MLEC Human

Malectin Human Recombinant
Shipped with Ice Packs
In Stock

Description

Glycoprotein Quality Control

MLEC participates in:

  • Initial N-glycosylation of nascent polypeptides

  • Retention of misfolded glycoproteins via diglucosylated oligosaccharide recognition

  • Interaction with ribophorin I (RPN1) of the OST complex

Viral Replication Mechanisms

Recent studies demonstrate MLEC's critical role in coronavirus infection:

  • Mouse Hepatitis Virus (MHV): MLEC knockdown (KD) reduces viral titers by 80% (MOI 0.1) and impairs replicase protein production .

  • SARS-CoV-2: MLEC KD suppresses Delta variant replication to 43% of control levels (p < 0.05) .

  • Mechanistic Action: Facilitates viral polyprotein biogenesis through glycan-dependent pathways, validated by OST inhibitor NGI-1 assays .

Table 1: MLEC in Coronavirus Studies

VirusExperimental ModelKey ResultCitation
MHVDBT cells (siRNA KD)80% reduction in viral titer at 10 hpi
SARS-CoV-2HEK293T replicon system57% reduction in Delta variant replication
MHVProteomics analysisGlobal reduction in viral glycoproteins

Therapeutic Implications

MLEC represents a potential broad-spectrum antiviral target due to its:

  • Conserved interaction with CoV non-structural proteins (nsp2/nsp4)

  • MOI-independent suppression of viral replication

  • Synergistic effects with OST inhibitors like NGI-1

Additional Applications

  • Antibody Development: Validated antibodies available for IHC and Western blot (Bio-Techne) .

  • Neurological Research: Altered MLEC-associated glycoforms observed in Alzheimer’s disease brains .

Product Specs

Introduction
MLEC, a member of the malectin family, resides on the endoplasmic reticulum membrane. This carbohydrate-binding protein exhibits a strong affinity for Glc2-N-glycan and plays a crucial role in the early stages of protein N-glycosylation.
Description
Recombinant human MLEC, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 264 amino acids (residues 29-269) with a molecular weight of 29.1 kDa. This protein is fused to a 23-amino acid His-tag at its N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The MLEC solution is formulated in 20 mM Tris-HCl buffer (pH 8.0), 0.15 M NaCl, 1 mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended 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 long-term storage. Repeated freezing and thawing cycles should be avoided.
Purity
The purity of this protein is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Malectin, KIAA0152, Oligosaccharyltransferase Complex Subunit (Non-Catalytic).
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSPGLGVAG VAGAAGAGLP ESVIWAVNAG GEAHVDVHGI HFRKDPLEGR VGRASDYGMK LPILRSNPED QILYQTERYN EETFGYEVPI KEEGDYVLVL KFAEVYFAQS QQKVFDVRLN GHVVVKDLDI FDRVGHSTAH DEIIPMSIRK GKLSVQGEVS TFTGKLYIEF VKGYYDNPKV CALYIMAGTV DDVPKLQPHP GLEKKEEEEE EEEYDEGSNL KKQTNKNRVQ SGPRTPNPYA SDNS

Q&A

What is MLEC and what is its primary function in human cells?

MLEC (Malectin) is a glycoprotein quality control factor that resides in the endoplasmic reticulum (ER) and plays a crucial role in the N-linked glycoprotein biogenesis pathway. It functions as a lectin that recognizes and binds to specific glycan structures, particularly diglucosyl oligosaccharides on newly synthesized glycoproteins. MLEC operates upstream of the calnexin/calreticulin cycle, participating in the early stages of glycoprotein quality control by monitoring proper folding and preventing the progression of misfolded proteins through the secretory pathway. Its primary function involves recognizing and managing glycoproteins as they progress through the ER quality control system, thereby ensuring only properly folded proteins continue to the Golgi apparatus for further processing .

How does MLEC interact with the oligosaccharyltransferase (OST) complex?

MLEC interacts extensively with multiple components of the oligosaccharyltransferase (OST) complex as part of its role in glycoprotein quality control. Proteomics studies have identified several OST complex members (RPN1, RPN2, MAGT1, DDOST, STT3A, STT3B) as significant MLEC-interacting partners. These interactions position MLEC strategically at the site of initial N-glycan transfer to nascent proteins, allowing it to monitor glycoproteins immediately after glycosylation occurs. The physical association between MLEC and the OST complex facilitates the coordinated transfer of newly glycosylated proteins from the OST complex to the quality control machinery, enabling MLEC to recognize and bind diglucosyl oligosaccharides after initial glucose trimming by glucosidase I (MOGS) .

