Recombinant Pongo abelii Complement C1r subcomponent (C1R), partial

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

C1r is a serine protease that, along with C1q and C1s, forms the C1 complex in the classical complement pathway . In innate immunity, this pathway facilitates pathogen recognition, inflammation, and clearance. The recombinant Pongo abelii C1R is a partial fragment of the full-length protein, expressed in heterologous systems such as E. coli, yeast, or mammalian cells .

Key Features

  • Expression Region: The partial protein spans amino acids 18–705 or specific subregions (e.g., 527–637 aa in some constructs) .

  • Tags: Variants include N-terminal 10xHis-tagged, C-terminal Myc-tagged, or Avi-tagged (for biotinylation) .

  • Molecular Weight: Approximately 19.8 kDa for the truncated form .

  • Purity: >85% as determined by SDS-PAGE .

Functional Domains

The full-length C1r contains:

  1. CUB1-EGF-CUB2-CCP1-CCP2-Activation Peptide domains in the A chain.

  2. Serine Protease Catalytic Domain in the B chain .
    The partial recombinant protein retains critical regions for enzymatic activity or binding assays .

Potential Uses

  • Binding Assays: To study interactions with complement components (e.g., C1s, C1q) .

  • Enzyme Activity: Assessing proteolytic activity using substrates like Z-Gly-Arg-SBzl .

  • Immunological Studies: Investigating complement-mediated immune responses in primates .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default glycerol concentration is 50% and may serve as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
C1RComplement C1r subcomponent; EC 3.4.21.41; Complement component 1 subcomponent r) [Cleaved into: Complement C1r subcomponent heavy chain; Complement C1r subcomponent light chain]
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Pongo abelii (Sumatran orangutan) (Pongo pygmaeus abelii)
Target Names
C1R
Uniprot No.

Target Background

Function
C1r B chain is a serine protease that, in combination with C1q and C1s, forms C1, the first component of the classical complement pathway.
Database Links
Protein Families
Peptidase S1 family

Q&A

What is Recombinant Pongo abelii Complement C1r subcomponent (C1R), partial?

Recombinant Pongo abelii Complement C1r subcomponent (C1R), partial is a serine protease derived from Sumatran orangutan (Pongo pygmaeus abelii) that forms part of the C1 complex in the classical pathway of the complement system. This protein belongs to the peptidase S1 family and is a critical component in initiating complement activation . The recombinant partial protein (UniProt No. Q5R544) is typically produced in expression systems such as yeast to achieve >85% purity as determined by SDS-PAGE . C1r functions by combining with C1q and C1s to form C1, the first component of the classical complement pathway, existing as a proenzyme until activation through binding to immune complexes .

How does C1R function within the complement activation cascade?

C1R functions as a crucial component in the classical pathway of complement activation through several sequential mechanisms. Initially, C1r exists in its proenzyme form, forming part of the C1 complex (C1q-C1r2-C1s2) where two C1r and two C1s molecules arrange in a Ca²⁺-dependent tetramer (C1s-C1r-C1r-C1s) that associates with C1q . When C1q binds to immune complexes (antibodies bound to antigens), a conformational change occurs that triggers C1r autoactivation through cleavage of the Arg463-Ile464 bond in its catalytic domain . Once activated, C1r cleaves and activates its associated C1s molecules, which then proceed to cleave C4 and C2 complement components, leading to the formation of C3 convertase and continuation of the complement cascade . Research has demonstrated that a single active C1r subunit within the C1 complex is sufficient for full activity of the entire complex .

What storage and handling protocols optimize the stability of recombinant C1R?

Proper storage and handling of recombinant C1R is critical for maintaining its stability and biological activity. For liquid formulations, the shelf life is typically 6 months when stored at -20°C/-80°C, while the lyophilized form maintains stability for approximately 12 months at the same temperature range . The recommended reconstitution protocol involves briefly centrifuging the vial prior to opening, reconstituting in deionized sterile water to a concentration of 0.1-1.0 mg/mL, and adding glycerol to a final concentration of 5-50% (typically 50% is recommended) for long-term storage . Working aliquots can be maintained at 4°C for up to one week, but repeated freezing and thawing cycles should be avoided as they can compromise protein integrity and activity . These protocols ensure that the recombinant protein maintains its structural integrity and functional properties for experimental applications.

