259N, R68H, V363M) linked to SCZ/ASD risk
Chromosomal22q11.2 deletion carriers exhibit 20–30% SCZ prevalence

A 2017 study identified the R292H variant in RTN4R as significantly associated with schizophrenia (P = 0.048) . Functional assays revealed this mutation disrupts growth cone formation, a critical process in neurodevelopment .

Rare Missense Mutations in RTN4R

VariantPopulationClinical AssociationFunctional ImpactSource
R292HSCZ patientsSchizophrenia susceptibilityAltered growth cone dynamics
D259NSCZ patientsSCZ riskUnknown (no functional data)
V363MASD patientsAutism susceptibilityUnknown (no functional data)

The R292H mutation is hypothesized to impair RTN4R’s ability to interact with co-receptors or ligands, leading to dysregulated neuronal signaling .

Rtn4r-Deficient Mice Studies

PhenotypeObservationClinical Relevance
Locomotor activityReduced activity in heterozygotesMotor deficits in SCZ/ASD
Sensorimotor gatingNormal prepulse inhibition (PPI)No direct SCZ link
Cognitive tasksNormal working memory performanceLimited role in SCZ-related deficits

While Rtn4r knockout mice show motor abnormalities, they do not replicate core SCZ-related behavioral deficits (e.g., impaired PPI or cognition) . This suggests RTN4R may modulate susceptibility indirectly, potentially through interactions with NMDA receptor dysfunction during development .

Targeting RTN4R in Neurological Disorders

ApproachMechanismPotential Applications
Antibody blockadeNeutralizes RTN4R-Nogo interactionPromote axonal regeneration post-spinal injury
Gene therapyCorrecting R292H or other pathogenic variantsTreating SCZ/ASD with rare mutations
ROCK inhibitorsDownstream pathway modulationReducing cytoskeletal instability

RTN4R inhibitors are under investigation for spinal cord injury and stroke, while genetic studies aim to identify therapeutic targets in psychiatric disorders .

Key Research Gaps and Future Directions

  1. Mechanistic clarity: Full pathway elucidation (e.g., RTN4R-p75 interaction dynamics).

  2. Population diversity: Limited data in non-European cohorts.

  3. Therapeutic translation: Assessing RTN4R inhibitors in human trials.

Product Specs

Introduction
Reticulon 4 Receptor, also known as RTN4R, functions as a receptor for RTN4, OMG, and MAG. This glycosylphosphoinositol (GPI)-anchored protein belongs to the Nogo receptor family and is primarily found on neurons and their axons. RTN4R plays a crucial role in regulating axonal regeneration and plasticity within the adult central nervous system, making it a promising drug target for treating various neurological conditions. These conditions include spinal cord injury, central nervous system lesions, stroke, and Alzheimer's disease.
Description
Produced in Sf9 Baculovirus cells, RTN4R is a single, glycosylated polypeptide chain consisting of 429 amino acids (27-447 a.a.). With a molecular mass of 46.3 kDa, it appears at approximately 40-57 kDa on SDS-PAGE analysis. This protein is expressed with an 8 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The RTN4R protein solution is provided at a concentration of 0.5 mg/ml and is formulated in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for extended storage periods. Avoid repeated freezing and thawing cycles.
Purity
The purity of RTN4R is greater than 90.0% as determined by SDS-PAGE analysis.
Synonyms
Reticulon 4 Receptor, Nogo-66 Receptor, Nogo Receptor, NOGOR, NGR , Reticulon-4 Receptor, UNQ330/PRO526.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
CPGACVCYNE PKVTTSCPQQ GLQAVPVGIP AASQRIFLHG NRISHVPAAS FRACRNLTIL WLHSNVLARI DAAAFTGLAL LEQLDLSDNA QLRSVDPATF HGLGRLHTLH LDRCGLQELG PGLFRGLAAL QYLYLQDNAL QALPDDTFRD LGNLTHLFLH GNRISSVPER AFRGLHSLDR LLLHQNRVAH VHPHAFRDLG RLMTLYLFAN NLSALPTEAL APLRALQYLR LNDNPWVCDC RARPLWAWLQ KFRGSSSEVP CSLPQRLAGR DLKRLAANDL QGCAVATGPY HPIWTGRATD EEPLGLPKCC QPDAADKASV LEPGRPASAG NALKGRVPPG DSPPGNGSGP RHINDSPFGT LPGSAEPPLT AVRPEGSEPP GFPTSGPRRR PGCSRKNRTR SHCRLGQAGS GGGGTGDSEG SLEHHHHHH.