What biochemical methods are used to study MLEC interactions with client glycoproteins?

Researchers employ several specialized biochemical approaches to study MLEC interactions with client glycoproteins. Cross-linking with dithiobis(succinimidyl propionate) (DSP) at concentrations around 0.5 mM is commonly used to capture the transient interactions characteristic of lectin-client relationships. Immunoprecipitation techniques using epitope-tagged MLEC (such as FLAG-tagged MLEC) allow for the isolation of MLEC along with its binding partners. These complexes are then analyzed using tandem mass spectrometry (LC-MS/MS) with quantitative labeling methods like TMTpro to identify and quantify the interactions. Additionally, comparative analysis between mock and experimental conditions helps distinguish between basal interactions and those that change during specific conditions such as viral infection. These methods collectively provide a comprehensive view of MLEC's dynamic interactome in various cellular states .

How does MLEC influence coronavirus replication in human cells?

MLEC significantly promotes coronavirus replication by facilitating viral protein biogenesis, particularly the production of viral replicase proteins. Research indicates that MLEC does not directly affect viral entry or genome release into cells, as demonstrated by experiments showing that MLEC knockdown actually leads to an increase in intracellular positive-sense genomic RNA ((+)gRNA) levels early in infection. Instead, MLEC exerts its pro-viral effects primarily during the translation and processing of viral proteins. Knockdown of MLEC results in reduced levels of viral replicase proteins (such as nsp2) beginning around 6 hours post-infection, with this effect becoming progressively more pronounced at 8 and 10 hours post-infection. This reduction in viral protein production consequently leads to decreased viral genome replication, as evidenced by significant reductions in negative-sense genomic RNA ((-)gRNA) levels, which serve as intermediates for genomic replication .

What effect does MLEC knockdown have on the viral proteome during infection?

MLEC knockdown causes a global reduction in the entire viral proteome during infection, affecting structural, non-structural, and accessory proteins. Quantitative proteomics comparing siMlec versus Scramble control conditions reveals significant decreases in viral glycoproteins, including Spike, nsp4, and nsp6. Importantly, MLEC knockdown also reduces levels of non-glycosylated viral proteins such as nucleocapsid and nsp2, demonstrating that the effect extends beyond direct glycoprotein interactions. This comprehensive reduction follows a cascade pattern, where decreased non-structural protein (nsp) levels (which are translated directly from the viral genome) subsequently lead to reduced viral genome replication, which in turn results in lower production of structural proteins that are translated from subgenomic RNAs. This pattern confirms that MLEC primarily impacts early viral protein production events, with effects that propagate throughout the viral replication cycle .

How can researchers experimentally differentiate between MLEC-dependent and MLEC-independent effects on viral replication?

Researchers can differentiate between MLEC-dependent and MLEC-independent effects on viral replication through several methodological approaches. A primary method involves pharmacological inhibition studies using compounds like NGI-1, which inhibits the glycan transfer activity of the OST complex upstream of MLEC in the glycoprotein biogenesis pathway. By comparing viral replication under conditions of MLEC knockdown alone, NGI-1 treatment alone, and combined MLEC knockdown with NGI-1 treatment, researchers can determine whether MLEC influences viral replication through its canonical glycoprotein biogenesis pathway or through alternative mechanisms. If MLEC operates through a pathway independent of glycoprotein biogenesis, the inhibitor and MLEC knockdown would produce additive suppressive effects on viral replication. Additionally, complementation studies using MLEC variants with mutations in specific functional domains can help identify which aspects of MLEC functionality are critical for viral replication support .

What changes occur to the MLEC interactome during viral infection?

During viral infection, the MLEC interactome undergoes substantial remodeling, characterized primarily by a large-scale loss of its basal interactions. Comparative proteomics of FLAG-tagged MLEC immunoprecipitations from mock versus infected cells reveals significant downregulation of numerous protein interactions. Among the most downregulated interactions are those with cytoskeletal proteins (CAPZB, VIM), microtubule-associated proteins (SLAIN2), and other lectins (LGALS1/3, also known as galectins). Notably, interactions with glycoprotein processing factors including glucosidase I (MOGS) and to a lesser extent glucosidase II (GANAB) are also reduced during infection. The calnexin (CANX) interaction and upstream regulator UGGT1 show modest downregulation, suggesting a potential decoupling of early and late glycoprotein quality control systems during infection. This interactome remodeling likely represents a viral strategy to redirect MLEC functionality toward supporting viral protein production at the expense of normal host protein quality control processes .

