BPNT1 Human

3(2) 5-Bisphosphate Nucleotidase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Enzymatic Activity and Biochemical Role

BPNT1 belongs to the inositol monophosphatase superfamily and catalyzes the breakdown of PAP, a byproduct of sulfation reactions . Its activity is essential for maintaining cytoplasmic nucleotide homeostasis and preventing PAP accumulation, which is toxic to cellular processes .

SubstrateProductActivity Notes
PAPSAPS + 3'-phosphatePrimary substrate; BPNT1’s activity is ~1000-fold higher for PAPS vs. inositol phosphates
PAPAMP + phosphateSecondary substrate; hydrolysis prevents PAP-mediated toxicity
Inositol phosphatesNot activeNo hydrolysis of Ins(1)P, Ins(1,4)P2, or Ins(1,3,4,5)P4

Metabolic Pathways and Functional Partners

BPNT1 interacts with enzymes in sulfate activation and nucleotide metabolism:

Partner ProteinFunctionInteraction Score
PAPSS1/PAPSS2Catalyze sulfate activation (APS → PAPS)0.972–0.975
IMPAD1Golgi-resident PAP phosphatase0.929
ISYNA1Myo-inositol biosynthesis0.782
FLAD1Flavin adenine dinucleotide (FAD) synthesis0.569

These interactions highlight BPNT1’s role in regulating sulfation capacity and coordinating with inositol metabolism .

Pathological Implications

BPNT1 dysfunction has been linked to severe metabolic and neurological phenotypes:

Hepatic Deficiency in Bpnt1 Knockout Mice

PhenotypeSeverityMechanism
PAP accumulation (50-fold)Liver-specificDisrupts ribosome biogenesis, nucleolar architecture, and protein synthesis
HypoproteinemiaSerum albumin ↓36%Impaired hepatic protein production (e.g., albumin, cholesterol)
EdemaWhole-bodyOsmotic imbalance due to reduced serum albumin
Elevated liver enzymesALT/AST ↑Hepatocellular damage

Double knockout with Papss2 (blocking PAP synthesis) rescues these phenotypes, confirming PAP toxicity as a primary driver .

Lithium Inhibition and Neuronal Effects

Lithium, a treatment for bipolar disorder, inhibits BPNT1, leading to PAP accumulation. In C. elegans, BPNT1 loss silences ASJ chemosensory neurons, impairing dauer exit and pathogen avoidance . Similar mechanisms may underlie lithium’s neuroprotective effects in humans, particularly in dopamine-secreting neurons .

Tissue-Specific Expression

BPNT1 is widely expressed, with high activity in:

  • Liver: Critical for detoxification and protein synthesis .

  • Kidney: Involved in sulfate metabolism and osmoregulation .

  • Brain: Expressed in neurons and astrocytes; implicated in lithium’s therapeutic and side effects .

Therapeutic and Research Relevance

  • Lithium Sensitivity: BPNT1 inhibition by lithium may contribute to its efficacy in bipolar disorder, though nephrotoxicity remains a concern .

  • Disease Models: Bpnt1-deficient mice serve as models for studying hepatic failure, edema, and alopecia .

  • Sulfation Disorders: Dysregulation of BPNT1 or PAPSS enzymes could underlie unexplained metabolic syndromes .