Q&A

What is the molecular structure of human RTN4R and how does it function at the cellular level?

Human RTN4R (also known as Nogo receptor or Nogo-66 receptor) is a glycosylphosphatidylinositol-linked (GPI-linked) cell surface molecule. The canonical protein has a length of 473 amino acid residues and a mass of 50.7 kDa, with its subcellular localization in the cell membrane . RTN4R forms a heteromeric receptor complex with either LINGO-1 and p75, or LINGO-1 and TROY (a tumor necrosis factor receptor family member) .

At the functional level, activation of RTN4R initiates a signaling cascade that leads to the activation of RhoA and, ultimately, the inhibition of axonal growth . This mechanism is particularly significant in the context of central nervous system injury and regeneration. RTN4R acts as a convergence point for three separate factors that inhibit neurite outgrowth and regeneration, making it a potential target for therapeutic intervention following CNS injury .

Where is RTN4R expressed in the human brain and how does its expression pattern relate to its function?

RTN4R is widely expressed throughout the human brain, with the highest levels detected in the gray matter . This expression pattern is consistent with its role in regulating neuronal growth and plasticity. The receptor is known to bind multiple ligands, including three gene products of the RTN4 gene (NoGo-A, B, and C), as well as oligodendrocyte myelin glycoprotein (OMgp) and myelin-associated glycoprotein (MAG) .

The predominant expression in gray matter suggests a primary role in neuronal function rather than in white matter or glial cells. This distribution pattern supports RTN4R's involvement in synaptic plasticity and neuronal remodeling, processes that are particularly active in gray matter regions of the brain.

What post-translational modifications affect RTN4R function and how can they be detected experimentally?

RTN4R undergoes several post-translational modifications, with the most significant being O-glycosylation and N-glycosylation . These modifications can affect protein folding, stability, and ligand-binding properties of the receptor.

Experimental detection methods:

  • Western Blot Analysis: This is one of the most widely used applications for detecting RTN4R and its modifications . Researchers can use specific antibodies that recognize either the core protein or glycosylated forms.

  • ELISA (Enzyme-Linked Immunosorbent Assay): Another common application for quantitative detection of RTN4R and its modified forms .

  • Mass Spectrometry: For detailed characterization of glycosylation sites and patterns.

  • Deglycosylation Enzymes: Treatment with enzymes like PNGase F (for N-glycans) or O-glycosidase (for O-glycans) followed by SDS-PAGE can reveal shifts in molecular weight corresponding to the removal of glycan moieties.

These post-translational modifications are critical for proper folding and trafficking of RTN4R to the cell membrane, as well as for its interactions with ligands and co-receptors in the signaling complex.

What are the known genetic variants of RTN4R in humans and how do they relate to neuropsychiatric disorders?

Several genetic variants of RTN4R have been identified through sequencing studies, with particular interest in their association with schizophrenia and autism spectrum disorder. Through comprehensive mutation screening, researchers have discovered four rare (minor allele frequency <1%) missense mutations in RTN4R: R68H, D259N, R292H, and V363M .

Among these variants, R292H has been found to be significantly associated with schizophrenia (P=0.048) . Functional assays have demonstrated that this mutation affects the formation of growth cones, providing a potential mechanistic link to neurodevelopmental abnormalities associated with schizophrenia .

Additionally, common polymorphisms in RTN4R have been investigated for association with schizophrenia in different populations, with some studies showing weak sex-specific evidence for association . For example, SNPs rs665780, rs701427, and rs696880 have shown varying degrees of association in different populations .

How does the RTN4R gene relate to the 22q11.2 microdeletion syndrome and its associated psychiatric symptoms?

