KHDC1L Human

KH Homology Domain Containing 1-Like Human Recombinant
Shipped with Ice Packs
In Stock

Description

Cancer-Related Functions

KHDC1L is implicated in head and neck squamous cell carcinoma (HNSCC), where it promotes proliferation and inhibits apoptosis via AKT and Bcl-2 pathways . Key findings include:

ParameterKHDC1L OverexpressionLow Expression
Cell ProliferationIncreased (CCK8, cell counting)Reduced
ApoptosisDecreased (flow cytometry)Elevated (Caspase-3/PARP-1 activity)
Signaling Pathways↑ pAKT/AKT, ↑ Bcl-2; ↓ BAXEnrichment in P53/apoptosis pathways

In HNSCC cell line CAL27, upregulation of KHDC1L activates the PI3K-AKT pathway, enhancing cell viability and colony formation .

Pan-Cancer Analysis

KHDC1L expression varies across cancers, with implications for survival outcomes:

Data derived from TCGA and GEPIA databases .

Molecular and Cellular Interactions

KHDC1L interacts with diverse biological entities, as per Harmonizome data :

CategoryExamples
Co-Expressed GenesBCL2, AKT1, CASP3 (survival/apoptosis pathways)
Chemical Interactions17β-estradiol, benzo[a]pyrene, methylmercury (modulates expression)
DiseasesOsteoarthritis (synovial cell proliferation), cancers (HNSCC, TGCT)

Recombinant Protein Availability

Commercially available recombinant KHDC1L proteins vary in production systems and tags:

SourceTagPurityPrice Range
E. coli (ProSpec, Abcam)His-tag>90%$60 – $3,600
E. coli (Bio-Techne)His-tag>90%Not listed
HEK293T (Boster Bio)C-Myc/DDK>80%~$500 (20 µg)

Prices and specifications from .

Research Applications

KHDC1L is used in:

  1. Cancer Biology: Studying survival pathways in HNSCC and other cancers.

  2. Protein Interactions: Exploring RNA-binding and kinase regulation.

  3. Therapeutic Targeting: Identifying inhibitors of AKT/Bcl-2 pathways.

Product Specs

Introduction
KH homology domain containing 1-like, also known as KHDC1L, is a member of the KHDC1 family. KHDC1L is a protein-coding gene; an important paralog of this gene is KHDC1.
Description
Recombinant KHDC1L protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. The protein sequence contains amino acids 1-128 of the KHDC1L protein and includes an N-terminal His-tag (23 amino acids). The molecular weight of the protein is 16.9 kDa. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
KHDC1L protein solution (0.25 mg/mL) in 20 mM Tris-HCl buffer (pH 8.0), 0.2 M NaCl, 50% glycerol, and 2 mM DTT.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Greater than 90% purity as determined by SDS-PAGE analysis.
Synonyms
KH Homology Domain Containing 1-Like, RP11-257K9.7, Putative KHDC1-Like Protein.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAVGTSA LSKEPWWTLP ENFHSPMVFH MEEDQEELIF GLDDTYLRCI ELHSHTLIQL ERCFTATGQT RVTVVGPPMA KQWLLLMFHC VGSQDSKCHA RGLKMLERVR SQPLTNDDLV TSVSLPPYTG D.

Q&A

What is KHDC1L and how is it classified molecularly?

KHDC1L is a 128-amino acid protein belonging to the K-homology domain-containing 1 (KHDC1) family of RNA-binding proteins (RBPs). It shares 84.4% sequence similarity with KHDC1 and is primarily localized in the cytoplasm . As an RBP, KHDC1L orchestrates cellular activities at the post-transcriptional level, affecting RNA stability and translation processes .

The protein is encoded by the KHDC1L gene located on chromosome 6q13. To study KHDC1L experimentally, researchers typically use vector constructs (e.g., pcDNA3.1-KHDC1L) with FLAG tags for detection since commercial antibodies specific to KHDC1L are currently limited .

What experimental methods are most effective for detecting KHDC1L expression in clinical samples?