What are the structural characteristics of C1R that enable its proteolytic function?

C1R possesses distinct structural domains that facilitate its function as a serine protease within the complement system. The protein contains a catalytic domain with serine protease activity, featuring the critical Arg463-Ile464 bond that undergoes cleavage during activation . Upon activation, C1r divides into two chains: a heavy chain and a light chain that remain connected by disulfide bonds . The protein also contains calcium-binding regions essential for forming the C1s-C1r-C1r-C1s tetramer within the C1 complex . Specialized interaction domains allow C1r to associate with C1q and C1s components, enabling proper assembly and function of the C1 complex . Understanding these structural features has been enhanced through the creation of various C1r mutants, including Mutant QI (Arg463 to Gln mutation), which creates a stable, nonactivable zymogen, and Mutants KI (Arg463 to Lys) and RF (Ile464 to Phe), which retain autoactivation capability but with increased stability .

What experimental approaches are most effective for studying C1R activation mechanisms?

Investigating C1R activation mechanisms requires sophisticated experimental approaches that address the protein's unique autoactivation properties. Site-directed mutagenesis studies targeting the Arg463-Ile464 cleavage site have proven particularly valuable—mutating Arg463 to Gln (mutant QI) creates a stable, nonactivable zymogen, while mutations to Lys (mutant KI) or changing Ile464 to Phe (mutant RF) increase stability while preserving autoactivation capability . Functional assessment through hemolytic assays offers critical insights, where recombinant C1r (wild-type or mutant) expressed in controlled systems (e.g., insect cells with serum-free medium) is used to reconstitute C1 complexes, with hemolytic activity serving as an indicator of complement activation .

The following table summarizes the properties of wild-type and mutant C1r variants:

C1r VariantMutationStability as ZymogenAutoactivation CapabilityHemolytic ActivityResearch Applications
Wild-typeNoneLow (spontaneous activation)YesFullReference standard
Mutant QIArg463→GlnHighNoNoneStable zymogen studies, mixed dimer experiments
Mutant KIArg463→LysModerateYesSimilar to wild-typeActivation mechanism studies
Mutant RFIle464→PheModerateYesSimilar to wild-typeActivation mechanism studies

Additionally, dimerization studies utilizing mixed dimers of wild-type and mutant C1r have revealed that one active C1r subunit is sufficient for full complex activity, with exchange of C1r monomers between dimers completing in less than 16 hours at pH 7 and 4°C . These methodologies provide critical insights into C1r's activation dynamics and its role in complement cascade initiation.

How does C1R contribute to kidney fibrosis pathogenesis and what experimental evidence supports this role?

C1R plays a significant role in kidney fibrosis pathogenesis through multiple mechanisms supported by robust experimental evidence. Studies have revealed distinct expression patterns where C1r is normally expressed at higher levels in liver compared to kidney tissue, but following kidney injury (e.g., folic acid administration), C1r mRNA and protein levels become significantly upregulated specifically in kidney tissue . Immunohistochemistry and in situ hybridization have localized this increased expression primarily to renal tubular epithelial cells .

The pathogenic role of C1r in kidney fibrosis has been demonstrated through studies using C1r-null mice, which showed reduced acute tubular injury and inflammation 2 days after folic acid administration, as well as reduced expression of C1s, C3 fragment formation, and organ fibrosis 14 days after treatment compared to wild-type mice . Mechanistically, interferon-γ has been identified as an inducer of both C1r and C1s proteases in renal epithelial cells, with differential gene expression analyses revealing that C1r-null mice show reduced expression of genes associated with acute phase response, complement activation, proliferation of connective tissue cells (e.g., platelet-derived growth factor receptor-β), and inflammation .