How do temporal dynamics of viral replication relate to MLEC dependency?

The temporal dynamics of viral replication reveal a critical window during which MLEC dependency is most pronounced. Using luminescence-based reporter viruses (such as MHV-FFL2) and time course experiments, researchers have determined that MLEC's impact becomes apparent approximately 6 hours post-infection and becomes increasingly significant at 8 and 10 hours post-infection. This timing coincides with the phase of active viral protein production and genome replication. Early events in viral infection (0-4 hours post-infection) show minimal MLEC dependency, consistent with MLEC not being involved in virus entry or initial genome release. The temporal progression of MLEC dependency correlates with the viral replication cycle transitioning from early events (entry and genome delivery) to protein production and genome replication. This time-dependent effect is observed at multiple MOIs (multiplicity of infection), though the magnitude of MLEC dependency decreases somewhat at higher MOIs (MOI of 1 versus 0.1), suggesting a partial dosage relationship between viral input and MLEC requirement .

What methodological approaches can measure MLEC's impact on different stages of the viral life cycle?

Researchers employ multiple complementary methodologies to precisely measure MLEC's impact on different viral life cycle stages. For entry and early events, quantitative reverse transcription PCR (RT-qPCR) measuring positive-sense genomic RNA ((+)gRNA) levels at early timepoints (e.g., 2 hours post-infection) provides direct assessment. For viral protein production, luminescence-based reporter viruses (like MHV-FFL2, where luciferase is fused to viral nsp2) enable real-time quantification of viral protein synthesis. Time course studies with samples collected at regular intervals (e.g., every 2 hours from 2-10 hours post-infection) allow temporal resolution of when MLEC's effects manifest. Genome replication is assessed through RT-qPCR quantification of negative-sense genomic RNA ((-)gRNA), the replication intermediate. Comprehensive viral proteome analysis uses quantitative mass spectrometry with appropriate controls to determine how MLEC affects all viral protein categories (structural, non-structural, and accessory). Finally, viral progeny production is measured through infectivity assays quantifying infectious viral titers in culture supernatants. Together, these approaches provide a multi-dimensional assessment of MLEC's role throughout the viral replication cycle .

What genetic manipulation approaches are most effective for studying MLEC function?

For studying MLEC function, several genetic manipulation approaches have proven particularly effective. RNA interference using small interfering RNAs (siRNAs) offers a versatile approach for transient knockdown of MLEC expression, with pooled siRNA treatments achieving approximately 80% reduction in endogenous MLEC protein levels. When implementing siRNA approaches, validation with individual constituent siRNAs is crucial to confirm specificity, as demonstrated by studies showing that at least two of four individual siRNAs targeting MLEC (#2 and #3) reproduce the phenotypes observed with pooled treatments. For stable genetic models, CRISPR-Cas9-mediated gene editing enables the generation of MLEC knockout cell lines, though complete knockout may affect cell viability in some contexts given MLEC's role in glycoprotein quality control. Complementary to these loss-of-function approaches, gain-of-function studies using epitope-tagged MLEC overexpression (such as FLAG-tagged MLEC) allow for both functional studies and interaction analyses through immunoprecipitation followed by proteomics. When designing genetic manipulation experiments, appropriate controls must be included, such as non-targeting scramble siRNAs for knockdown studies and empty vector transductions for overexpression studies .

How can researchers distinguish MLEC-specific effects from general disruption of the glycoprotein quality control pathway?

Distinguishing MLEC-specific effects from general disruption of the glycoprotein quality control pathway requires a multi-faceted experimental approach. Comparative studies targeting different components of the pathway provide crucial insights—researchers should perform parallel knockdowns or inhibition of other pathway components such as glucosidase I (MOGS), glucosidase II (GANAB), calnexin (CANX), or UDP-glucose:glycoprotein glucosyltransferase 1 (UGGT1). Pharmacological approaches using specific inhibitors at carefully titrated concentrations help isolate particular steps in the pathway; for example, NGI-1 at 5 μM inhibits the oligosaccharyltransferase (OST) complex upstream of MLEC. Rescue experiments represent another powerful approach, where MLEC mutants with selective functional defects (such as impaired lectin activity but intact protein-protein interaction capabilities) can determine which MLEC functions are essential for observed phenotypes. Additionally, temporal analysis comparing the kinetics of effects from MLEC disruption versus other pathway components can reveal unique aspects of MLEC function. Finally, global versus targeted analysis of glycoprotein folding and trafficking through pulse-chase experiments with specific client proteins can distinguish between general quality control defects and substrate-specific effects of MLEC disruption .