For KHDC1L detection in clinical contexts, RNA-based methods have proven most reliable due to antibody limitations:

RNA detection methods:

  • RT-PCR using specific primers (forward: 5′-GACTTCATGACACGTACCTTCG-3′ and reverse: 5′-AGCGTGACACTTGGAGTCCT-3′)

  • RNA sequencing (RNA-seq) for transcriptomic analysis

  • In situ hybridization for tissue localization

Protein detection methods:

  • Western blotting using FLAG-tagged constructs for experimental models

  • Developing custom antibodies (currently limited commercial options)

When working with clinical samples, researchers should normalize KHDC1L expression to housekeeping genes like GAPDH for accurate quantification. Bioinformatic tools like GEPIA (Gene Expression Profiling Interactive Analysis) can also help analyze KHDC1L expression patterns across different cancer subtypes using existing datasets like TCGA .

How does KHDC1L expression differ between normal and cancerous tissues?

KHDC1L exhibits distinct expression patterns between normal and cancerous tissues:

Tissue TypeKHDC1L Expression LevelEvidence
Normal head and neck tissuesLowTCGA data analysis via GEPIA
HNSCC tissuesHighTCGA data analysis
Normal oral epithelial cells (HOK)LowRT-PCR verification
Oral cancer cells (SCC9, CAL27)HighRT-PCR verification
Testicular Germ Cell TumorsHighGEPIA pan-cancer analysis
Normal testicular tissueLower than tumorGEPIA analysis

The aberrant high expression of KHDC1L in multiple cancer types suggests its potential role as a biomarker. In HNSCC specifically, elevated expression was observed across all molecular subtypes (atypical, basal, classical, and mesenchymal) , indicating this may be a consistent feature of head and neck malignancies regardless of subclassification.

What are the primary cellular pathways affected by KHDC1L in cancer cells?

KHDC1L influences several critical cellular pathways in cancer cells, primarily affecting cell proliferation and apoptosis regulation. Based on both bioinformatic analyses and experimental validation, the following pathways are significantly impacted:

Primary pathways affected:

  • PI3K-AKT signaling pathway - KHDC1L overexpression enhances AKT phosphorylation at Ser473, activating this pro-survival pathway

  • Bcl-2/BAX-mediated apoptosis regulation - Increased KHDC1L leads to higher Bcl-2/BAX ratio, inhibiting mitochondrial apoptosis

  • Caspase cascade - KHDC1L overexpression decreases cleaved Caspase-3 and cleaved PARP-1 levels

Secondary enriched pathways based on transcriptome analysis:

  • HPV infection pathway

  • Viral carcinogenesis

  • mTOR signaling

  • Transcriptional misregulation in cancer

  • RNA polymerase II-mediated transcription

Mechanistically, KHDC1L appears to first activate AKT signaling, which subsequently upregulates Bcl-2 expression, shifting the Bcl-2/BAX ratio to favor cell survival and suppress apoptotic processes .

How does KHDC1L modulation affect cancer cell proliferation and survival?

Experimental evidence demonstrates that KHDC1L has significant effects on cancer cell behavior:

Effects of KHDC1L overexpression (as seen in CAL27 HNSCC cells):

  • Increased cell viability (measured by CCK8 assay)

  • Enhanced cell proliferation (measured by cell counting)

  • Greater colony formation capacity

  • Decreased apoptosis (measured by flow cytometry)

  • Reduced cell detachment and death (morphological observation)

Molecular changes observed:

  • Increased phosphorylation of AKT at Ser473

  • Elevated Bcl-2/BAX ratio

  • Decreased cleaved Caspase-3 and cleaved PARP-1

These findings suggest that KHDC1L acts as an oncogenic factor by simultaneously promoting proliferation and inhibiting apoptosis, likely through its RNA-binding activities that affect post-transcriptional regulation of genes involved in these processes.

What is the prognostic significance of KHDC1L expression across different cancer types?

KHDC1L expression correlates differently with clinical outcomes depending on cancer type:

This variable prognostic significance suggests that KHDC1L may play different roles depending on the tissue context and tumor microenvironment. The contrasting effects in different cancers highlight the complexity of KHDC1L's functions and the need for tissue-specific research approaches when evaluating its potential as a biomarker.

What are the optimal in vitro models for studying KHDC1L function?