Single nuclei RNA sequencing studies have further clarified the spatial distribution of complement components during kidney injury, showing that increased synthesis of complement C3 and C5 occurs primarily in renal tubular epithelial cells (both proximal and distal), while increased expression of complement receptors C3ar1 and C5ar1 occurs in interstitial cells including immune cells like monocytes/macrophages . This compartmentalization suggests a complex intercellular signaling network mediated by complement components in kidney fibrosis development.

What methods are available for reconstituting C1R activity in depleted systems for functional studies?

Reconstituting C1R activity in depleted systems requires precise methodological approaches to ensure valid experimental outcomes. The standard approach utilizes C1r-depleted serum (C1r-Dpl), which is prepared by removing endogenous C1r through immunoaffinity chromatography while preserving all other complement components . This depleted serum serves as an excellent platform for reconstitution experiments, as it maintains a functional alternative pathway while specifically lacking classical pathway activity due to C1r absence .

The reconstitution protocol involves first verifying the absence of C1r in the depleted serum through functional assays for classical pathway activity and protein detection methods such as double immunodiffusion . Researchers can then add purified C1r enzyme or proenzyme at a concentration of 31 µg/mL to restore classical pathway functionality . Quality control measures should include testing reconstituted activity using functional hemolytic assays and verifying that all other complement components necessary for classical pathway activation remain present and active in the depleted serum .

When designing reconstitution experiments, researchers should consider whether their experimental question requires active enzyme or zymogen proenzyme, as C1r naturally exists in serum in its inactive zymogen form and becomes activated only when C1 binds to immune complexes . This reconstitution approach allows for precise control over the C1r variable while maintaining a physiologically relevant context for studying complement activation mechanisms.

What are the molecular mechanisms by which C1R influences tissue inflammation and fibrosis?

The molecular mechanisms by which C1R influences tissue inflammation and fibrosis involve complex interactions between complement activation and tissue repair pathways. C1r initiates the classical pathway of complement activation, leading to C3 fragment formation, which has been directly implicated in fibrosis development . Gene expression analyses in kidney fibrosis models have identified several downstream mechanisms through which C1r promotes pathological tissue remodeling.

C1r deletion in experimental models results in reduced expression of genes associated with the acute phase response, suggesting that C1r activation contributes to the initial inflammatory response to tissue injury . Additionally, C1r influences the expression of genes involved in the proliferation of connective tissue cells, including platelet-derived growth factor receptor-β, a key mediator of fibroblast activation and proliferation . The interplay between C1r and interferon-γ signaling also appears significant, as interferon-γ induces the expression of both C1r and C1s proteases in renal epithelial cells, potentially creating a feed-forward loop that amplifies complement activation in injured tissues .

Spatial studies using single nuclei RNA sequencing have revealed a compartmentalization of complement components during kidney injury, with tubular epithelial cells producing C3 and C5, while interstitial cells express the receptors for these components . This arrangement suggests that C1r-initiated complement activation leads to the production of complement fragments that act as signaling molecules between different cell populations in the injured tissue, coordinating inflammatory and fibrotic responses . These molecular mechanisms highlight C1r as a potential therapeutic target for fibrotic diseases, where inhibiting its activity might disrupt the progression from acute injury to chronic fibrosis.

What controls should be included when designing experiments to study C1R function?

Designing robust experiments to study C1R function requires carefully selected controls to ensure valid and interpretable results. For negative controls, researchers should consider using C1r-depleted serum (C1r-Dpl), which provides a system lacking C1r while maintaining other complement components . In animal studies, C1r-null mice serve as genetic negative controls, while inactive C1r mutants such as QI (Arg463 to Gln) that lack activation capacity can serve as protein-level negative controls .

Positive controls should include purified native C1r to establish baseline activity for comparison with recombinant variants, as well as wild-type recombinant C1r when studying mutant forms . Known C1r activators, such as immune complexes that trigger the classical pathway, can confirm the responsiveness of the experimental system .

Specificity controls are crucial for distinguishing C1r activity from other proteases. These should include related proteases like C1s to confirm assay specificity, selective protease inhibitors that differentiate between complement proteases, and pathway-specific activators and inhibitors to distinguish classical from alternative pathway effects .

