PQBP1 Human

Polyglutamine Binding Protein 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of PQBP1 Human

PQBP1 (polyglutamine-binding protein 1) is a nuclear and cytoplasmic protein encoded by the PQBP1 gene located on the X chromosome (Xp11.23) . It is highly expressed in neural progenitor cells, neurons, and immune cells, playing critical roles in mRNA splicing, transcription regulation, innate immunity, and neurodevelopment . Structural analysis reveals a WW domain for protein interactions and a C-terminal YxxPxxVL motif essential for binding spliceosomal proteins like U5-15kD .

Molecular Structure and Functional Domains

DomainFunctionInteracting Partners
WW DomainBinds proline-rich motifs (e.g., WBP11) to regulate splicing and transcription WBP11, Dynamin 2, TXNL4A
YxxPxxVL MotifAnchors PQBP1 to U5-15kD in the spliceosome; lost in frameshift mutations U5-15kD, U5-52K
C-TerminalMediates RNA granule formation and stress granule dynamics RNA-binding proteins, SRRM2

Frameshift mutations disrupting the YxxPxxVL motif impair splicing and transcription, linking to X-linked intellectual disabilities .

RNA Metabolism

  • Splicing Regulation: Interacts with U5-15kD to stabilize the B-complex spliceosome, ensuring accurate mRNA processing of cell cycle and synaptic genes .

  • Transcription Coupling: Associates with RNA polymerase II to coordinate transcription and splicing .

Innate Immunity

  • Viral Sensing: Recognizes HIV-1 capsids via its N-terminus, triggering cGAS recruitment and antiviral responses .

  • Tauopathy Response: Binds extracellular tau in microglia, initiating neuroinflammatory pathways .

Neurodevelopment

  • Neural Proliferation: Regulates neural stem progenitor cell cycle timing in the ventricular/subventricular zone .

  • Synaptic Plasticity: Modulates NMDA receptor subunit (NR1) expression in Drosophila models .

Genetic Mutations and Syndromes

SyndromeMutation TypeKey SymptomsReferences
Renpenning SyndromeFrameshift, missenseMicrocephaly, intellectual disability, short stature
Golabi-Ito-HallMissense (WW domain)Coloboma, heart defects, developmental delays
Non-syndromic IDDeletions, truncationsCognitive impairment without distinct dysmorphia

Frameshift mutations account for ~75% of cases, disrupting the YxxPxxVL motif critical for splicing .

Animal Models and Pathogenic Insights

ModelManipulationPhenotypeMechanism
Mouse KDsiRNA knockdownAnxiety, impaired memory, microcephaly Reduced cell cycle efficiency in NSPCs
Mouse cKOConditional knockoutCognitive deficits, synaptic dysfunction Altered synapse-related gene expression
DrosophilaHypomorph expressionImpaired learning, reduced NR1 expression Disrupted NMDA receptor function

Alzheimer’s Disease and Tauopathy

  • SRRM2 Dysregulation: Phosphorylation of SRRM2 reduces nuclear PQBP1, impairing RNA metabolism and cognition .

  • Tau Pathology: PQBP1 binds tau aggregates, activating microglial inflammation .

HIV-1 and Innate Immunity

PQBP1 acts as a capsid sensor, recruiting cGAS to detect lentiviral DNA and trigger interferon responses .

Protein Interaction Network

PartnerInteraction SiteFunctional RoleSource
WBP11WW domainSplicing activation, PP1 phosphatase inhibition
U5-15kDYxxPxxVL motifSpliceosome assembly, mRNA processing
TXNL4AC-terminusNuclear import, stress granule regulation
ATXN1N-terminusRNA metabolism, spinocerebellar ataxia

Future Directions

  1. Therapeutic Targets: Replenishing PQBP1 or stabilizing its interactions (e.g., WBP11, U5-15kD) may address splicing deficits in ID.