When selecting experimental models for KHDC1L research, consider:

Cell line models with validated KHDC1L expression:

  • CAL27 (HNSCC cell line) - Successfully used for KHDC1L overexpression studies

  • SCC9 (Oral cancer cell line) - Shows elevated endogenous KHDC1L expression

  • HOK (Human oral keratinocytes) - Normal control with low KHDC1L expression

Experimental approaches for functional studies:

  • Gain-of-function studies:

    • Transfection with pcDNA3.1-KHDC1L (with 3× flag tag for detection)

    • Assessment via cell viability (CCK8), cell counting, and colony formation assays

  • Loss-of-function studies:

    • siRNA or shRNA knockdown targeting KHDC1L

    • CRISPR-Cas9 gene editing for knockout models

  • Pathway analysis:

    • Western blotting for key signaling proteins (pAKT/AKT, Bcl-2, BAX, cleaved Caspase-3, cleaved PARP-1)

    • Transcriptome sequencing to identify downstream targets

For reproducible results, researchers should validate KHDC1L modulation at both mRNA and protein levels and include appropriate vector controls in all experiments.

How can researchers effectively analyze KHDC1L's RNA-binding targets and mechanisms?

As an RNA-binding protein, identifying KHDC1L's target transcripts is crucial for understanding its function:

Recommended methodologies:

  • RNA Immunoprecipitation (RIP):

    • Utilize FLAG-tagged KHDC1L constructs for pulldown

    • Combine with sequencing (RIP-seq) to identify bound transcripts

    • Validate specific targets via RT-qPCR

  • Cross-linking Immunoprecipitation (CLIP):

    • CLIP-seq or eCLIP for higher resolution mapping of binding sites

    • Identify sequence motifs recognized by KHDC1L's KH domain

  • RNA stability assays:

    • Actinomycin D chase experiments to determine if KHDC1L affects mRNA half-life

    • Compare decay rates of potential target mRNAs between KHDC1L-overexpressing and control cells

  • Translational efficiency analysis:

    • Polysome profiling to determine if KHDC1L affects translation of target mRNAs

    • Ribosome profiling to assess translational impact at genome-wide scale

  • Structural analysis:

    • RNA electrophoretic mobility shift assays (EMSA) to confirm direct binding

    • Mapping the critical residues in the KH domain responsible for RNA recognition

These approaches can help establish whether KHDC1L primarily affects mRNA stability, translation, or other post-transcriptional processes of specific target transcripts that mediate its biological effects.

What strategies can resolve challenges in detecting endogenous KHDC1L protein?

The lack of commercial antibodies for KHDC1L creates challenges for protein detection:

Short-term solutions:

  • Epitope tagging:

    • Generate FLAG-tagged or HA-tagged KHDC1L constructs for overexpression studies

    • Validate expression using tag-specific antibodies

  • Custom antibody development:

    • Design peptides based on unique regions of KHDC1L (avoiding the highly similar regions shared with other KHDC1 family proteins)

    • Validate specificity using overexpression and knockdown controls

  • Mass spectrometry:

    • Targeted proteomics approaches to detect and quantify endogenous KHDC1L

    • Useful for tissues with higher endogenous expression

Long-term strategies:

  • CRISPR knock-in:

    • Generate cell lines with endogenously tagged KHDC1L

    • Enables physiological level detection without overexpression artifacts

  • Proximity labeling:

    • BioID or APEX2 fusion constructs to identify proximal proteins

    • Can indirectly confirm KHDC1L expression and localization

When reporting results, researchers should clearly acknowledge the detection method's limitations and include appropriate controls to demonstrate specificity.

What is known about KHDC1L's role in embryonic development?

While research on KHDC1L in embryonic development is emerging, evidence suggests important developmental functions:

Current knowledge:

  • KHDC1L belongs to a family of RNA-binding proteins with roles in early developmental processes

  • Recent research indicates involvement in paternal gene expression dynamics during early embryo development

  • The STRING database shows interactions with developmental proteins like LEUTX (Leucine twenty homeobox) and transcriptional regulators

Research gaps and future directions:

  • The specific transcripts regulated by KHDC1L during embryogenesis remain largely unknown

  • Temporal expression patterns across developmental stages need further characterization

  • Knockout models would help establish developmental requirements and phenotypes

Researchers investigating KHDC1L in development should consider both its RNA-binding functions and potential roles in translational regulation during critical developmental windows.

How does KHDC1L interact with other members of the KHDC1 family?