Technical controls should address experimental variables such as storage time (comparing freshly prepared versus stored C1r to account for potential activity loss), temperature (assessing activity at different temperatures to optimize experimental conditions), and buffer composition (evaluating the impact of different ionic conditions and pH) . These comprehensive controls ensure that observed effects can be confidently attributed to C1r function rather than experimental artifacts or contributions from other complement components.

What techniques can differentiate between C1R and closely related complement components?

Differentiating between C1R and closely related complement components requires specialized techniques targeting their unique characteristics. Biochemical differentiation approaches include selective depletion of C1r using immunoaffinity chromatography while preserving other complement components, followed by reconstitution studies that add back purified C1r to depleted serum to confirm specific effects . Activity assays employing substrates specific to C1r's serine protease activity provide functional differentiation, while careful attention to the temporal sequence of activation can help distinguish C1r (which activates before C1s) from other components in the cascade .

Molecular differentiation techniques utilize genetic models such as C1r-null mice that specifically lack C1r expression, highly specific antibodies that recognize C1r but not related proteases like C1s, and PCR analysis with primers designed to detect C1r-specific sequences . Domain-specific studies targeting the unique structural elements of C1r and mutational analyses that affect specific functions while preserving others offer additional approaches to distinguishing C1r from related proteins .

Spatial differentiation can be achieved through immunohistochemistry with specific antibodies to localize C1r in tissues, in situ hybridization to detect C1r mRNA expression patterns, and single-cell or single-nuclei RNA sequencing to identify cell types expressing C1r versus other complement components . The combination of these techniques allows researchers to confidently attribute observed effects to C1r rather than related complement components, enabling more precise characterization of C1r's specific roles in biological processes.

How can researchers assess the functional activity of recombinant C1R in experimental systems?

Assessing the functional activity of recombinant C1R in experimental systems requires specialized approaches that target its serine protease activity and role in complement activation. The hemolytic assay represents the gold standard for evaluating C1r functionality, where recombinant C1r is incorporated into reconstituted C1 complexes and tested for its ability to initiate complement-mediated lysis of appropriately sensitized erythrocytes . This assay provides a physiologically relevant measure of C1r's ability to function within the complete complement cascade.

Enzymatic activity assays using specific peptide substrates designed to be cleaved by C1r's serine protease activity offer a more direct assessment of the protein's catalytic function. These assays can measure the kinetics of substrate cleavage to quantify enzymatic activity and can be adapted to high-throughput formats for screening multiple conditions or variants . Researchers can also assess C1r activation state through SDS-PAGE analysis under reducing conditions, which separates the cleavage products generated during activation (heavy and light chains) from the intact proenzyme form .

For more complex biological systems, researchers can evaluate downstream effects of C1r activity by measuring the activation of C1s and subsequent generation of C3 fragments using techniques such as Western blotting, ELISA, or immunohistochemistry . When working with mutant C1r variants, mixed dimer formation with wild-type C1r can be used to evaluate the impact of mutations on C1r function, as demonstrated in studies showing that one active C1r subunit is sufficient for activity of the entire complex . These diverse approaches allow researchers to comprehensively characterize the functional properties of recombinant C1r in various experimental contexts.

What comparative differences exist between human and Pongo abelii C1R that researchers should consider?

Understanding the comparative differences between human and Pongo abelii (Sumatran orangutan) C1R is essential for translational research applications. Human and Pongo abelii C1R share high sequence homology, reflecting their close evolutionary relationship, with the Pongo abelii C1R (UniProt No. Q5R544) maintaining the core functional domains present in human C1R . The fundamental mechanisms of C1r function in the complement system appear conserved between the species, including serine protease activity, role in C1 complex formation, calcium-dependent interactions with C1s, and autoactivation mechanisms .