  2. Biomarkers: Monitoring nuclear PQBP1 levels in AD or tauopathy to predict cognitive decline .

  3. Viral Pathogenesis: Inhibiting PQBP1-capsid interactions to modulate HIV-1 immunity .

Product Specs

Introduction
PQBP1, a transcription repressor, interacts with polyglutamine tract-containing transcription regulators and genes associated with neurodegenerative disorders. It is primarily located in the nucleus of neurons throughout the brain, exhibiting elevated levels in the hippocampus, olfactory bulb, and cerebellar cortex. PQBP1 possesses a WWP/WW domain for binding proline-rich motifs and a C2 domain capable of mediating Ca2+-dependent phospholipid signaling.
Description
Recombinantly produced in E. coli, PQBP1 Human is a single polypeptide chain comprising 289 amino acids (residues 1-265) with a molecular weight of 33.0 kDa. A 24 amino acid His-tag is fused to the N-terminus of PQBP1, which is purified using proprietary chromatographic techniques.
Physical Appearance
A sterile, colorless solution that has been filtered.
Formulation
The PQBP1 solution is provided at a concentration of 0.5 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 1 mM DTT, 100 mM NaCl, and 20% glycerol.
Stability
For optimal storage, refrigerate the product at 4°C if the entire vial will be used within 2-4 weeks. For extended storage, freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity greater than 95%.
Synonyms
Golyglutamine binding protein 1, Polyglutamine tract-binding protein 1, 38kDa nuclear protein containing a WW domain, mental retardation X-linked 55, Sutherland-Haan X-linked mental retardation syndrome, Npw38, MRXS3, MRX55, MRXS8, RENS1, SHS.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMPLPVA LQTRLAKRGI LKHLEPEPEE EIIAEDYDDD PVDYEATRLE GLPPSWYKVF DPSCGLPYYW NADTDLVSWL SPHDPNSVVT KSAKKLRSSN ADAEEKLDRS HDKSDRGHDK SDRSHEKLDR GHDKSDRGHD KSDRDRERGY DKVDRERERD RERDRDRGYD KADREEGKER RHHRREELAP YPKSKKAVSR KDEELDPMDP SSYSDAPRGT WSTGLPKRNE AKTGADTTAA GPLFQQRPYP SPGAVLRANA EASRTKQQD

Q&A

What is PQBP1 and what are its primary structural domains?

PQBP1 (polyglutamine tract-binding protein 1) is a multifunctional protein encoded by the PQBP1 gene located on the X chromosome. The protein contains several functional domains that contribute to its diverse cellular roles. These include a WW domain that mediates protein-protein interactions, particularly with proline-rich sequences; a polar amino acid-rich domain; and a C-terminal YxxPxxVL motif that interacts with spliceosomal proteins. This C-terminal motif is particularly crucial as it forms a complex with the U5-15kD spliceosomal protein by binding to its hydrophobic groove. Notably, this YxxPxxVL motif is absent in all PQBP1 frameshift mutants associated with intellectual disability syndromes, highlighting its functional significance .

What are the primary functions of PQBP1 in neuronal cells?

PQBP1 plays essential roles in multiple aspects of neural development and function. Research has demonstrated its involvement in regulating neural progenitor proliferation, neural projection formation, synaptic growth, neuronal survival, and cognitive function. These processes are mediated through both mRNA transcription and splicing-dependent mechanisms, as well as through splicing-independent pathways . PQBP1 functions as part of the spliceosome complex, influencing pre-mRNA processing, which is critical for proper gene expression patterns in neuronal cells. Additionally, it participates in DNA damage responses and has been implicated in cell cycle regulation, particularly in neuroblast exit from quiescence during development .

What is Renpenning syndrome and how is it associated with PQBP1?

Renpenning syndrome is an X-linked intellectual disability disorder characterized by a constellation of clinical features including moderate to severe intellectual disability, microcephaly, short stature, small testes, and distinctive facial features. Patients may also present with heart defects, muscular atrophy, and midline defects such as cleft palate, anal atresia, iris coloboma, and Tetralogy of Fallot . This syndrome is caused by mutations in the PQBP1 gene, particularly frameshift mutations involving duplications or deletions of AG dinucleotides in the fourth coding exon. These mutations result in the loss of the C-terminal YxxPxxVL motif, which prevents PQBP1 from properly interacting with spliceosomal proteins, thereby disrupting normal pre-mRNA processing .

How do mutations in PQBP1 lead to intellectual disability at the molecular level?