The human RTN4R gene is located within the 22q11.2 locus, where relatively common hemizygous microdeletions occur at a frequency of 1 in 5000 live births . These deletions are primarily de novo events occurring on different haplotype backgrounds .

Patients with 22q11.2 deletion syndrome display a pattern of cognitive impairments and behavioral deficits, including deficits in working memory, conflict monitoring, visuospatial short-term memory, and executive visual attention . The possibility that RTN4R deficiency contributes to the psychiatric symptoms associated with the 22q11.2 microdeletion is supported by preliminary human genetic and gene expression studies .

While several genes within the 22q11.2 region have been implicated in schizophrenia susceptibility (primarily PRODH, ZDHHC8, and COMT), the potential role of RTN4R is intriguing given its function in neurodevelopment and synaptic plasticity . The contribution of RTN4R to the neuropsychiatric phenotype of 22q11.2 deletion syndrome requires further investigation through integrative approaches combining human genetics, gene expression analysis, and animal models.

What is the significance of the R292H mutation in RTN4R and what methodologies can be used to study its functional impact?

The R292H mutation in RTN4R has been identified as significantly associated with schizophrenia (P=0.048) . This rare variant appears to have functional consequences that may contribute to the pathophysiology of the disorder.

Methodological approaches to study R292H functional impact:

  • In vitro growth cone assays: These have already demonstrated that the R292H mutation affects growth cone formation , a process critical for proper neuronal development and connectivity.

  • Receptor binding studies: To determine if the mutation alters the binding affinity for RTN4R ligands such as Nogo-A, OMgp, or MAG.

  • RhoA activation assays: Since RTN4R signals through RhoA, measuring changes in RhoA activation in cells expressing the mutant receptor can provide insights into altered signaling.

  • Neuronal morphology analysis: Transfection of primary neurons with wild-type versus R292H mutant RTN4R followed by morphometric analysis of neurite length, branching, and spine density.

  • Animal models: Generation of knock-in mice carrying the R292H mutation to study behavioral phenotypes relevant to schizophrenia and other neuropsychiatric disorders.

The R292H mutation provides a valuable entry point for understanding the molecular mechanisms underlying neurodevelopmental disorders and could potentially serve as a target for therapeutic intervention.

What are the most effective methods for analyzing RTN4R expression and function in neuronal cells?

Expression Analysis:

  • Quantitative RT-PCR: For measuring RTNAR mRNA levels. Studies have shown that RTN4R knockdown does not affect ATXN2 mRNA levels, indicating regulation at the protein level .

  • Western blotting: Widely used for detecting RTN4R protein levels . This method can also be used to assess downstream signaling effects, such as changes in ataxin-2 levels following RTN4R manipulation .

  • Immunohistochemistry/Immunofluorescence: For visualizing the spatial distribution of RTN4R in brain tissue or cultured neurons.

Functional Analysis:

  • Lentiviral shRNA knockdown: Has been successfully used to reduce RTN4R expression in both mouse cortical neurons and human iPSC-derived neurons, resulting in approximately 50% reduction of ataxin-2 levels .

  • Peptide inhibition: The NEP1-40 inhibitor peptide has been shown to cause a dose-dependent reduction of ataxin-2 in both mouse and human neurons , providing a pharmacological approach to manipulate RTN4R function.

  • Growth cone analysis: Essential for studying the effects of RTN4R mutations or manipulations on neuronal morphology and development .

  • RhoA activation assays: Since RTN4R signals through RhoA, measuring its activation is crucial for understanding the functional consequences of RTN4R variants or interventions.

How can researchers effectively design genetic association studies for RTN4R variants in neuropsychiatric disorders?

Designing robust genetic association studies for RTN4R variants requires careful consideration of several methodological aspects:

  • Sample Selection: Previous studies have utilized large samples of unrelated individuals (e.g., 1716 schizophrenia patients, 382 autism spectrum disorder patients, and 4009 controls) . Family-based samples can also be valuable, as demonstrated in studies with Afrikaner families .

  • SNP Selection: Tag SNPs can be selected to capture common variation across the RTN4R gene. In European populations, five SNPs (rs665780, rs701427, rs854971, rs696880, and rs1567871) have been used to tag common SNPs in RTN4R with an average R² of 0.75 .