What quantitative proteomics workflows are optimal for analyzing MLEC interactome changes during infection?

For analyzing MLEC interactome changes during infection, optimal quantitative proteomics workflows involve several critical components. Sample preparation should include chemical crosslinking with reagents like dithiobis(succinimidyl propionate) (DSP) at 0.5 mM to capture transient lectin-client interactions, followed by immunoprecipitation using epitope-tagged MLEC (e.g., FLAG-tagged MLEC). Experimental design must include appropriate controls: parental cells (without tagged MLEC) subjected to the same immunoprecipitation protocol serve as background controls, while mock-infected cells expressing tagged MLEC establish the basal interactome. Multiplexed quantitative labeling with TMTpro enables direct comparison across conditions within a single mass spectrometry run, minimizing technical variability. Data analysis should include normalization to bait (MLEC) levels across samples, followed by statistical analysis to identify significantly changed interactions (enriched or depleted). Downstream bioinformatic analysis should classify interacting proteins by function, localization, and pathway involvement to recognize patterns in interactome changes. Validation of key interactions using orthogonal methods (such as co-immunoprecipitation followed by Western blotting) confirms mass spectrometry findings. This comprehensive workflow allows researchers to obtain a systems-level view of how viral infection remodels the functional networks of MLEC within the cell .

How does MLEC function differ between normal and disease states in human tissues?

MLEC function shows distinct differences between normal and disease states in human tissues, particularly in contexts of ER stress and viral infection. In normal physiological conditions, MLEC serves as a quality control checkpoint for newly synthesized glycoproteins, operating in concert with other ER quality control factors like glucosidases, calnexin, and UGGT1. During viral infection, MLEC's interactome undergoes significant remodeling, with substantial loss of interactions with normal glycoprotein processing factors like MOGS (glucosidase I) and GANAB (glucosidase II), suggesting a repurposing of MLEC function to support viral replication. This repurposing comes at the expense of normal host glycoprotein quality control, potentially contributing to ER stress observed during viral infection. The preferential reduction in interactions with cytoskeletal proteins (CAPZB, VIM) and other lectins (LGALS1/3) during infection suggests virus-specific modulation of MLEC's cellular functions. These alterations in MLEC's functional network likely represent a viral strategy to optimize cellular resources for viral replication, though they may contribute to cellular dysfunction in infected tissues .

How can insights from MLEC research inform our understanding of emerging viral threats?

Insights from MLEC research provide valuable frameworks for understanding and combating emerging viral threats through several mechanisms. First, the identification of MLEC as a pro-viral host factor for coronaviruses suggests it may play similar roles in other emerging viruses with extensive glycoprotein requirements, potentially offering a common vulnerability across diverse viral families. Second, the detailed understanding of how MLEC promotes viral protein production reveals a critical checkpoint in viral replication that could be targeted preemptively in newly emerging viral threats. Third, the methodological approaches developed to study MLEC—including proteomics workflows for analyzing dynamic interactomes during infection, reporter systems for monitoring viral protein production, and integrated approaches for distinguishing effects on different viral life cycle stages—provide a technical roadmap for rapidly characterizing host-pathogen interactions in emerging viruses. Fourth, the observed remodeling of the MLEC interactome during infection highlights how viruses repurpose host quality control machinery, a principle likely applicable to many emerging pathogens. Finally, these insights suggest that therapeutic strategies targeting shared host factors like MLEC could provide advantage against future pandemic threats by offering intervention options that may remain effective even as viruses evolve, potentially complementing traditional approaches like vaccines and direct-acting antivirals .

What are the key experimental findings regarding MLEC knockdown on viral replication?

