IMPA1 Human

Inositol Monophosphatase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

IMPA1 regulates two primary pathways:

  • Phosphatidylinositol Recycling: Converts inositol monophosphates (e.g., Ins(1)P, Ins(4)P) to free inositol, sustaining PI synthesis for secondary messengers like IP3 and diacylglycerol (DAG) .

  • Cellular Homeostasis: Modulates autophagy, apoptosis, and neuronal excitability via inositol depletion or accumulation .

Key Interactions:

  • Binds Bergmann glial S100B and calbindin, influencing calcium signaling .

  • Regulates mTOR and epithelial-mesenchymal transition (EMT) in cancer .

Intellectual Disability

A homozygous IMPA1 mutation (c.238C>T) causes severe autosomal recessive intellectual disability (ID) and behavioral abnormalities. Patient-derived neurons exhibit:

  • Impaired neurogenesis and glial differentiation .

  • Cell cycle arrest and apoptosis in neural progenitor cells .
    These defects are rescued by myo-inositol supplementation, confirming IMPA1’s role in neuronal survival .

Cancer

IMPA1 is upregulated in triple-negative breast cancer (TNBC) and castration-resistant prostate cancer (CRPC):

  • TNBC: Drives metastasis via mTOR activation and EMT .

  • CRPC: Maintains cancer stemness by binding IMPDH2 to sustain guanylate nucleotide synthesis .
    Pharmacological IMPA1 inhibition reduces tumor growth in xenograft models .

Experimental Tools and Inhibitors

  • Recombinant IMPA1: Used to study enzymatic kinetics and lithium’s effects .

  • Chemical Inhibitors:

    • L-690,330: Competitive inhibitor (Ki = 1.5 µM) with limited bioavailability .

    • L-690,488: Cell-permeable prodrug of L-690,330 .

Therapeutic Implications

ConditionIMPA1 InvolvementTherapeutic Approach
Bipolar DisorderLithium-mediated IMPA1 inhibition reduces PIP2 turnover and neuronal excitability .Lithium, valproate, carbamazepine
CRPCIMPA1/inositol/IMPDH2 axis sustains stemness and ABT resistance .IMPDH2 inhibitors (e.g., mycophenolate)
Intellectual DisabilityIMPA1 mutations disrupt neurogenesis; inositol supplementation rescues defects .Dietary inositol trials

Unresolved Questions

  • Why do IMPA1 knockout mice retain normal brain inositol levels despite reduced IMPase activity ?

  • How does IMPA1 coordinate with IMPA2 in tissues where both are expressed ?

  • Can IMPA1-specific inhibitors achieve lithium’s therapeutic effects without off-target toxicity ?

Product Specs

Introduction
Inositol monophosphatase 1 (IMPA1) plays a crucial role in generating inositol, a vital component for synthesizing phosphatidylinositol and polyphosphoinositides. It functions within the phosphatidylinositol signaling pathway by hydrolyzing inositol monophosphates. IMPA1 is significant as a target for lithium's action in the brain. This enzyme exhibits magnesium-dependent phosphatase activity, which therapeutic lithium concentrations can inhibit. The therapeutic effects of lithium in treating bipolar disorder, such as its anti-manic and anti-depressive properties, might be linked to its inhibition of inositol monophosphate hydrolysis, consequently reducing the availability of inositol for phosphatidylinositol synthesis.
Description
Recombinant human IMPA1, expressed in E. coli, is a single, non-glycosylated polypeptide chain with a His tag (20 amino acids) at its N-terminus. This protein consists of 297 amino acids (including the 20-amino acid His tag; the IMPA1 sequence spans from amino acid 1 to 277) and has a molecular weight of 32.3 kDa. The purification process for IMPA1 involves proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The IMPA1 solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), keep the IMPA1 vial refrigerated at 4°C. For extended storage, freeze the solution at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage to enhance stability. Minimize repeated freeze-thaw cycles to prevent protein degradation.
Purity
The purity of IMPA1 is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
Inositol monophosphatase 1, IMP 1, IMPase 1, Inositol-1(or 4)-monophosphatase 1, Lithium-sensitive myo-inositol monophosphatase A1, IMPA1, IMPA, IMP.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MADPWQECMD YAVTLARQAG EVVCEAIKNE MNVMLKSSPV DLVTATDQKV EKMLISSIKE KYPSHSFIGE ESVAAGEKSI LTDNPTWIID PIDGTTNFVH RFPFVAVSIG FAVNKKIEFG VVYSCVEGKM YTARKGKGAF CNGQKLQVSQ QEDITKSLLV TELGSSRTPE TVRMVLSNME KLFCIPVHGI RSVGTAAVNM CLVATGGADA YYEMGIHCWD VAGAGIIVTE AGGVLMDVTG GPFDLMSRRV IAANNRILAE RIAKEIQVIP LQRDDED.