  • Linkage Disequilibrium Analysis: Assessing LD patterns is important for interpreting association results. For example, SNPs rs701427, rs854971, and rs696880 have been found to be in high LD with each other (R²≥0.90) .

  • Sex-Stratified Analysis: Given previous reports of sex-specific associations between RTN4R variants and schizophrenia, conducting both combined and sex-stratified analyses is recommended .

  • Rare Variant Analysis: For rare variants, targeted sequencing of RTN4R exons is necessary, as done in studies that identified rare missense mutations R68H, D259N, R292H, and V363M .

  • Functional Validation: Following identification of associated variants, functional assays (as described in previous sections) should be performed to establish biological relevance.

What in vitro and in vivo models are most suitable for studying RTN4R function in neurological and psychiatric disorders?

In Vitro Models:

  • Primary Neuronal Cultures: Mouse cortical neurons have been successfully used to study RTN4R function and its effects on proteins like ataxin-2 .

  • Human iPSC-derived Neurons (iNeurons): Provide a human-specific model system that has been used to confirm findings from rodent models, particularly regarding the effects of RTN4R knockdown or inhibition on ataxin-2 levels .

  • Neuroblastoma Cell Lines: SH-SY5Y cells have been used as a model system for studying RTN4R knockdown effects on ataxin-2 levels .

In Vivo Models:

  • RTN4R-deficient Mice: Studies have identified a haploinsufficient effect of Rtn4r on locomotor activity, though with normal performance in schizophrenia-related behavioral tasks . These models can be valuable for studying the long-term behavioral effects of RTN4R deficiency.

  • Conditional Knockout Models: Allow for tissue-specific and temporally controlled deletion of RTN4R, which can help distinguish developmental versus adult functions of the receptor.

  • Knock-in Models of Specific Variants: Generation of mice carrying specific mutations (e.g., R292H) can provide insights into the effects of these variants on brain development and behavior.

  • NMDA Receptor Hypofunction Models: RTN4R deficiency has been shown to modulate the long-term behavioral effects of transient postnatal NMDA receptor hypofunction , suggesting interaction models as valuable tools.

How does RTN4R interact with other signaling pathways in neuronal development and neurodegeneration?

RTN4R interacts with multiple signaling pathways that regulate neuronal development, plasticity, and degeneration:

  • RhoA Signaling: The primary downstream effector of RTN4R activation is RhoA, which inhibits axonal growth by regulating cytoskeletal dynamics .

  • Ataxin-2 Regulation: RTN4R appears to regulate ataxin-2 protein levels, with RTN4R knockdown reducing ataxin-2 levels in both mouse and human neurons . This regulation occurs at the protein level, as RTN4R knockdown does not affect ATXN2 mRNA levels .

  • TDP-43 and Stress Granule Formation: As a functional readout of decreased ataxin-2 function, RTN4R knockdown has been shown to reduce recruitment of TDP-43 to stress granules , suggesting a role in RNA metabolism and stress response.

  • NMDA Receptor Signaling: Evidence suggests that RTN4R deficiency can modulate the long-term behavioral effects of transient postnatal NMDA receptor hypofunction , indicating interaction with glutamatergic signaling.

  • Additional Ligand Interactions: Beyond its canonical ligands (Nogo-A, OMgp, MAG), RTN4R also interacts with BAI adhesion-GPCRs, LGI1, BLyS, and LOTUS , suggesting integration with multiple signaling networks.

Understanding these pathway interactions is crucial for developing targeted therapeutic approaches for neurological and psychiatric disorders involving RTN4R dysfunction.

What is the potential of targeting RTN4R for therapeutic interventions in neurodegenerative diseases like ALS?

RTN4R has emerged as a potential therapeutic target for neurodegenerative diseases, particularly Amyotrophic Lateral Sclerosis (ALS) and Spinocerebellar Ataxia Type 2 (SCA2):

  • Ataxin-2 Reduction Strategy: RTN4R knockdown or inhibition reduces ataxin-2 protein levels . Since reducing ATXN2 is a validated method for rescuing degeneration phenotypes in mouse models of TDP-43 toxicity, motor neuron degeneration, and SCA2 , targeting RTN4R represents a potential upstream approach to achieve this therapeutic effect.