The following table summarizes key experimental findings regarding the effects of MLEC knockdown on various aspects of viral replication:

Viral ParameterTime Post-InfectionEffect of MLEC KnockdownMagnitude of EffectExperimental Method
Entry/Early Events ((+)gRNA)2 hoursIncreaseModerate increaseRT-qPCR
Viral Replicase Production (nsp2)6 hoursDecreaseModerate decreaseLuminescence (MHV-FFL2)
Viral Replicase Production (nsp2)8-10 hoursDecreaseSignificant decreaseLuminescence (MHV-FFL2)
Genome Replication ((-)gRNA)10 hoursDecreaseSignificant decreaseRT-qPCR
Viral Transcript Levels (N gene)10 hoursDecreaseSignificant decreaseRT-qPCR
Viral Structural Proteins10 hoursDecreaseGlobal reductionQuantitative Proteomics
Viral Non-structural Proteins10 hoursDecreaseGlobal reductionQuantitative Proteomics
Infectious Viral Titers10 hoursDecreaseExceeds receptor KD effectViral Titer Assay

These findings collectively demonstrate that MLEC knockdown does not affect viral entry but significantly impairs viral protein production, which subsequently leads to reduced genome replication and ultimately lower viral titers. The effect becomes more pronounced as the viral replication cycle progresses, with the most substantial impact occurring during the later stages of infection (8-10 hours post-infection) .

How does the MLEC interactome change during viral infection?

The table below details significant changes in the MLEC interactome during viral infection compared to mock-infected conditions:

Protein CategorySpecific ProteinsInteraction Change During InfectionPotential Functional Significance
OST Complex MembersRPN1, RPN2, MAGT1, DDOST, STT3A, STT3BMaintained (canonical interactions)Preserved core glycoprotein processing function
Cytoskeletal ProteinsCAPZB, VIMStrongly DownregulatedAltered cellular architecture/trafficking
Microtubule-AssociatedSLAIN2Strongly DownregulatedChanged cellular transport pathways
LectinsLGALS1/3 (Galectins)Strongly DownregulatedModified glycan recognition network
Early Glycan ProcessingMOGS (Glucosidase I)Moderately DownregulatedAltered glycoprotein quality control
Late Glycan ProcessingGANAB (Glucosidase II)Mildly DownregulatedModified progression through quality control
Folding CycleCANX (Calnexin)Mildly DownregulatedChanged client protein handling
Quality ControlUGGT1Mildly DownregulatedAltered reglucosylation of misfolded proteins
Viral ProteinsSpikeMildly EnrichedDirect or indirect viral protein interaction

This interactome remodeling reveals a virus-induced shift in MLEC's functional network, with preservation of core OST complex interactions but significant loss of connections to downstream quality control components and cytoskeletal elements. This pattern suggests viral hijacking of MLEC's function, redirecting it from normal quality control processes toward supporting viral replication .

What methodological parameters are critical for studying MLEC in research contexts?

The following table outlines critical methodological parameters and considerations for studying MLEC in various research contexts:

Experimental ApproachCritical ParametersOptimal ConditionsImportant ControlsAnalytical Considerations
siRNA KnockdownsiRNA concentration, Transfection efficiency10-50 nM siRNA, ≥80% knockdownScramble siRNA, Individual siRNAs validationVerify protein reduction by Western blot
CRISPR Gene EditingGuide RNA design, Clonal selectionMultiple guide RNAs targeting different exonsWild-type cells, Empty vectorValidate knockout by sequencing and protein detection
Viral InfectionMOI, Infection durationMOI 0.1-1, 10 hours post-infectionMock infection, UV-inactivated virusConsider MOI-dependent effects
Crosslinking IP-MSCrosslinker concentration, IP conditions0.5 mM DSP, Optimized wash stringencyParental cells (no tag), Mock infectionNormalize to bait (MLEC) levels
Pharmacological InhibitionInhibitor concentration, Treatment timing5 μM NGI-1, At start of infectionVehicle (DMSO) controlConsider off-target effects
Reporter Virus AssaysMOI, Timepoint selectionMOI 0.1-1, Multiple timepoints (2-10 hpi)Positive controls (receptor KD)Account for assay dynamic range
RT-qPCRPrimer design, Reference genesVirus-specific and host control primersNo-template control, No-RT controlNormalize to stable reference genes

These methodological parameters provide critical guidance for researchers designing experiments to study MLEC function in viral infection and glycoprotein quality control contexts. Careful attention to these parameters ensures robust, reproducible results and proper interpretation of experimental outcomes .

What are the key unanswered questions regarding MLEC function in human cells?