Product Specs

Introduction
BPNT1, a member of the magnesium-dependent, lithium-sensitive phosphomono-esterase superfamily, catalyzes the conversion of PAPS (adenosine 3'-phosphate 5' phosphosulfate) to APS (adenosine 5'-phosphosulfate) and PAP (3'(2')-phosphoadenosine 5' phosphate) to AMP (adenosine 5'-phosphate) using magnesium as a cofactor. Ubiquitously expressed, BPNT1 reaches its highest levels in the brain and kidney. The potent inhibition of BPNT1 by lithium, a drug used to treat manic depression and bipolar affective disorder, suggests a potential role for BPNT1 in the etiology of mood disorders.
Description
Recombinant BPNT1, expressed in E. coli, is a single, non-glycosylated polypeptide chain containing 344 amino acids (1-308a.a.) with a molecular mass of 37.5 kDa. The N-terminus of BPNT1 is fused to a 36 amino acid His-tag. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered clear solution.
Formulation
The BPNT1 protein solution (0.5 mg/mL) is formulated in 20 mM Tris-HCl buffer (pH 8.0) containing 5 mM DTT, 0.1 M NaCl, and 10% glycerol.
Purity
Greater than 90% as determined by SDS-PAGE.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, store frozen at -20°C. Avoid repeated freeze-thaw cycles.
Synonyms
3'(2'), 5'-bisphosphate nucleotidase 1, Bisphosphate 3'-nucleotidase 1, PAP-inositol-1,4-phosphatase, PIP, EC 3.1.3.7, BPntase.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSMASS NTVLMRLVAS AYSIAQKAGM IVRRVIAEGD LGIVEKTCAT DLQTKADRLA QMSICSSLAR KFPKLTIIGE EDLPSEEVDQ ELIEDSQWEE ILKQPCPSQY SAIKEEDLVV WVDPLDGTKE YTEGLLDNVT VLIGIAYEGK AIAGVINQPY YNYEAGPDAV LGRTIWGVLG LGAFGFQLKE VPAGKHIITT TRSHSNKLVT DCVAAMNPDA VLRVGGAGNK IIQLIEGKAS AYVFASPGCK KWDTCAPEVI LHAVGGKLTD IHGNVLQYHK DVKHMNSAGV LATLRNYDYY ASRVPESIKN ALVP

Q&A

What is BPNT1 and what is its primary function in human cells?

BPNT1 is a metal-dependent, lithium-sensitive phosphatase that catalyzes the breakdown of 3′-phosphoadenosine 5′-phosphate (PAP), a byproduct of sulfation reactions utilizing 3′-phosphoadenosine 5′-phosphosulfate (PAPS) . The enzyme belongs to a conserved family that includes Golgi-resident PAP phosphatase (gPAPP), but BPNT1 specifically operates in the cytoplasm. Its primary function is preventing toxic accumulation of PAP, which is essential for normal cellular processes including protein synthesis and nucleolar function . Methodology for investigating BPNT1's function typically includes enzymatic assays with recombinant protein, subcellular fractionation studies, and knockout models to observe physiological consequences of BPNT1 deficiency.

How is BPNT1 activity regulated across different tissues?

BPNT1 shows tissue-specific regulation patterns with particularly important roles in liver and neuronal tissues. In liver, BPNT1 activity is crucial for maintaining protein synthesis and preventing PAP accumulation, correlating with the liver's high metabolic and protein synthesis demands . In neuronal tissues, BPNT1 is expressed in specific neurons, particularly those that secrete dopamine, epinephrine, or norepinephrine . To study this tissue-specific regulation, researchers should utilize tissue-specific knockout models, quantitative expression analysis across tissues, and comparative enzymatic activity assays accounting for the different microenvironments of each tissue type.

What are the known substrates of BPNT1 beyond PAP?

While PAP is the primary established substrate, BPNT1 has been shown to hydrolyze other 3′-phosphorylated nucleotides in vitro, including 3′-phosphocytosine 5′-phosphate . The enzyme shows specificity for 3′,5′-bisphosphorylated nucleotides. Researchers investigating alternative substrates should employ in vitro enzyme assays with purified recombinant BPNT1, HPLC or mass spectrometry to detect substrate hydrolysis, and comparative kinetic analyses to determine substrate preferences. While other substrates have been reported in vitro, analysis of Bpnt1 knockout tissues has primarily detected accumulation of PAP, with lesser amounts of PAPS, suggesting these may be the physiologically relevant substrates .

How can researchers accurately map BPNT1 expression in human tissues?