  • Peptide Inhibitor Approach: NEP1-40, a peptide inhibitor of RTN4R, has shown dose-dependent reduction of ataxin-2 in both mouse cortical neurons and human iNeurons . This provides a pharmacological approach that could be developed into a therapeutic intervention.

  • Clinical Translation: Clinical trials are already underway to test a NoGo-Receptor inhibitor in spinal cord injury (ClinicalTrials.gov identifier: NCT03989440) . Lessons learned from this trial could aid in testing similar approaches for ALS and SCA2.

  • Genetic Validation: Human genetics has provided compelling validation for ATXN2 as a therapeutic target for both ALS and SCA2 , strengthening the rationale for pursuing RTN4R-targeted approaches.

The effectiveness of RTN4R knockdown in rescuing degeneration in mouse models of ALS remains to be fully tested, but the existing evidence provides a strong foundation for future investigations in this direction.

How can advanced genomic and proteomic approaches enhance our understanding of RTN4R function in human neurological disorders?

Advanced genomic and proteomic approaches offer powerful tools for elucidating RTN4R function in neurological disorders:

  • Whole-Exome/Genome Sequencing: Can identify rare variants in RTN4R with potential functional consequences. Previous studies have already discovered rare missense mutations (R68H, D259N, R292H, V363M) through targeted sequencing , but more comprehensive approaches may reveal additional variants.

  • Single-Cell Transcriptomics: Can provide insights into cell-type-specific expression patterns of RTN4R and its signaling partners in human brain tissue, potentially revealing unique vulnerabilities in specific neuronal populations.

  • Proteomics of RTN4R Complexes: Immunoprecipitation followed by mass spectrometry can identify novel interacting partners of RTN4R, expanding our understanding of its signaling networks.

  • Post-Translational Modification Analysis: Advanced mass spectrometry techniques can characterize the glycosylation patterns and other modifications of RTN4R, which may be altered in disease states.

  • CRISPR-Based Genomic Screening: Genome-wide CRISPR screens in neuronal cells can identify genes that modulate RTN4R function or that show synthetic lethality with RTN4R mutations, revealing potential therapeutic targets.

  • Patient-Derived Cellular Models: iPSC-derived neurons from patients with RTN4R mutations or disorders like schizophrenia and ALS can be analyzed using multi-omics approaches to understand disease-specific alterations in RTN4R signaling.

Integration of these advanced technologies can provide a more comprehensive understanding of RTN4R's role in human neurological disorders and identify novel therapeutic opportunities.

What are the most important considerations when designing antibodies and reagents for RTN4R research?

When designing antibodies and reagents for RTN4R research, several key considerations should be addressed:

  • Epitope Selection: RTN4R undergoes O-glycosylation and N-glycosylation , so antibodies should target epitopes that are not affected by these modifications unless specifically designed to distinguish glycosylated forms.

  • Species Cross-Reactivity: RTN4R orthologs have been reported in mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken species . Researchers should consider whether cross-species reactivity is desired or problematic based on their experimental design.

  • Isoform Specificity: Ensure antibodies can distinguish RTN4R from related family members or potential splice variants.

  • Application Validation: Antibodies should be validated for specific applications such as Western Blot and ELISA, which are commonly used for RTN4R detection .

  • Peptide Inhibitors: When designing peptide inhibitors like NEP1-40, considerations should include stability, cell permeability, and potential off-target effects.

  • shRNA/siRNA Design: For knockdown experiments, RNA interference tools should be carefully designed to maximize specificity and efficiency. Previous studies have successfully used lentiviral shRNA to achieve approximately 50% reduction of RTN4R in neurons .

What methodological challenges exist in studying RTN4R signaling in human brain tissue and how can they be overcome?

Studying RTN4R signaling in human brain tissue presents several methodological challenges that require specific strategies to overcome:

  • Tissue Preservation: RTN4R is a membrane protein that may be particularly sensitive to post-mortem degradation. Using rapid fixation protocols and controlling for post-mortem interval is critical.