Mutations in PQBP1, particularly frameshift mutations that result in the loss of the C-terminal YxxPxxVL motif, disrupt the protein's ability to interact with the U5-15kD spliceosomal protein . This interaction is critical for proper pre-mRNA splicing, and its disruption leads to aberrant splicing patterns that affect multiple downstream targets important for neurodevelopment and neuronal function.

The loss of PQBP1 function affects neural progenitor proliferation, potentially leading to alterations in brain development and neural circuit formation. Studies have shown that PQBP1 regulates the translation of specific mRNAs, and disruption of this process may contribute to the pathogenesis of intellectual disability. Additionally, PQBP1 mutations may impair synaptic growth and function, which are fundamental processes for learning and memory . The cumulative effect of these molecular disruptions manifests as the intellectual disability and associated features observed in Renpenning syndrome.

What is the role of PQBP1 in neuroinflammation and neurodegenerative diseases?

PQBP1 functions as an intracellular receptor that can recognize specific molecular patterns and trigger inflammatory responses. Recent research has revealed that PQBP1 can bind to tau protein, which is associated with neurodegenerative conditions such as Alzheimer's and Parkinson's diseases . This interaction activates the cGAS-STING pathway, leading to inflammatory responses in the brain.

The PQBP1-mediated inflammatory response represents a novel mechanism linking protein misfolding diseases with neuroinflammation. In microglia, which are the brain's resident immune cells, PQBP1 recognition of tau induces activation of the cGAS-STING pathway, potentially contributing to the chronic inflammation observed in neurodegenerative conditions . This finding suggests that PQBP1 may serve as a bridge between protein quality control mechanisms and innate immune responses in the brain, with significant implications for understanding and potentially treating neurodegenerative diseases.

How does PQBP1 contribute to innate immunity against viral infections?

PQBP1 plays a critical role in the cellular recognition of viral components, particularly HIV-1 DNA. Research has demonstrated that PQBP1 initiates a two-step recognition process of HIV-1 reverse-transcribed DNA products that ensures a type 1 interferon response . The mechanism involves:

  • Initial binding of PQBP1 to the viral capsid upon HIV-1 entry into the cell

  • Recruitment of cyclic GMP-AMP synthase (cGAS) to the capsid in a PQBP1-dependent manner once capsid disassembly and HIV-1 DNA synthesis begin

  • Positioning of cGAS at the site of pathogen-associated molecular pattern (PAMP) formation, enabling sensing of temporally and spatially constrained HIV-1 DNA

  • Production of cyclic GMP-AMP (cGAMP) and activation of downstream STING-IRF3 signaling, leading to interferon production

This mechanism highlights PQBP1's role as a critical sensor in innate immune responses to viral infections, functioning similarly in both viral defense and neurodegenerative disease contexts by activating the cGAS-STING pathway .

What are the key experimental models for studying PQBP1 function?

Research on PQBP1 has employed various experimental models that provide complementary insights into its function:

  • Cellular models: Human cell lines (neuronal and non-neuronal) with PQBP1 knockdown or overexpression have been used to study molecular interactions and cellular pathways. Patient-derived cells, particularly those from individuals with Renpenning syndrome, offer valuable insights into pathogenic mechanisms.

  • Drosophila models: Studies have shown conservation of PQBP1 function in mRNA translation between Drosophila and mammals. Drosophila Fmr1 (related to PQBP1 pathways) mutants display locomotory deficits and decreased photoreceptor function, mirroring some aspects of human conditions .

  • Mouse models: PQBP1 knockout or conditional knockout mice have been used to study neurodevelopmental processes. These models show increased neural stem cell proliferation and density of intermediate progenitors and pyramidal cells in the early postnatal cortex, consistent with a role for PQBP1 in regulating neurogenesis .

  • In vitro biochemical systems: Purified PQBP1 and interacting proteins have been used to study structural biology and biochemical interactions, including the PQBP1-U5-15kD complex formation and interactions with tau protein .

Each model system offers distinct advantages for investigating specific aspects of PQBP1 biology, from molecular interactions to physiological functions in complex organisms.

How can researchers differentiate between PQBP1's roles in splicing versus its non-splicing functions?

Differentiating between splicing-dependent and splicing-independent functions of PQBP1 requires sophisticated experimental approaches:

  • Domain-specific mutations: Creating constructs with mutations in specific PQBP1 domains (e.g., the YxxPxxVL motif) that affect splicing while preserving other functions, or vice versa, can help dissect these distinct roles.