BPNT1 is expressed throughout various human tissues, with notable expression in the liver and throughout the mammalian brain . Accurate expression mapping requires multiple complementary approaches:

MethodApplicationKey Considerations
RNA-seq/qPCRQuantitative expression across tissuesInclude multiple reference genes
Western blottingProtein expression levelsValidate antibody specificity
ImmunohistochemistrySpatial distribution in tissue sectionsInclude knockout controls
Single-cell RNA-seqCell-type specific expressionSufficient sequencing depth
In situ hybridizationmRNA localization within tissuesProbe specificity validation

Researchers should correlate expression patterns with functional studies to understand the physiological significance of differential expression across tissues and cell types.

What are the most reliable assays for measuring BPNT1 enzyme activity?

For reliable measurement of BPNT1 enzymatic activity, researchers should consider multiple complementary approaches:

  • Colorimetric assays: A validated method involves isolating small-molecule extracts from tissues using boiling glycine, followed by a PAP-dependent enzymatic assay where color development rate depends on PAP/PAPS concentration .

  • HPLC-based methods: HPLC can resolve mono-, di-, and triphosphorylated nucleotides, allowing direct quantification of substrates and products. This approach revealed dramatic PAP accumulation in Bpnt1 knockout liver, with levels increasing 30-50 fold compared to wild-type .

  • Radioactive assays: Using radiolabeled substrates provides highly sensitive measurement, particularly useful with limited samples or when detecting low activity levels.

When implementing these assays, researchers must include appropriate controls (heat-inactivated enzyme), validate substrate specificity, optimize buffer conditions (particularly metal ion concentrations), and account for potential interference from other phosphatases.

How can researchers effectively generate and validate BPNT1 knockout models?

Developing effective BPNT1 knockout models requires careful methodology:

  • Generation strategies:

    • Homologous recombination has been successfully used for global Bpnt1 knockout mice

    • CRISPR/Cas9 systems for both cellular and animal models

    • Conditional knockout approaches may mitigate lethality issues seen in global knockouts

  • Validation methods:

    • Western blotting to confirm absence of protein expression

    • Enzymatic activity assays to confirm functional loss

    • HPLC or mass spectrometry to confirm substrate accumulation

    • Phenotypic characterization consistent with published models

  • Study design considerations:

    • Include heterozygous animals as controls (no haploinsufficiency has been observed)

    • Account for age-dependent phenotypes (Bpnt1 null mice develop phenotypes by approximately 45 days)

    • Examine tissue-specific effects, particularly in liver and neuronal tissues

    • Consider genetic interaction studies (e.g., with Papss2 mutants) to test mechanistic hypotheses

What methodological approaches reveal BPNT1's role in nucleolar function?

BPNT1 deficiency dramatically impacts nucleolar morphology and function, particularly in hepatocytes . To investigate these effects, researchers should:

  • Analyze nucleolar morphology:

    • Immunofluorescence with nucleolar markers (fibrillarin, nucleophosmin)

    • Transmission electron microscopy for ultrastructural analysis

    • Quantitative image analysis of nucleolar size, number, and architecture

  • Assess ribosomal RNA processing:

    • Northern blotting to detect processing intermediates

    • qRT-PCR for specific rRNA precursors

    • RNA-seq to comprehensively analyze rRNA processing defects

  • Evaluate ribosome biogenesis:

    • Polysome profiling to assess ribosome assembly

    • Ribosome half-transit time measurements

    • Pulse-chase labeling of rRNA

  • Examine nucleolar protein dynamics:

    • Fluorescence recovery after photobleaching (FRAP)

    • Live-cell imaging with fluorescently tagged nucleolar proteins

    • Proteomics of isolated nucleoli

Research in Bpnt1 knockout mice revealed hypertrophied nuclei with abnormal subnuclear structures and dramatically altered ultrastructure, including reduced inner membrane-bound DNA and disrupted rough endoplasmic reticulum .

How should researchers approach studying lithium effects on BPNT1?