Understanding the relationship between KHDC1L and other KHDC1 family members is important for functional characterization:

Key protein-protein interactions:

  • KHDC1L shares 84.4% sequence similarity with KHDC1

  • KHDC1A has been implicated in apoptosis induction in T cells

  • The KHDC1 family contains several members with RNA-binding capabilities

Methodological approaches to study interactions:

  • Co-immunoprecipitation:

    • Tag different family members and assess physical interactions

    • Determine if they form heterodimers or compete for binding partners

  • Functional redundancy analysis:

    • Compare phenotypes between individual knockdowns and combined knockdowns

    • Assess rescue capabilities between family members

  • Expression correlation:

    • Analyze co-expression patterns across tissues and disease states

    • Determine if they show compensatory regulation

  • Domain swapping experiments:

    • Create chimeric proteins to identify functional domains

    • Determine which regions confer specific activities

These approaches can help establish whether KHDC1L functions independently or in concert with other family members, which is critical for interpreting experimental results and developing targeted interventions.

How might single-cell analysis advance our understanding of KHDC1L heterogeneity in tumors?

Single-cell technologies offer powerful approaches to address KHDC1L heterogeneity:

Methodological considerations:

  • Single-cell RNA sequencing (scRNA-seq):

    • Map KHDC1L expression across different cell populations within tumors

    • Correlate with markers of proliferation, stemness, and resistance

    • Identify cell states associated with high KHDC1L expression

  • Spatial transcriptomics:

    • Visualize KHDC1L expression patterns within the tumor microenvironment

    • Associate expression with specific niches (hypoxic regions, invasive front)

  • Trajectory analysis:

    • Track KHDC1L expression changes during tumor evolution

    • Identify whether KHDC1L marks specific developmental or differentiation states

  • Integration with multi-omics:

    • Correlate KHDC1L mRNA with proteomic and epigenomic features at single-cell level

    • Develop comprehensive models of KHDC1L regulation

These approaches could reveal whether KHDC1L marks specific tumor cell subpopulations with distinct functional properties, potentially explaining the variable prognostic associations observed across cancer types.

What therapeutic strategies could target KHDC1L or its downstream pathways?

Given KHDC1L's role in promoting cancer cell proliferation and inhibiting apoptosis, several therapeutic approaches warrant investigation:

Direct targeting strategies:

  • RNAi-based therapeutics:

    • siRNA or antisense oligonucleotides targeting KHDC1L mRNA

    • Delivery challenges need consideration (nanoparticles, conjugates)

  • Small molecule inhibitors:

    • Target the RNA-binding pocket of KHDC1L's KH domain

    • Screen for compounds that disrupt KHDC1L-RNA interactions

Indirect targeting approaches:

  • AKT pathway inhibitors:

    • Since KHDC1L activates AKT, combining AKT inhibitors with standard therapy could be effective

    • Several AKT inhibitors in clinical development could be assessed

  • Bcl-2 antagonists:

    • As KHDC1L increases Bcl-2/BAX ratio, BH3 mimetics like venetoclax might counteract its effects

    • Consider combination approaches targeting both AKT and apoptotic machinery

  • Synthetic lethality:

    • Identify genes that, when inhibited, cause selective death in KHDC1L-overexpressing cells

    • Screen for compounds that exploit KHDC1L-dependent vulnerabilities

Research should focus on determining which patient populations might benefit most from KHDC1L-targeted therapies based on comprehensive biomarker analysis.

How can the contradictory prognostic associations of KHDC1L across different cancers be reconciled?

The variable prognostic significance of KHDC1L across cancer types presents an intriguing research question:

Methodological approaches to address this contradiction:

  • Context-dependent interaction mapping:

    • Compare KHDC1L protein-protein and protein-RNA interactions across different tissue contexts

    • Identify tissue-specific binding partners that might alter function

  • Pathway analysis across cancer types:

    • Conduct comparative transcriptomics of KHDC1L-high vs. KHDC1L-low tumors across cancer types

    • Identify which downstream pathways are consistently or differentially affected

  • Genetic background assessment:

    • Evaluate how co-occurring mutations modify KHDC1L's impact

    • Develop genetic interaction maps to identify synergistic or antagonistic effects

  • Microenvironmental influences:

    • Assess how tumor microenvironment factors affect KHDC1L function

    • Determine if immune infiltration patterns correlate with KHDC1L's prognostic impact

  • Isoform analysis:

    • Investigate whether different KHDC1L splice variants predominate in different cancers

    • Characterize functional differences between potential isoforms

These approaches could reveal whether KHDC1L functions as a context-dependent modifier of cancer progression, potentially explaining its divergent prognostic associations from positive in ovarian cancer to negative in sarcoma and other malignancies .