Q&A

What is the primary biochemical function of IMPA1 in human cellular metabolism?

IMPA1 (inositol monophosphatase 1) is a critical enzyme responsible for the dephosphorylation of inositol-1-phosphate to free inositol. This enzymatic function serves two essential metabolic purposes: facilitating de novo biosynthesis of inositol and enabling the recycling of inositol from inositol polyphosphates. Through these mechanisms, IMPA1 plays a fundamental role in the phosphatidylinositol (PI) signaling pathway, which functions as a key intracellular second messenger system in the brain and numerous other tissues throughout the body . The enzyme's activity directly influences the availability of free inositol for subsequent incorporation into phosphatidylinositol 4,5-bisphosphate (PIP2), which is essential for various cellular signaling cascades involved in neuronal function and other cellular processes .

In neurons specifically, IMPA1 activity has been shown to be crucial for the maintenance of proper calcium signaling dynamics. Research indicates that when IMPA1 function is compromised, there is a reduction in receptor-activated calcium release from intracellular stores and a delay in PIP2 resynthesis after phospholipase C (PLC) signaling events . These findings suggest that IMPA1 is not merely involved in basic metabolite processing but plays an active role in regulating neuronal excitability and signaling.

How is IMPA1 gene expression regulated in different neural cell types?

The expression regulation of IMPA1 exhibits distinct patterns across neural cell lineages, with significant implications for understanding both normal development and pathological conditions. Transcriptome analyses from patient-derived cells have revealed that IMPA1 deficiency leads to extensive deregulation of gene expression pathways necessary for neurogenesis, with concurrent upregulation of gliogenic genes . This suggests that IMPA1 expression itself likely follows developmental stage-specific regulation patterns.

Research utilizing induced pluripotent stem cells (iPSCs) differentiated into various neural lineages has demonstrated that IMPA1 function appears to be particularly critical in neuronal progenitor cells (NPCs) compared to other neural cell types. While IMPA1 deficiency profoundly affects cell cycle progression and survival in NPCs, it does not similarly impact these processes in iPSCs, glial progenitor cells, or differentiating astrocytes . This cell-type specificity indicates sophisticated regulatory mechanisms that modulate IMPA1 expression and function differently across neural lineages.

The regulatory relationship between IMPA1 and lithium response pathways has also been investigated, with evidence suggesting that lithium's inhibition of IMPA1 can alter transcriptional programs in human cortical neurons. Following lithium treatment, transcriptome analyses have identified down-regulation of signaling pathways related to glutamate, a key excitatory neurotransmitter in the human brain , further illustrating how IMPA1 regulation intersects with broader neuronal signaling networks.

What are the known pathogenic variants of IMPA1 in humans and their associated phenotypes?

The most well-documented pathogenic variant of IMPA1 is a homozygous frameshift mutation (c.489_493dupGGGCT) that has been linked to severe intellectual disability (ID) in geographically isolated consanguineous families. This specific mutation has been identified in two distinct populations: a large family in Northeastern Brazil and another consanguineous cohort in Pakistan . In the Brazilian cohort, which has been extensively studied, the family structure includes 28 consanguineous marriages with 59 genotyped family members, among which nine individuals were initially identified with the homozygous mutation and corresponding severe intellectual disability phenotype .