  • RNA-seq and splicing analysis: Comprehensive analysis of splicing patterns using RNA-seq in PQBP1-deficient or mutant cells can identify specific splicing events dependent on PQBP1.

  • Protein interaction studies: Identifying PQBP1 interacting partners through techniques like proximity labeling, co-immunoprecipitation, or yeast two-hybrid assays can distinguish between splicing complex components and other functional partners.

  • Temporal separation of functions: Using rapid protein degradation systems or inducible expression systems to manipulate PQBP1 levels at different time points can help separate immediate (likely non-splicing) effects from longer-term (splicing-dependent) consequences.

  • Subcellular localization studies: Investigating the localization of PQBP1 in different cellular compartments can provide clues about its various functions, as splicing occurs primarily in the nucleus while many non-splicing functions may occur in the cytoplasm.

By combining these approaches, researchers can develop a more nuanced understanding of how PQBP1 contributes to cellular function through both splicing and non-splicing mechanisms .

What are the current discrepancies in understanding PQBP1 function across different experimental systems?

Several notable discrepancies exist in the PQBP1 literature that warrant careful consideration:

  • Translational regulation: While some studies suggest PQBP1 activates translation initiation of large proteins (similar to FMRP), others indicate it acts as a negative translational regulator. These contradictory findings may reflect context-dependent functions or methodological differences .

  • Cell type-specific effects: PQBP1 appears to have different roles in different cell types. For example, its impact on synaptic function varies between central histaminergic photoreceptor synapses and peripheral glutamatergic neuromuscular junctions, with opposite effects observed in these different contexts .

  • Species differences: While many functions of PQBP1 are conserved across species (from Drosophila to humans), there may be species-specific roles or regulatory mechanisms that complicate the translation of findings between model systems .

  • Pathogen sensing versus neurodegeneration: The dual role of PQBP1 in both pathogen recognition (e.g., HIV-1) and neurodegeneration (e.g., tau recognition) suggests common mechanistic pathways, but the specific molecular details and consequences may differ between these contexts .

Resolving these discrepancies requires careful experimental design with appropriate controls, consideration of cellular context, and direct comparative studies across different model systems.

What are the most effective approaches for studying PQBP1 interactions with the spliceosome?

To investigate PQBP1's interactions with the spliceosome, researchers can employ several sophisticated approaches:

  • Structural biology techniques: X-ray crystallography and cryo-electron microscopy have been successfully used to determine the structure of PQBP1 in complex with spliceosomal proteins like U5-15kD, revealing critical interaction motifs such as the YxxPxxVL sequence .

  • In vitro splicing assays: Cell-free splicing systems using purified components or nuclear extracts allow direct assessment of how PQBP1 and its mutants affect splicing of reporter pre-mRNAs.

  • Spliceosome isolation: Biochemical purification of spliceosomes at different assembly stages (E, A, B, B*, and C complexes) followed by proteomic analysis can reveal the dynamic association and dissociation of PQBP1 during the splicing cycle .

  • RNA-protein crosslinking: Techniques like CLIP-seq (Cross-linking and immunoprecipitation followed by sequencing) can identify the direct RNA targets of PQBP1 within the spliceosome context.

  • Live-cell imaging: Fluorescently tagged PQBP1 and spliceosomal components can be used to visualize their dynamic interactions in living cells, particularly in nuclear speckles where splicing factors concentrate.

These approaches, used in combination, provide comprehensive insights into how PQBP1 participates in the complex and dynamic process of pre-mRNA splicing.

How can researchers effectively model Renpenning syndrome in vitro and in vivo?

Modeling Renpenning syndrome requires multifaceted approaches to recapitulate the complex features of this disorder:

  • Patient-derived cells: Fibroblasts or lymphoblastoid cell lines from Renpenning syndrome patients provide naturally occurring disease models. These can be reprogrammed into induced pluripotent stem cells (iPSCs) and differentiated into neurons or brain organoids to study neurodevelopmental aspects of the disease.

  • CRISPR/Cas9 gene editing: Introducing specific PQBP1 mutations found in Renpenning syndrome patients (particularly frameshift mutations affecting the YxxPxxVL motif) into cellular or animal models creates precise disease models.