Lithium inhibits BPNT1 activity, potentially contributing to its therapeutic effects in bipolar disorder . Methodologically sound approaches include:

  • Biochemical characterization:

    • In vitro enzymatic assays with varying lithium concentrations

    • Determination of inhibition kinetics (Ki values)

    • Competition studies with other ions

  • Cellular studies:

    • Measure PAP accumulation after lithium treatment

    • Assess reversibility of effects after lithium withdrawal

    • Compare with genetic BPNT1 knockdown

  • Neuronal specificity:

    • Evaluate effects in different neuronal populations

    • Correlate with expression patterns of cytosolic sulfotransferase (e.g., SSU-1 in C. elegans)

    • Assess impact on neurotransmitter systems, particularly dopaminergic neurons

  • Behavioral correlates:

    • Compare behavioral effects of lithium with genetic BPNT1 manipulation

    • Assess dose-dependency of behavioral and molecular effects

    • Investigate interaction with other lithium targets

Studies in C. elegans demonstrated that lithium causes selective dysfunction of ASJ neurons through BPNT1 inhibition, with effects that are reversible upon lithium withdrawal .

How does BPNT1 deficiency lead to liver dysfunction at the molecular level?

BPNT1 deficiency triggers a cascade of molecular events leading to severe liver dysfunction:

  • PAP accumulation: Knockout mice show 30-50 fold elevation of PAP in liver tissue .

  • Translational repression:

    • Significantly reduced incorporation of amino acids into newly synthesized proteins

    • Dramatic decrease in serum albumin production (0.9 g/dL vs normal 3.0 g/dL)

    • Reduced synthesis of apolipoproteins E and A1

  • Nucleolar dysfunction:

    • Abnormal nucleolar morphology and architecture

    • Accumulation of unprocessed ribosomal RNA

    • Disruption of ribosome biogenesis pathways

  • Cellular structural abnormalities:

    • Hypertrophied nuclei with abnormal subnuclear structures

    • Reduced inner membrane-bound DNA

    • Decreased contiguous rough endoplasmic reticulum

    • Irregular mitochondria and reduced glycogen

    • Accumulated lipid droplets containing cholesteryl esters and triglycerides

These molecular changes collectively lead to hypoproteinemia, hepatocellular damage, and in severe cases, whole-body edema and death .

What mechanisms connect PAP accumulation to hepatocellular damage?

The connection between PAP accumulation and hepatocellular damage involves several key mechanisms:

  • Inhibition of RNA processing:

    • PAP inhibits exoribonucleases involved in RNA processing

    • This disrupts ribosomal RNA maturation

    • Consequently affects ribosome biogenesis and protein synthesis capacity

  • Translational repression:

    • Impaired protein synthesis affects production of essential liver proteins

    • Reduced albumin production (36% reduction) leads to hypoalbuminemia and contributes to edema

    • Decreased apolipoprotein synthesis affects lipid metabolism, leading to lipid accumulation

  • Metabolic derangements:

    • Altered lipid metabolism leads to hepatic steatosis

    • Disrupted cholesterol handling (63% reduction in serum cholesterol)

    • Potential impairment of other metabolic functions

  • Cellular stress:

    • Evidence of hepatocellular damage (elevated ALT, AST, and ALKP)

    • Potential activation of stress response pathways

    • Progressive deterioration of hepatocyte function

The direct causality of PAP in this process is supported by genetic rescue experiments: double knockout mice lacking both Bpnt1 and Papss2 (blocking both PAP hydrolysis and synthesis) show complete reversal of liver phenotypes .

How does BPNT1 affect protein synthesis in hepatocytes?

BPNT1's impact on protein synthesis in hepatocytes involves multiple aspects of the translation machinery:

  • Direct translation effects:

    • In vivo radioactive metabolic labeling shows dramatically reduced incorporation of 3H-leucine into newly synthesized albumin in Bpnt1 null mice

    • Reduced production of other liver-derived proteins, including apolipoproteins E and A1

  • Ribosome biogenesis disruption:

    • Abnormal nucleolar morphology suggests disrupted ribosome production

    • PAP accumulation affects 5.8S rRNA processing

    • Ultrastructural changes include drastically reduced rough endoplasmic reticulum

  • Translational capacity:

    • Reduced availability of functional ribosomes limits global protein synthesis

    • Particularly affects highly translated liver-specific mRNAs

    • May have differential effects on specific mRNA populations

The severity of translation impairment correlates with PAP levels and phenotypic severity. Mice presenting with edema show more severe hypoalbuminemia (0.9 g/dL vs 1.9 g/dL in non-edematous knockouts and 3.0 g/dL in wild-type) .