This pathogenic variant is now recognized as causing Mental Retardation, Autosomal Recessive 59 (MRT59), a condition characterized by severe intellectual disability and frequently accompanied by disruptive behavior . The intellectual disability associated with homozygous IMPA1 mutation appears to be substantial, as evidenced by systematic neuropsychological assessment using standardized measures such as the Wechsler Abbreviated Scale of Intelligence (WASI) .

The specificity of the phenotype is notable - despite IMPA1's widespread expression throughout the body, the pathology associated with its mutation appears predominantly neurological in nature, with minimal reported effects on other organ systems. This neural specificity aligns with experimental findings in cellular models that demonstrate IMPA1 deficiency specifically affects neuronal progenitor cell survival and differentiation while sparing other cell lineages .

How does IMPA1 mutation affect neurophysiological function as measured by quantitative EEG?

Electrophysiological investigations using quantitative EEG have revealed specific neurophysiologic signatures associated with IMPA1 mutation. In a family-based study of the Brazilian cohort with the homozygous IMPA1 frameshift mutation, researchers identified several significant abnormalities in resting-state EEG measures between mutation carriers and wild-type relatives.

During eyes-open conditions, IMPA1 mutation was associated with:

  • Relative decreases in frontal theta band power

  • Altered alpha-band variability without regional specificity

During eyes-closed conditions, researchers observed:

  • Altered dominant theta frequency variability specifically in central and parietal regions

Statistical analysis using multivariate family-based association testing with generalized estimating equations (FBAT-GEE) identified significant associations between the IMPA1 mutation and several quantitative EEG phenotypes. Specifically, multivariate testing revealed significant associations with θ-band power (χ² = 18.451, p = .018) and dominant α-band variability (χ² = 19.771, p = .011) for the eyes-open condition, and dominant θ-band variability (χ² = 15.848, p = .045) for the eyes-closed condition .

Subsequent univariate analysis of individual scalp regions demonstrated that lower than expected θ power over the left frontal scalp region was significantly associated (Z = −2.211, p = .027) with the mutated allele, while higher than expected θ power over the right frontal scalp region was also significantly associated (Z = 2.248, p < .05) . These findings represent the first human in vivo phenotypic assessment of brain function disturbances associated with a loss-of-function IMPA1 mutation, providing insight into the pathophysiologic mechanisms underlying the associated intellectual disability.

What demographic and clinical characteristics are observed in patients with IMPA1-associated intellectual disability?

Patients with IMPA1-associated intellectual disability (MRT59) present with a distinct clinical profile that has been systematically documented in the Brazilian cohort. Demographic and clinical data from this population reveal important insights about the condition.

The following table summarizes the demographic characteristics of individuals stratified by IMPA1 genotype from one study :

IMPA1 MutationnFemaleMalePsychosisAge (SD)
HOM +/+440251.5 (5.3)
HET +/−927344.8 (12.6)
WT −/−17116345.0 (12.4)

In the MRT59 population, many patients were taking various medications, including antipsychotics, benzodiazepines, SSRIs, antihypertensives, oral hypoglycemics, and allopurinol . This suggests comorbid conditions requiring pharmacological management are common in this population.

Functional assessment of MRT59 patients using standardized measures such as the Functional Independence Measure Scale (FIM) has revealed significant impairments in daily living skills, confirming the profound impact of IMPA1 deficiency on adaptive functioning . The intellectual disability is categorized as severe based on DSM-V criteria for Intellectual Disability Severity.

Despite these significant neurological and cognitive impairments, structural brain imaging has not revealed consistent abnormalities. MRI findings in one subject from the Brazilian cohort showed no structural abnormalities and no reduction of myo-inositol in the basal ganglia, suggesting that neural disturbances may be more present and detectable at the circuit/systems level rather than gross anatomical changes .

What cellular models are most effective for studying IMPA1 function and dysfunction?