  • Conditional knockout animals: Tissue-specific or temporally controlled deletion of PQBP1 can help dissect its role in different developmental stages and brain regions, addressing the complex phenotypes seen in Renpenning syndrome.

  • Brain organoids: Three-dimensional brain organoids derived from patient iPSCs or genetically engineered stem cells can recapitulate aspects of microcephaly and abnormal neuronal development characteristic of Renpenning syndrome.

  • Behavioral testing in animal models: Comprehensive behavioral phenotyping of PQBP1 mutant animals, focusing on learning, memory, social interaction, and motor coordination, can provide functional correlates to the intellectual disability observed in humans.

These approaches provide complementary insights into the molecular, cellular, developmental, and behavioral consequences of PQBP1 mutations .

What techniques are most suitable for investigating PQBP1's role in innate immunity and inflammation?

To explore PQBP1's role in innate immunity and inflammation, researchers can employ several specialized techniques:

  • Pathogen recognition assays: Using purified components or cellular systems to assess PQBP1 binding to viral components (e.g., HIV-1 capsid) or endogenous danger signals (e.g., tau protein) provides direct evidence of its recognition function .

  • cGAS-STING pathway monitoring: Measuring downstream signaling events including cGAMP production, STING phosphorylation, IRF3 nuclear translocation, and interferon gene expression can quantify pathway activation in response to PQBP1-recognized ligands.

  • Live-cell microscopy: Visualizing the recruitment of PQBP1 and subsequent cGAS localization to HIV-1 capsids or tau aggregates provides spatial and temporal information about the recognition process .

  • Microglia models: Primary microglia cultures, microglia-like cell lines, or iPSC-derived microglia with PQBP1 manipulation are particularly relevant for studying neuroinflammation in the context of neurodegenerative diseases.

  • In vivo inflammation models: Animal models with PQBP1 mutations challenged with viral infections or expressing neurodegenerative disease proteins can reveal the systemic consequences of altered PQBP1-mediated immune responses.

These approaches allow researchers to dissect the complex role of PQBP1 in bridging pathogen recognition and inflammatory responses in both infectious and neurodegenerative contexts .

What are the potential therapeutic strategies targeting PQBP1 pathways?

Several therapeutic strategies targeting PQBP1 pathways show promise for treating associated disorders:

  • Splicing modulators: Small molecules that can partially restore proper splicing function in the presence of mutant PQBP1 could address the fundamental defect in Renpenning syndrome.

  • Anti-inflammatory approaches: For PQBP1's role in neuroinflammation, inhibitors of the cGAS-STING pathway could potentially mitigate excessive inflammatory responses triggered by tau recognition in neurodegenerative diseases .

  • Gene therapy: Delivery of functional PQBP1 to affected tissues using viral vectors could potentially correct deficiencies in Renpenning syndrome, though this approach faces challenges regarding delivery, expression control, and safety.

  • Antisense oligonucleotides: These could be designed to modulate splicing of specific PQBP1 target transcripts that are misprocessed in disease states, potentially correcting downstream effects of PQBP1 dysfunction.

  • PQBP1 interaction modulators: Small molecules designed to enhance or inhibit specific PQBP1 protein-protein interactions could selectively modify its function in particular pathways while preserving others.

While these approaches are still largely theoretical, they represent promising avenues for future therapeutic development based on our growing understanding of PQBP1 biology .

How might single-cell technologies advance our understanding of PQBP1 function in heterogeneous tissues?

Single-cell technologies offer unprecedented opportunities to dissect PQBP1 function in complex tissues:

  • Single-cell RNA-seq (scRNA-seq): This can reveal cell type-specific expression patterns of PQBP1 and its target genes, identifying particularly vulnerable cell populations in disease states.

  • Single-cell ATAC-seq: Assessing chromatin accessibility at the single-cell level can provide insights into how PQBP1 might influence gene regulation through chromatin-associated mechanisms.

  • Spatial transcriptomics: These techniques maintain spatial information while providing transcriptomic data, allowing researchers to understand how PQBP1 function varies across anatomical regions in the brain or other tissues.