What are the implications of BPNT1 research for human liver diseases?

BPNT1 research has several important implications for human liver diseases:

  • Novel pathomechanism:

    • Identifies PAP accumulation as a potential mechanism in certain liver pathologies

    • Establishes link between nucleotide metabolism and liver function

  • Hypoalbuminemia:

    • Bpnt1 null mice develop severe hypoalbuminemia (0.9 g/dL vs normal 3.0 g/dL)

    • Mirrors clinical presentations of certain liver diseases in humans

    • Suggests potential role for BPNT1 in unexplained hypoalbuminemia cases

  • Hepatic steatosis:

    • The significant lipid accumulation in Bpnt1 null hepatocytes resembles non-alcoholic fatty liver disease (NAFLD)

    • Provides novel connection between protein synthesis impairment and lipid accumulation

    • May identify new pathway involved in hepatic steatosis

  • Therapeutic implications:

    • The genetic rescue by Papss2 mutation suggests possible therapeutic strategies targeting PAPS synthesis

    • Identifies potential for modulating PAP levels as a therapeutic approach

    • Could inform understanding of unexplained drug-induced liver injury

  • Biomarker potential:

    • PAP levels might serve as biomarkers for specific liver pathologies

    • Nucleolar morphology changes could provide diagnostic insights

Researchers should investigate BPNT1 expression and function in human liver disease samples, screen for BPNT1 mutations in patients with unexplained liver dysfunction, and explore pharmacological approaches to modulate the PAP metabolic pathway.

How does lithium inhibition of BPNT1 potentially contribute to its therapeutic effects?

Lithium's inhibition of BPNT1 may contribute to its therapeutic effects through several mechanisms:

  • Selective neuronal effects:

    • Research in C. elegans demonstrated that lithium causes dysfunction of specific neurons (ASJ neurons) through BPNT1 inhibition

    • This selective effect may explain how lithium affects specific neuronal circuits rather than causing global brain dysfunction

  • Neurotransmitter modulation:

    • In humans, PAP (which accumulates when BPNT1 is inhibited) is found in neurons that secrete dopamine, epinephrine, or norepinephrine

    • These neurotransmitters are implicated in mood regulation and bipolar disorder

    • "Silencing dopaminergic neurons [through BPNT1 inhibition] would make you less manic because of how dopamine affects the brain"

  • Pathway integration:

    • BPNT1 inhibition represents a novel mechanism that may complement other known lithium targets

    • May explain aspects of lithium's effects not accounted for by other mechanisms

    • Provides new perspective on lithium's complex actions in the brain

The selective effect on specific neurons is particularly relevant, as lithium's therapeutic benefit comes without global neuronal dysfunction. In C. elegans, this selectivity is partly due to limited expression of cytosolic sulfotransferase SSU-1 in the ASJ neuron pair .

What neuronal populations in humans most heavily rely on BPNT1 function?

Based on current research, the neuronal populations most likely to rely on BPNT1 function include:

  • Monoaminergic neurons:

    • PAP, which BPNT1 degrades, is typically found in neurons that secrete dopamine, epinephrine, or norepinephrine

    • These neurons play critical roles in mood regulation, reward processing, and arousal

  • Specific chemosensory neurons:

    • In C. elegans, BPNT1 is critical for function of ASJ chemosensory neurons

    • This suggests potential importance in human sensory processing neurons

  • High BPNT1-expressing neurons:

    • BPNT1 is expressed throughout the mammalian brain

    • Neurons with particularly high expression may be most dependent on its function

    • Detailed expression mapping in human brain is needed

The differential dependence on BPNT1 may explain why lithium and other treatments have selective effects on behavior despite their potential to affect multiple neuronal types. Research methodologies should include single-cell RNA sequencing, immunohistochemistry, and functional studies correlating BPNT1 levels with neuronal properties.