The development of induced pluripotent stem cell (iPSC) models has emerged as a particularly powerful approach for investigating IMPA1 function in human neural cells. Researchers have successfully generated iPSCs from patients with IMPA1 mutations and neurotypical controls, which can then be differentiated into specific neural cell types of interest . This methodology allows for the study of IMPA1's role in human neural development and function in a system that maintains the genetic background of affected individuals.

For studying IMPA1 in the context of neuronal development, protocols for differentiating iPSCs into hippocampal dentate gyrus-like neurons have proven especially valuable. This approach has revealed that IMPA1-deficient neuronal progenitor cells (NPCs) exhibit substantial deficits in proliferation and neurogenic potential . Specifically, at low passage NPCs (P1 to P3), researchers observed cell cycle arrest, apoptosis, progressive change to a glial morphology, and reduction in neuronal differentiation.

The validity of these cellular phenotypes has been confirmed through two complementary approaches:

  • Rescue experiments using myo-inositol supplemented media during differentiation of patient-derived iPSCs into neurons

  • Recapitulation of the phenotype in control NPCs expressing shRNA against IMPA1 (shIMPA1)

Additionally, CRISPR-Cas9 gene editing has been employed to create IMPA1-deleted cell lines, which provide a clean genetic background for studying the specific effects of IMPA1 absence on cellular processes like PIP2 resynthesis and calcium signaling . These engineered cellular models have demonstrated that treatment effects of lithium on receptor-activated calcium release and PIP2 dynamics are abrogated in IMPA1-deleted cells, confirming the specificity of these effects to IMPA1 inhibition.

For advanced electrophysiological studies, human forebrain cortical neurons derived from iPSCs have been utilized to investigate how IMPA1 inhibition (via lithium treatment) affects neuronal excitability and calcium signaling . This model system allows for detailed functional assessments that would be impossible to perform in patient brain tissue.

What are the most sensitive biochemical assays for measuring IMPA1 activity in human samples?

Measuring IMPA1 enzymatic activity requires specialized biochemical assays that can detect the dephosphorylation of inositol monophosphate to free inositol. The gold standard approaches involve quantifying either the reduction of substrate or the production of inositol product.

For determining IMPase activity levels in brain tissue, researchers have successfully employed assays that can detect regional differences in enzyme activity. In studies with IMPA1 deficient mice, IMPase activity levels were found to be reduced by up to 65% in hippocampus , demonstrating the sensitivity of these assays for detecting partial reductions in enzyme function.

For more sensitive cellular-level analysis, researchers have developed assays to measure phosphatidylinositol 4,5-bisphosphate (PIP2) resynthesis dynamics following PLC activation. These assays can detect delays in PIP2 recovery that result from IMPA1 inhibition or deficiency . Additionally, calcium imaging techniques using fluorescent indicators have proven valuable for assessing the downstream functional consequences of altered IMPA1 activity on receptor-activated calcium release from intracellular stores.

For genetic confirmation of IMPA1 mutations, standard DNA sequencing techniques are employed. In the Brazilian cohort, for example, genotyping identified the specific frameshift mutation (c.489_493dupGGGCT) in homozygosity among affected individuals .

How does IMPA1 activity influence neuronal excitability and synaptic transmission?

IMPA1 exerts significant influence on neuronal excitability and synaptic transmission through its critical role in phosphoinositide metabolism. Research has demonstrated that inhibition of IMPA1, whether through genetic deletion or pharmacological means (e.g., lithium treatment), results in measurable changes to neuronal excitability parameters.

In human forebrain cortical neurons, treatment with lithium—which inhibits IMPA1—reduces neuronal excitability and dampens calcium signals . This effect appears to be mediated through IMPA1's role in the maintenance of phosphatidylinositol 4,5-bisphosphate (PIP2) levels, a critical phospholipid that regulates numerous ion channels and signaling processes at the synapse. When IMPA1 is inhibited, there is a delay in PIP2 resynthesis following receptor activation, which consequently affects downstream signaling cascades .