  • CyTOF and spectral flow cytometry: These approaches can simultaneously measure multiple protein markers at the single-cell level, allowing assessment of PQBP1 pathway activation across heterogeneous cell populations.

  • Live-cell imaging at single-cell resolution: This can reveal dynamic aspects of PQBP1 function, such as its translocation between cellular compartments or its recruitment to specific cellular structures in response to stimuli.

These technologies are particularly valuable for studying PQBP1 in the context of the brain, where cellular heterogeneity is pronounced and may explain some of the complex phenotypes associated with PQBP1 mutations .

How does PQBP1 function compare with other intellectual disability-associated RNA-binding proteins?

PQBP1 shares functional similarities with several other RNA-binding proteins implicated in intellectual disability, yet maintains distinct mechanisms:

ProteinAssociated DisorderPrimary FunctionsRNA TargetsComparison to PQBP1
FMRPFragile X SyndromeTranslational regulation, primarily repressionLong, autism-relevant transcriptsBoth can function in translation regulation, but PQBP1 has additional splicing roles; FMRP may activate translation of large proteins similar to PQBP1
PURAPURA SyndromeTranscriptional activation, mRNA transportGGN repeats, specific mRNAsBoth affect neurodevelopment, but through different mechanisms
RBFOX1Autism Spectrum DisordersAlternative splicing regulation(U)GCAUG motifsBoth involved in splicing but recognize different sequence motifs
MBNL1/2Myotonic DystrophySplicing regulationCUG repeatsBoth affect RNA processing, but MBNL proteins are sequestered by toxic RNA in disease

This comparative analysis highlights that while many RNA-binding proteins contribute to intellectual disability when mutated, they operate through distinct molecular mechanisms and affect different subsets of RNA targets. PQBP1 is unusual in its dual role in both splicing regulation and innate immunity pathways, distinguishing it from many other intellectual disability-associated RNA-binding proteins .

What is the evolutionary conservation of PQBP1 structure and function across species?

PQBP1 exhibits remarkable evolutionary conservation, suggesting fundamental biological importance:

  • Structural conservation: The key functional domains of PQBP1, including the WW domain and the YxxPxxVL motif that interacts with the spliceosome, are highly conserved from Drosophila to humans .

  • Functional conservation: Studies comparing mammalian and Drosophila models show similar effects of PQBP1/Fmr1 deficiency on translation of target mRNAs. In both species, translation of PQBP1-bound mRNAs decreases to a similar magnitude in PQBP1-deficient tissues .

  • Phenotypic conservation: Both Drosophila and mammalian PQBP1 mutants exhibit neurological deficits, including locomotor abnormalities and synaptic dysfunction, though the specific manifestations vary by species .

  • Divergent aspects: Despite the similarities, there are species-specific aspects of PQBP1 function. For instance, the role of PQBP1 in innate immunity, particularly in HIV-1 recognition, is likely more recently evolved and specific to mammals .

This evolutionary perspective provides valuable insights into which aspects of PQBP1 function are fundamental (and thus conserved) versus those that may represent more recently evolved specializations in higher organisms.

Product Science Overview

Gene and Protein Structure

The PQBP1 gene is located on the X chromosome (Xp11.23) and encodes a nuclear polyglutamine-binding protein that contains a WW domain . This domain is crucial for protein-protein interactions, particularly with polyglutamine tracts found in certain transcription factors and disease-related proteins .

Function and Mechanism

PQBP1 acts as a scaffold protein, interacting with various splicing-related factors and regulating alternative splicing of target pre-mRNA species . It also plays a role in transcription regulation by interacting with the transcription machinery and can either activate or suppress transcription depending on the context . Additionally, PQBP1 is involved in the innate immune response and neuron development .

Clinical Significance

Mutations in the PQBP1 gene have been associated with several X-linked cognitive disabilities, including Renpenning syndrome 1 . These mutations can lead to various developmental and neurological issues, highlighting the importance of PQBP1 in normal brain function and development .

Research and Applications

Research on PQBP1 has provided insights into its role in cellular processes and its potential involvement in neurodegenerative diseases. The recombinant form of PQBP1 is used in various studies to understand its interactions and functions better . This research is crucial for developing potential therapeutic strategies for conditions associated with PQBP1 mutations .

Quick Inquiry

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