When designing comparative studies, researchers should consider using parallel experiments with both human and Pongo abelii C1R to identify any species-specific variations in activation kinetics, substrate specificity, or interactions with other complement components. Additionally, when using Pongo abelii C1R as a model for human complement function, validation with human proteins should be included whenever possible to confirm the translational relevance of findings. These considerations ensure that insights gained from studying the orangutan protein can be appropriately applied to understanding human complement biology and related disease processes.

What disease models have demonstrated roles for C1R dysregulation, and what mechanisms are involved?

C1R dysregulation has been implicated in several disease models through distinct pathological mechanisms. In kidney fibrosis models, increased expression of complement C1r in renal tubular epithelial cells plays a significant role in disease progression . The mechanisms involve C1r-mediated activation of the classical complement pathway, leading to increased C3 fragment formation and subsequent inflammatory and fibrotic responses . Studies using C1r-null mice demonstrate reduced acute tubular injury, inflammation, and organ fibrosis following folic acid administration compared to wild-type mice, providing direct evidence for C1r's pathogenic role .

Genetic studies have revealed associations between C1R mutations and Ehlers-Danlos Syndrome, Periodontal Type, 1, highlighting the role of complement components in connective tissue disorders . This unexpected connection between a complement protein and connective tissue integrity suggests broader roles for C1r beyond traditional immune functions. The mechanisms likely involve altered interactions with extracellular matrix components or dysregulated inflammatory responses that affect tissue maintenance and repair .

Molecular studies indicate that dysregulated C1r can contribute to disease through aberrant activation leading to uncontrolled complement activity, altered expression levels influencing inflammatory signaling cascades, or structural alterations affecting autoactivation dynamics . These findings suggest potential therapeutic approaches targeting C1r in various disease contexts. For example, inhibition of C1r might provide beneficial effects in conditions like kidney fibrosis, while recombinant C1r research facilitates the development of specific inhibitors or modulators that could have therapeutic applications .

How can single-cell analysis techniques enhance our understanding of C1R biology in complex tissues?

Single-cell analysis techniques offer powerful approaches for understanding C1R biology in complex tissues by revealing cell-specific expression patterns and functional compartmentalization that would be obscured in bulk tissue analyses. Single nuclei RNA sequencing studies in kidney fibrosis models have demonstrated that increased synthesis of complement C3 and C5 occurs primarily in renal tubular epithelial cells (both proximal and distal), while increased expression of complement receptors C3ar1 and C5ar1 occurs in interstitial cells including immune cells like monocytes/macrophages . This spatial distribution suggests a complex intercellular signaling network mediated by complement components, with C1r-initiated classical pathway activation in epithelial cells influencing surrounding interstitial cell populations .

These techniques can identify previously unrecognized cell populations expressing C1r, reveal dynamic changes in expression during disease progression, and elucidate the complete complement expression profile at single-cell resolution. By combining single-cell RNA sequencing with spatial transcriptomics, researchers can map the physical relationships between C1r-expressing cells and cells expressing other complement components or receptors, providing insights into how complement signaling networks operate within tissue microenvironments .

Additionally, single-cell proteomics approaches, though still emerging, could potentially detect active versus zymogen forms of C1r in individual cells, offering unprecedented insights into the activation state of the complement system at cellular resolution. These advanced technologies are transforming our understanding of complement biology from a primarily serum-based system to a complex network of cellular interactions where local production and activation of components like C1r play crucial roles in tissue homeostasis and pathology .

What strategies can researchers employ to manipulate C1R activity for therapeutic development?

Developing therapeutic strategies targeting C1R activity requires sophisticated approaches that capitalize on our understanding of its structure and function. Direct inhibition strategies include the development of small molecule inhibitors targeting C1r's serine protease activity, antibodies that bind to and neutralize C1r, or peptide-based inhibitors designed to interfere with the autoactivation cleavage site (Arg463-Ile464) . Research using mutant forms like QI (Arg463 to Gln) provides valuable insights for designing inhibitors that stabilize the zymogen form and prevent activation .