What experimental controls are essential when studying KHDC1L in cancer models?

Rigorous experimental design requires appropriate controls:

Essential controls for KHDC1L research:

  • Expression validation controls:

    • Empty vector controls for overexpression studies (e.g., pcDNA3.1)

    • Non-targeting siRNA/shRNA controls for knockdown studies

    • Validation at both mRNA (RT-qPCR) and protein levels (western blot with tagged constructs)

  • Cell type controls:

    • Multiple cancer cell lines to ensure observations aren't cell-line specific

    • Matched normal cells as baseline controls (e.g., HOK for oral cancer studies)

    • Patient-derived primary cells to confirm relevance beyond established cell lines

  • Pathway validation controls:

    • Pathway inhibitors (e.g., AKT inhibitors) to confirm mechanism

    • Rescue experiments with downstream effectors

    • Time-course analyses to establish causality in signaling cascades

  • Phenotypic assay controls:

    • Multiple complementary assays for each phenotype (e.g., CCK8, cell counting, and colony formation for proliferation)

    • Both short-term and long-term readouts where applicable

    • Appropriate positive controls for each assay

Implementing these controls helps ensure that observed effects are specifically attributable to KHDC1L rather than experimental artifacts or non-specific effects.

How can researchers overcome the challenges of KHDC1L's similarity to other KHDC1 family members?

The high sequence similarity between KHDC1L and other KHDC1 family members presents specificity challenges:

Strategies for ensuring specificity:

  • Primer and siRNA design:

    • Target unique regions that differ between family members

    • Validate specificity by measuring expression of all family members after intervention

    • Use multiple independent siRNAs/primers and confirm consistent results

  • Expression construct considerations:

    • Use full-length cDNA confirmed by sequencing

    • Include appropriate tags that don't interfere with function

    • Validate specificity of overexpression at mRNA level

  • Bioinformatic approaches:

    • When analyzing public datasets, assess probe or primer specificity

    • For RNA-seq data, use alignment parameters that distinguish between highly similar transcripts

    • Validate key findings with orthogonal methods

  • Functional validation:

    • Compare phenotypes between manipulations of different family members

    • Perform rescue experiments with specific family members

    • Use domain-specific approaches to distinguish functional differences

These strategies help ensure that observed effects are attributable specifically to KHDC1L rather than to related family members, improving the reliability and reproducibility of research findings.

How does KHDC1L's role in embryonic development relate to its functions in cancer?

The dual involvement of KHDC1L in both embryonic development and cancer suggests potential connections worth exploring:

Comparative research approaches:

  • Developmental pathway reactivation analysis:

    • Compare transcriptional profiles between KHDC1L-expressing embryonic cells and cancer cells

    • Identify common regulatory networks and target genes

    • Determine if KHDC1L regulates similar RNA targets in both contexts

  • Epigenetic regulation studies:

    • Analyze chromatin states and DNA methylation patterns at the KHDC1L locus

    • Compare epigenetic regulation between developmental stages and cancer progression

    • Investigate whether cancer-specific epigenetic changes drive KHDC1L dysregulation

  • Lineage tracing experiments:

    • Determine if KHDC1L marks specific progenitor populations during development

    • Assess whether KHDC1L-expressing cancer cells show stem-like properties

    • Investigate potential roles in cellular plasticity and differentiation

Understanding these connections could reveal whether KHDC1L's role in cancer represents an aberrant reactivation of developmental programs, potentially providing new insights into both normal development and malignant transformation.

What is the relationship between KHDC1L and the immune microenvironment in cancer?