Several critical questions regarding MLEC function in human cells remain unanswered and represent important areas for future research. First, the substrate specificity of MLEC needs further characterization—while it's known to bind diglucosyl oligosaccharides, the structural features that determine preferential binding to certain glycoproteins over others remain poorly defined. Second, the mechanistic details of how MLEC coordinates with other quality control factors (glucosidases, calnexin/calreticulin, UGGT1) to form a functional network for glycoprotein maturation require further elucidation. Third, the regulatory mechanisms controlling MLEC expression and activity during normal development, stress conditions, and disease states remain largely unexplored. Fourth, the role of MLEC in human disease pathogenesis beyond viral infections—particularly in conditions associated with ER stress and misfolded protein accumulation like neurodegenerative diseases—warrants investigation. Fifth, the evolutionary conservation of MLEC function across species suggests important biological roles that extend beyond current understanding. Finally, the potential compensatory mechanisms that may activate when MLEC function is compromised represent an important knowledge gap, as these could impact therapeutic strategies targeting MLEC. Addressing these questions will require integrative approaches combining structural biology, systems-level analyses, and physiological studies in relevant human cell and tissue models .

How might advanced techniques in glycobiology advance our understanding of MLEC function?

Advanced techniques in glycobiology hold significant promise for deepening our understanding of MLEC function. Glycan microarrays featuring systematically varied oligosaccharide structures would enable precise determination of MLEC's binding preferences and how structural variations in glycans affect recognition strength. Cryo-electron microscopy and X-ray crystallography of MLEC in complex with client glycoproteins would reveal the structural basis of substrate recognition and how binding induces conformational changes in either partner. Proximity labeling approaches like BioID or APEX2 could map the spatial organization of MLEC relative to other ER quality control components under various conditions. Chemoenzymatic glycan labeling combined with super-resolution microscopy would allow visualization of MLEC-glycoprotein interactions in situ. Glycoproteomics using selective enrichment of MLEC-bound glycoproteins followed by mass spectrometry would identify preferential substrates across the proteome. CRISPR screens targeting glycosylation pathway components could reveal synthetic interactions with MLEC, identifying compensatory or cooperative relationships. Finally, reconstitution of minimal glycoprotein quality control systems in vitro using purified components would allow mechanistic dissection of MLEC's direct effects on glycoprotein folding trajectories. These advanced approaches would collectively provide unprecedented insights into MLEC's role in glycoprotein biogenesis and quality control .

What interdisciplinary approaches might yield new insights into MLEC biology?

Interdisciplinary approaches spanning multiple scientific domains offer tremendous potential for generating novel insights into MLEC biology. Integrating structural biology with computational modeling could predict how MLEC recognizes diverse glycan structures and interacts with client proteins, guiding rational design of selective inhibitors. Combining systems biology with mathematical modeling would help decode the complex networks connecting MLEC to broader ER proteostasis systems and predict system-level consequences of MLEC perturbation. Incorporating viral evolution studies with molecular virology could reveal how different virus families have adapted to exploit or evade MLEC-dependent processes during infection. Merging immunology with glycobiology approaches would elucidate potential roles for MLEC in immune recognition, as glycan structures significantly influence immune system interactions. Employing tissue engineering and organoid models would allow examination of MLEC function in physiologically relevant three-dimensional contexts mimicking human tissues. Utilizing patient-derived cells with clinical data could establish connections between MLEC variants and disease susceptibility or progression. Finally, developing translational approaches that target MLEC-dependent processes could yield novel therapeutic strategies for viral infections and other diseases involving glycoprotein misfolding. These interdisciplinary approaches would collectively push beyond current knowledge boundaries, revealing new facets of MLEC biology with potential clinical applications .

Product Science Overview

Structure and Expression

Recombinant Human Malectin is a fragment protein that ranges from amino acids 29 to 269. It is expressed in Escherichia coli and has a purity greater than 95%, making it suitable for applications such as SDS-PAGE and mass spectrometry (MS) . The protein is tagged with a His tag at the N-terminus to facilitate purification and detection.

Biological Function

Malectin is involved in the quality control of glycoproteins. It binds to specific glycan structures and plays a role in the early steps of protein N-glycosylation. This process is essential for the proper folding and function of glycoproteins, which are critical for various cellular functions .

Role in Viral Infections

Recent studies have highlighted the role of Malectin in viral infections, particularly in the replication and biogenesis of viral proteins. For instance, Malectin has been shown to promote the replication of coronaviruses by interacting with viral nonstructural proteins and host factors. This interaction helps stabilize the association with the Oligosaccharyltransferase (OST) complex, which is crucial for viral glycoprotein production . This makes Malectin a potential target for antiviral therapies, especially for coronaviruses like SARS-CoV-2 .

Applications

Recombinant Human Malectin is widely used in research to study glycoprotein quality control and protein N-glycosylation. It is also used in high-throughput screening assays and other biochemical applications . The protein’s high purity and specific binding properties make it a valuable tool for researchers studying glycoprotein interactions and functions.

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.