Transcriptome analyses of human cortical neurons treated with lithium have revealed downregulation of signaling pathways related to glutamate, the primary excitatory neurotransmitter in the brain . This finding suggests that IMPA1 inhibition may specifically modulate excitatory neurotransmission, potentially explaining some of the therapeutic effects observed with lithium in conditions characterized by neuronal hyperexcitability.

The role of IMPA1 in synaptic transmission is further supported by electrophysiological findings from individuals with IMPA1 mutations. Quantitative EEG studies have identified altered neural oscillation patterns, particularly in theta and alpha frequency bands, which are important for coordinated network activity in the brain . These changes in oscillatory activity likely reflect altered synaptic function resulting from disrupted phosphoinositide signaling.

What is the relationship between IMPA1 function and neural progenitor cell proliferation and differentiation?

IMPA1 plays a critical and remarkably specific role in neural progenitor cell (NPC) development, with clear implications for neurogenesis. Studies using patient-derived induced pluripotent stem cells (iPSCs) have revealed that IMPA1 deficiency leads to substantial deficits in NPC proliferation and neurogenic potential .

The effects of IMPA1 deficiency on NPCs are both profound and progressive. At low passage (P1 to P3), IMPA1-deficient NPCs exhibit:

  • Cell cycle arrest

  • Increased apoptosis

  • Progressive changes toward glial morphology

  • Reduced capacity for neuronal differentiation

Importantly, these effects appear to be highly specific to the neuronal lineage. When researchers examined the impact of IMPA1 deficiency across different cell types, they found that it did not affect cell cycle progression or survival in iPSCs or glial progenitor cells, nor did it impair astrocyte differentiation . This cell-type specificity suggests that NPCs have unique requirements for inositol metabolism that are not shared by other neural or progenitor cell populations.

The causal relationship between IMPA1 deficiency and impaired neurogenesis has been confirmed through rescue experiments. When IMPA1-deficient iPSCs were differentiated into neurons in media supplemented with myo-inositol, the neurogenic deficits were reversed . Conversely, when control NPCs were made to express shRNA against IMPA1 (shIMPA1), they showed reduced neurogenic potential similar to that observed in patient-derived cells .

Transcriptome analysis has provided further insight into the molecular mechanisms underlying these phenotypes. NPCs and neurons derived from patients with IMPA1 mutations show extensive deregulation of gene expression affecting pathways necessary for neurogenesis, alongside upregulation of gliogenic genes . This suggests that IMPA1's influence on neural development extends beyond its enzymatic function to include regulation of broader transcriptional programs that govern cell fate determination in the nervous system.

How does lithium inhibition of IMPA1 contribute to its therapeutic effects in bipolar disorder?

Lithium has been the standard pharmacological treatment for bipolar disorder for over 50 years, yet the precise molecular mechanisms underlying its therapeutic effects remain incompletely understood. IMPA1 inhibition represents one of lithium's most well-characterized molecular actions, with significant evidence pointing to this interaction as potentially central to lithium's mood-stabilizing properties.

Lithium inhibits IMPA1 with a Ki within the therapeutic range required for effective treatment of bipolar disorder . This inhibition affects the recycling of inositol and consequently influences the availability of phosphatidylinositol 4,5-bisphosphate (PIP2), a critical signaling molecule in neurons. Research has demonstrated that therapeutic concentrations of lithium reduce receptor-activated calcium release from intracellular stores and delay PIP2 resynthesis—effects that are completely abrogated in IMPA1-deleted cells . This provides strong evidence that IMPA1 inhibition is a specific mechanism of lithium action.

In human forebrain cortical neurons, lithium treatment reduces neuronal excitability and dampens calcium signals, suggesting a potential mechanism for stabilizing aberrant neural activity in bipolar disorder . Transcriptome analyses following lithium treatment have revealed downregulation of glutamate signaling pathways , which is particularly relevant given that glutamate is the primary excitatory neurotransmitter and its dysregulation has been implicated in mood disorders.