Genetic modulation approaches could utilize RNA interference or CRISPR-based technologies to reduce C1r expression in specific tissues. Studies in C1r-null mice demonstrating reduced kidney fibrosis suggest that targeted reduction of C1r in renal tissue could have therapeutic benefits in kidney diseases . Alternatively, delivery of decoy substrates or inhibitory domains could compete with natural substrates or interaction partners of C1r, mitigating its downstream effects without completely blocking its activity.

For diseases associated with C1r deficiency or dysfunction (rather than overactivity), enzyme replacement therapies using recombinant C1r could potentially restore complement function. The demonstrated ability to reconstitute classical pathway activity in C1r-depleted serum by adding purified C1r (31 µg/mL) provides proof of concept for such approaches .

Advanced therapeutic strategies might leverage the finding that one active C1r subunit is sufficient for C1 complex activity, suggesting that partial inhibition might be sufficient to modulate complement activation while maintaining basal function . Additionally, targeting the inducers of C1r expression, such as interferon-γ in renal epithelial cells, represents an indirect approach to modulating C1r-mediated pathology in specific disease contexts . These diverse strategies highlight the potential of C1r as a therapeutic target in complement-mediated diseases.

What are the most critical unresolved questions in C1R research?

Despite significant advances in understanding C1R biology, several critical questions remain unresolved. The precise structural rearrangements that occur during C1r autoactivation within the C1 complex remain incompletely characterized, particularly how C1q binding to immune complexes transmits an activation signal to C1r . The recently discovered role of C1r in conditions like kidney fibrosis and Ehlers-Danlos Syndrome raises questions about potential non-canonical functions beyond its established role in complement activation . How C1r interacts with other cellular systems beyond complement, including potential direct effects on extracellular matrix components or cell-surface receptors, requires further investigation.

The cell-specific regulation of C1r expression in different tissues, particularly during disease states, remains poorly understood despite evidence of tissue-specific upregulation in conditions like kidney injury . The evolutionary conservation and diversity of C1r across species, including the similarities and differences between human and Pongo abelii C1r, present opportunities for comparative studies that could reveal crucial functional insights . Additionally, the potential of C1r as a therapeutic target remains to be fully explored, including questions about optimal inhibition strategies and potential side effects of C1r modulation in various disease contexts .

These unresolved questions highlight the need for continued research on C1r biology, utilizing emerging technologies like single-cell analysis, structural biology approaches, and advanced genetic models to further elucidate this protein's complex roles in health and disease. The answers to these questions could potentially transform our understanding of complement biology and open new therapeutic avenues for complement-related disorders.

How might emerging technologies advance our understanding of C1R biology in the coming years?

Emerging technologies promise to revolutionize our understanding of C1R biology in several key areas. Cryo-electron microscopy and advanced structural biology techniques will likely provide unprecedented insights into the conformational changes that occur during C1r activation within the C1 complex, potentially revealing new targetable sites for therapeutic development . Single-cell multi-omics approaches combining transcriptomics, proteomics, and metabolomics at single-cell resolution will further elucidate the cell-specific roles of C1r in complex tissues, building on current findings of compartmentalized expression patterns in kidney injury models .

CRISPR-based genetic engineering technologies enable the creation of more sophisticated model systems, including temporal and cell-type-specific C1r knockout or mutation models that could reveal context-dependent functions of C1r beyond what has been observed in global knockout studies . Advances in protein engineering may facilitate the development of reporter systems where C1r activation state can be monitored in real-time in living cells or tissues, transforming our ability to study dynamic complement activation in complex biological systems.

Computational biology approaches, including machine learning algorithms applied to large-scale genomic and proteomic datasets, could identify previously unrecognized associations between C1r variants and human diseases, expanding our understanding of its pathophysiological roles . Additionally, advances in drug delivery systems may enable targeted modulation of C1r activity in specific tissues, overcoming potential limitations of systemic complement inhibition.

These technological innovations, combined with growing recognition of complement's roles beyond traditional immune functions, position C1r research at the intersection of immunology, tissue biology, and precision medicine. The coming years will likely witness significant advances in our understanding of this fascinating protein and its diverse roles in human health and 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 2025 TheBiotek. All Rights Reserved.