How does BPNT1 dysfunction affect neural signaling pathways?

BPNT1 dysfunction impacts neural signaling through multiple mechanisms:

  • Neuronal silencing:

    • Evidence from C. elegans shows that BPNT1 inhibition or knockout causes neurons to enter a dormant state

    • Affects neuronal excitability and responsiveness

    • May preferentially impact high-activity neurons

  • Behavioral consequences:

    • In C. elegans, BPNT1 loss affects behaviors dependent on ASJ neurons, such as dauer exit and pathogen avoidance

    • Suggests effects on both sensory processing and behavioral outputs

    • May explain specific behavioral effects of lithium

  • Signaling pathway interactions:

    • PAP accumulation may affect multiple intracellular signaling pathways

    • Could interact with neurotransmitter synthesis or release mechanisms

    • May have downstream effects on neuronal gene expression

  • Circuit-level effects:

    • Selective effects on specific neurons would alter circuit dynamics

    • May affect balance between excitatory and inhibitory transmission

    • Could modify information processing in neural networks

The selective nature of these effects is particularly important, as it helps explain how therapeutic benefits can occur without global neural dysfunction, consistent with lithium's clinical properties.

What is the relevance of C. elegans BPNT1 findings to human neurological research?

C. elegans BPNT1 findings provide valuable insights for human neurological research:

  • Conservation of mechanism:

    • The fundamental role of BPNT1 in PAP metabolism is conserved across species

    • Lithium inhibition of BPNT1 occurs in both C. elegans and mammals

    • Provides molecular framework for understanding lithium's effects

  • Selective neuronal vulnerability:

    • In C. elegans, BPNT1 loss affects specific neurons (ASJ) rather than all neurons

    • This selective vulnerability could parallel specific effects in human brain circuits

    • Helps explain how lithium can affect mood without causing global brain dysfunction

  • Molecular pathway insights:

    • C. elegans studies identified that the selective effect of lithium is partly due to limited expression of cytosolic sulfotransferase SSU-1 in specific neurons

    • Suggests examining sulfotransferase expression patterns in human brain regions

    • Provides testable hypotheses about cell-specific vulnerability

  • Behavioral correlates:

    • BPNT1 mutation in C. elegans affects complex behaviors requiring sensory integration

    • Similar principles may apply to lithium's effects on human mood and behavior

    • Offers framework for understanding circuit-level effects

These findings suggest new directions for human studies, including mapping BPNT1 and sulfotransferase expression in human brain, examining PAP metabolism in neuropsychiatric disorders, and investigating BPNT1 variants in treatment-responsive versus non-responsive patients.

How do BPNT1 and gPAPP functionally differentiate despite their similar enzymatic activities?

Despite their similar enzymatic activities, BPNT1 and gPAPP have evolved distinct physiological roles:

This functional differentiation provides an elegant example of how subcellular compartmentalization, combined with tissue-specific expression patterns, allows related enzymes to evolve specialized roles in distinct physiological processes.

What approaches can resolve contradictions in BPNT1 function across model systems?

To resolve contradictory findings across model systems, researchers should employ:

  • Standardized methodologies:

    • Consistent assay conditions for BPNT1 activity measurements

    • Standardized knockout validation approaches

    • Comparable phenotypic characterization protocols

  • Cross-species functional analysis:

    • Complementation studies with BPNT1 orthologs

    • Analysis of conserved versus divergent domains

    • Detailed comparative expression mapping

  • Integrated multiple approach strategy:

ApproachApplicationKey Advantage
GeneticKnockout/knockdown modelsHigh specificity
BiochemicalIn vitro enzyme studiesMolecular mechanism
PharmacologicalLithium and other inhibitorsTemporal control
StructuralCrystallography/modelingStructure-function
ComputationalSystems biology modelingIntegration of data
  • Context-dependent analyses:

    • Evaluate tissue-specific requirements

    • Consider developmental timing

    • Examine environmental influences on phenotypes

  • Direct hypothesis testing:

    • Design experiments specifically addressing contradictions

    • Use rescue experiments to test causal mechanisms

    • Employ genome editing for precise genetic manipulation

This multi-faceted approach can help reconcile apparently contradictory findings by identifying context-dependent factors and species-specific differences in BPNT1 function.