Supporting the hypothesis that IMPA1 inhibition contributes to lithium's therapeutic effects, genetic studies in mice have shown that IMPA1 knockout animals exhibit lithium-like behaviors in certain experimental paradigms. Specifically, IMPA1-/- mice display increased motor activity in both open-field and forced-swim tests, as well as increased sensitivity to pilocarpine-induced seizures—phenotypes that parallel some of lithium's behavioral and physiological effects .

What does research on IMPA1-deficient models reveal about lithium's mechanism of action?

Studies of IMPA1-deficient models have provided valuable insights into lithium's mechanisms of action, offering a unique perspective that complements pharmacological inhibition studies. The IMPA1-/- mouse represents a novel model for studying inositol homeostasis and has demonstrated that genetic inactivation of IMPA1 can mimic certain actions of lithium .

One of the most striking findings from IMPA1 knockout mice is that homozygous deletion (IMPA1-/-) results in embryonic lethality between days 9.5 and 10.5 post coitum (p.c.), highlighting the essential role of IMPA1 in early development . Intriguingly, this lethality can be reversed by myo-inositol supplementation administered to pregnant mothers , demonstrating that the developmental defects are specifically related to inositol deficiency rather than other potential functions of the IMPA1 protein.

Behavioral analysis of IMPA1-/- mice has revealed phenotypes that resemble certain effects of lithium treatment, including:

  • Increased motor activity in the open-field test

  • Increased activity in the forced-swim test

  • Strongly increased sensitivity to pilocarpine-induced seizures

The last finding is particularly noteworthy as it supports the hypothesis that IMPA1 represents a physiologically relevant target for lithium's effects on neuronal excitability. These parallel effects between genetic IMPA1 deficiency and lithium treatment provide compelling evidence that IMPA1 inhibition contributes significantly to lithium's therapeutic actions.

Cellular studies using human iPSC-derived neurons with IMPA1 deficiency have further illuminated lithium's mechanisms, particularly in relation to phosphoinositide signaling and calcium dynamics. Research has demonstrated that lithium treatment reduces receptor-activated calcium release and delays PIP2 resynthesis—effects that are eliminated in IMPA1-deleted cells . This confirms that these specific actions of lithium are mediated through IMPA1 inhibition rather than lithium's other potential molecular targets.

What are the current limitations in studying IMPA1 function in human brain tissue?

Investigating IMPA1 function in human brain tissue presents several significant methodological challenges that limit our comprehensive understanding of its role in neural physiology and pathology. One fundamental limitation is the inability to directly sample and analyze living human brain tissue in most research contexts, forcing researchers to rely on indirect measures or post-mortem samples that may not accurately reflect in vivo conditions.

Even in cases where human subjects with known IMPA1 mutations are available for study, accessing brain tissue for detailed biochemical analysis remains impractical. This challenge is evident in research with the Brazilian IMPA1 mutation cohort, where despite identifying clear genetic mutations and associated cognitive phenotypes, researchers noted "difficulties in transportation and access to healthcare services in the isolated region where the patients live," limiting them to performing MRI scans on only a single patient as a "proof-of-concept" . These logistical constraints severely restrict the collection of crucial physiological data.

Current neuroimaging methods also present limitations. While magnetic resonance spectroscopy (MRS) can theoretically measure brain inositol levels, studies have shown that parenchymal inositol concentrations may not be detectably altered even in individuals with confirmed IMPA1 mutations . This suggests that either the resolution of current imaging techniques is insufficient to detect subtle cellular changes, or that compensatory mechanisms maintain bulk tissue inositol levels despite altered enzymatic activity.

The interpretation of IMPA1's role is further complicated by medication effects in patient populations. Many individuals with IMPA1 mutations are treated with various psychotropic and other medications, including antipsychotics, benzodiazepines, and SSRIs , which may independently affect inositol metabolism and neural function, potentially confounding research findings.

What novel experimental approaches could advance our understanding of IMPA1 in neurodevelopmental disorders?

Advancing our understanding of IMPA1's role in neurodevelopmental disorders will require innovative experimental approaches that overcome current limitations. Several promising methodologies could significantly accelerate progress in this field.