  • Cellular Heterogeneity: The human brain contains diverse cell types with potentially different RTN4R signaling mechanisms. Single-cell approaches or cell type-specific isolation techniques can help address this challenge.

  • Regional Variability: RTN4R is expressed at different levels across brain regions, with highest levels in gray matter . Systematic sampling across multiple brain regions is necessary for comprehensive analysis.

  • Protein-Protein Interactions: Capturing the dynamic interactions between RTN4R and its binding partners in human tissue is challenging. Proximity ligation assays or in situ hybridization combined with immunohistochemistry can help visualize these interactions.

  • Functional Readouts: Assessing RTN4R function in fixed human tissue is limited. Complementary approaches using fresh human tissue slices or patient-derived iPSC neurons can provide functional insights.

  • Disease-Specific Changes: Distinguishing primary RTN4R alterations from secondary changes in disease states requires careful experimental design with appropriate controls and correlation with clinical and genetic data.

How can researchers effectively isolate and characterize RTN4R protein complexes from neural tissues?

Isolating and characterizing RTN4R protein complexes from neural tissues requires specialized techniques:

  • Membrane Protein Extraction: As a GPI-anchored protein, RTN4R requires detergent-based extraction methods optimized for membrane proteins, such as using mild non-ionic detergents (e.g., Triton X-100, CHAPS, or digitonin).

  • Immunoprecipitation Strategies:

    • Standard IP using RTN4R-specific antibodies

    • Tandem affinity purification using tagged RTN4R constructs in cellular or animal models

    • Chemical cross-linking prior to lysis to capture transient interactions

  • Mass Spectrometry Analysis:

    • Label-free quantitative proteomics to identify interacting partners

    • Targeted proteomics to monitor specific known interactions

    • Cross-linking mass spectrometry to map interaction interfaces

  • Proximity-Based Methods:

    • BioID or TurboID approaches, where a biotin ligase is fused to RTN4R to biotinylate proximal proteins

    • APEX2 proximity labeling for electron microscopy-compatible labeling

  • Blue Native PAGE: For preserving and analyzing native protein complexes containing RTN4R

  • Functional Validation:

    • Co-immunoprecipitation followed by Western blotting to confirm specific interactions

    • Microscopy-based techniques such as FRET or BiFC to visualize protein interactions in cells

    • Functional assays to assess the impact of disrupting specific interactions

Product Science Overview

Introduction

Reticulon 4 Receptor (RTN4R), also known as Nogo Receptor, is a protein encoded by the RTN4R gene in humans. This receptor is primarily involved in the inhibition of neurite outgrowth and plays a crucial role in regulating axonal regeneration and plasticity in the adult central nervous system . The receptor is known to interact with several ligands, including Reticulon 4 (Nogo), oligodendrocyte myelin glycoprotein (OMG), and myelin-associated glycoprotein (MAG) .

Preparation Methods

The human recombinant Reticulon 4 Receptor is typically produced using recombinant DNA technology. The process involves the following steps:

  1. Gene Cloning: The RTN4R gene is cloned into an appropriate expression vector.
  2. Transformation: The vector is introduced into a host cell, commonly E. coli or mammalian cells, through a process called transformation.
  3. Expression: The host cells are cultured under conditions that promote the expression of the RTN4R protein.
  4. Purification: The expressed protein is purified using techniques such as affinity chromatography, which ensures the isolation of the recombinant protein with high purity.
Chemical Reactions Analysis

The Reticulon 4 Receptor is involved in several biochemical pathways and interactions:

  • Axonal Growth Inhibition: RTN4R mediates axonal growth inhibition through its interaction with Nogo, OMG, and MAG . This interaction triggers intracellular signaling cascades via the coreceptor NGFR, leading to the activation of Rho and subsequent reorganization of the actin cytoskeleton .
  • Neuronal Plasticity: RTN4R plays a role in regulating neuronal plasticity, which is essential for the formation and maintenance of synapses during brain development .
  • Apoptosis Protection: The receptor protects motoneurons against apoptosis, likely through its interaction with MAG .

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 2025 TheBiotek. All Rights Reserved.