How does genetic rescue inform mechanism in BPNT1 research?

Genetic rescue experiments have provided critical mechanistic insights in BPNT1 research:

These genetic experiments demonstrate the power of combining mutations in related pathway components to dissect mechanisms and identify potential therapeutic approaches.

What is the relationship between BPNT1, PAP metabolism, and RNA processing?

BPNT1 connects PAP metabolism to RNA processing through several mechanisms:

  • Exoribonuclease inhibition:

    • PAP, which accumulates in BPNT1 deficiency, inhibits exoribonucleases

    • These enzymes are critical for proper RNA processing, particularly ribosomal RNA

    • Creates link between sulfur metabolism and RNA maturation

  • Nucleolar dysfunction:

    • Bpnt1 knockout mice show dramatic nucleolar abnormalities

    • This correlates with accumulation of unprocessed ribosomal RNA

    • Demonstrates importance of PAP metabolism for nucleolar function

  • Translational consequences:

    • Disrupted rRNA processing affects ribosome biogenesis

    • Leads to reduced protein synthesis capacity

    • Creates cascade from PAP metabolism to translation efficiency

  • Specificity of effects:

    • The effects appear most pronounced in tissues with high protein synthesis demands (like liver)

    • Suggests tissue-specific vulnerability based on translational requirements

    • Explains why liver is particularly affected in Bpnt1 knockout mice

This relationship establishes an unexpected but critical connection between sulfur metabolism and RNA processing, revealing how disruption of a seemingly unrelated metabolic pathway can profoundly impact fundamental cellular processes like translation.

Product Science Overview

Biological Properties

BPNT1 is primarily involved in the hydrolysis of 3’(2’)-phosphoadenosine 5’-phosphate (PAP) to adenosine monophosphate (AMP) and inositol 1,4-bisphosphate (Ins(1,4)P2) to inositol 4-phosphate . It also has the ability to hydrolyze adenosine 3’-phosphate 5’-phosphosulfate (PAPS) to adenosine 5’-phosphosulfate (APS) . This activity is crucial for preventing the toxic accumulation of PAP, which can inhibit various proteins, including PAPS-utilizing enzymes such as sulfotransferases and RNA processing enzymes .

Expression Patterns and Tissue Distribution

BPNT1 is expressed in various tissues, with significant expression in the testicular tissue, where it is also known as Testicular Tissue Protein Li 29 or Epididymis Luminal Protein 20 . The protein is highly conserved across species, with the human protein being 92% identical to its mouse counterpart .

Biological Functions

The primary function of BPNT1 is to regulate nucleotide metabolism through its phosphatase activity. By converting PAP to AMP, BPNT1 plays a role in inositol recycling and phosphoinositide metabolism . This regulation is essential for maintaining cellular homeostasis and preventing the accumulation of toxic intermediates .

Modes of Action

BPNT1’s enzymatic activity is magnesium-dependent, and it acts as a phosphatase to hydrolyze specific nucleotide substrates . Lithium, a major drug used to treat manic depression, acts as an uncompetitive inhibitor of BPNT1, which may account for lithium-induced nephrotoxicity .

Regulatory Mechanisms

The activity of BPNT1 is regulated by inositol signaling pathways, which are crucial for its role in nucleotide metabolism . The inhibition of BPNT1 by lithium highlights the enzyme’s significance in therapeutic contexts and its potential impact on drug-induced side effects .

Associated Diseases

Mutations or dysregulation of BPNT1 have been associated with several diseases, including Brachydactyly, Type B1 and Fanconi Anemia, Complementation Group D1 . These associations underscore the importance of BPNT1 in maintaining normal cellular functions and its potential role in disease pathogenesis.

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.