The interaction between KHDC1L and tumor immunity represents an unexplored frontier:

Research directions to explore this relationship:

  • Correlation analyses in public datasets:

    • Analyze associations between KHDC1L expression and immune infiltration patterns

    • Examine relationships with immune checkpoint molecules and cytokine signatures

    • Compare these patterns across cancer types with different KHDC1L prognostic associations

  • In vitro co-culture systems:

    • Assess how KHDC1L modulation in cancer cells affects interactions with immune cells

    • Investigate impacts on cytokine production and immune cell activation

    • Determine if KHDC1L affects antigen presentation or recognition

  • In vivo models with intact immunity:

    • Compare tumor growth and immune infiltration in KHDC1L-modulated tumors

    • Assess responses to immune checkpoint inhibitors

    • Evaluate potential synergies between KHDC1L targeting and immunotherapy

These approaches could reveal whether KHDC1L's variable prognostic significance across cancers relates to differential effects on tumor-immune interactions, potentially identifying new therapeutic opportunities combining KHDC1L targeting with immunomodulatory approaches.

How might KHDC1L function in non-cancer pathologies?

Beyond cancer, KHDC1L may have roles in other diseases that warrant investigation:

Potential non-cancer roles and research approaches:

  • Inflammatory diseases:

    • Previous research identified KHDC1L upregulation in osteoarthritis synovial cells

    • Investigate expression in other inflammatory conditions

    • Assess impact on inflammatory signaling pathways and cell survival

  • Developmental disorders:

    • Given its role in embryogenesis, analyze potential contributions to developmental abnormalities

    • Assess genetic variations in patients with relevant phenotypes

    • Study interactions with known developmental disorder genes

  • Degenerative diseases:

    • Investigate potential roles in cellular stress responses and apoptosis regulation

    • Examine expression in tissues affected by degenerative conditions

    • Assess contributions to cell survival under stress conditions

Expanding KHDC1L research beyond cancer contexts could reveal broader biological functions and potential therapeutic applications across multiple disease states, providing a more comprehensive understanding of this protein's significance in human health and disease.

Product Science Overview

Introduction

The KH Homology (KH) domain is a protein domain that was first identified in the human heterogeneous nuclear ribonucleoprotein (hnRNP) K. This domain is evolutionarily conserved and consists of approximately 70 amino acids. It is present in a wide variety of nucleic acid-binding proteins and plays a crucial role in RNA recognition and binding .

Structure and Function

The KH domain binds RNA and can function in RNA recognition. It is found in multiple copies in several proteins, where they can function cooperatively or independently. For example, in the AU-rich element RNA-binding protein KSRP, which has four KH domains, KH domains 3 and 4 behave as independent binding modules to interact with different regions of the AU-rich RNA targets .

The solution structure of the first KH domain of FMR1 and the C-terminal KH domain of hnRNP K determined by nuclear magnetic resonance (NMR) revealed a beta-alpha-alpha-beta-beta-alpha structure . The KH domain binds to nucleic acids in an extended conformation across one side of the domain. The binding occurs in a cleft formed between alpha helix 1, alpha helix 2, the GXXG loop (which contains a highly conserved sequence motif), and the variable loop .

Types of KH Domains

There are two structurally different types of KH domains identified by Grishin, known as type I and type II . The type I domains are mainly found in eukaryotic proteins, while the type II domains are predominantly found in prokaryotes. While both types share a minimal consensus sequence motif, they have different structural folds. The type I KH domains have a three-stranded beta-sheet where all three strands are anti-parallel. In contrast, the type II domain has two of the three beta strands in a parallel orientation .

Human Proteins Containing KH Domains

Several human proteins contain KH domains, including:

  • AKAP1
  • ANKHD1
  • ANKRD17
  • ASCC1
  • BICC1
  • DDX43
  • DDX53
  • DPPA5
  • ERAL1
  • FMR1
  • FUBP1
  • FUBP3
  • FXR1
  • FXR2
  • GLD1
  • HDLBP
  • HNRPK
  • IGF2BP1
  • IGF2BP2
  • IGF2BP3
  • KHDRBS1
  • KHDRBS2
  • KHDRBS3
  • KHSRP
  • KRR1
  • MEX3A
  • MEX3B
  • MEX3C
  • MEX3D
  • NOVA1
  • NOVA2
  • PCBP1
  • PCBP2
  • PCBP3
  • PCBP4
  • PNO1
  • PNPT1
  • QKI
  • SF1
  • TDRKH .

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.