Brain organoids derived from patient iPSCs represent a particularly powerful emerging approach. These three-dimensional cellular models can recapitulate aspects of human brain development and organization that are impossible to study in conventional two-dimensional cultures. For IMPA1 research specifically, brain organoids could allow examination of how IMPA1 deficiency affects not just individual cells but also circuit formation, cell migration, and region-specific development—all processes relevant to intellectual disability. Building on existing successes with iPSC-derived neuronal cultures , brain organoids could provide a more comprehensive model system.

Advanced genetic approaches using CRISPR-Cas9 technology offer another promising avenue. While complete IMPA1 knockout in mice causes embryonic lethality , conditional and/or inducible knockout models could circumvent this limitation, allowing investigation of IMPA1 function in specific brain regions and developmental time points. Human cellular models with precise, isogenic CRISPR-engineered mutations would similarly provide clean genetic backgrounds for detailed mechanistic studies.

Multimodal in vivo assessment combining neuroimaging with electrophysiological and behavioral measures could provide a more comprehensive understanding of how IMPA1 mutations affect brain function across multiple scales. While some work has been done using quantitative EEG , combining this with functional MRI, MR spectroscopy, and detailed cognitive assessment would create a more complete picture of how IMPA1 deficiency alters neural circuit function.

Pharmacological rescue experiments, building on the observation that embryonic lethality in IMPA1 knockout mice can be reversed with myo-inositol supplementation , could lead to novel therapeutic approaches. Systematically testing various compounds that modulate inositol metabolism or downstream signaling pathways could identify potential treatments for IMPA1-associated intellectual disability.

Lastly, broader genetic studies investigating potential modifier genes could help explain the variable penetrance and expressivity observed in IMPA1-associated disorders. Whole genome sequencing of affected individuals and family members could identify genetic factors that influence the severity of phenotypes resulting from IMPA1 mutations, potentially revealing new therapeutic targets.

What are the key outstanding questions regarding IMPA1 function in human neurobiology?

Despite significant advances in our understanding of IMPA1's role in human neurobiology, several crucial questions remain unanswered. Perhaps most fundamentally, we still lack a complete understanding of why IMPA1 deficiency specifically affects neuronal development and function while largely sparing other tissues, despite the enzyme's widespread expression throughout the body. This neural specificity is evident in both human patients with IMPA1 mutations, who primarily present with intellectual disability , and in cellular models where IMPA1 deficiency specifically affects neuronal progenitor cells but not other cell types like iPSCs or glial progenitors .

The precise mechanism by which IMPA1 deficiency leads to intellectual disability also remains incompletely understood. While studies have demonstrated that IMPA1-deficient neural progenitor cells show impaired proliferation and differentiation , how these cellular deficits translate to the complex cognitive phenotypes observed in patients requires further investigation. Similarly, the electrophysiological abnormalities identified in quantitative EEG studies provide clues about altered neural circuit function, but the causal relationship between these changes and cognitive impairment needs clarification.

The relationship between IMPA1, lithium response, and therapeutic outcomes in bipolar disorder continues to be an area of active investigation. While evidence strongly suggests that IMPA1 inhibition contributes to lithium's effects on neuronal excitability and calcium signaling , the complete pathway from enzyme inhibition to mood stabilization remains to be fully elucidated. Additionally, whether genetic variation in IMPA1 contributes to individual differences in lithium response among bipolar patients is an important clinical question that warrants further study.

Finally, the potential for therapeutic targeting of the inositol pathway in IMPA1-associated disorders remains largely unexplored. The observation that embryonic lethality in IMPA1 knockout mice can be reversed with myo-inositol supplementation suggests possibilities for metabolic interventions, but the efficacy and feasibility of such approaches in human patients with IMPA1 mutations have yet to be determined.

How might advances in IMPA1 research translate to clinical applications for neurodevelopmental and psychiatric disorders?

Advances in IMPA1 research hold considerable promise for translational applications across a spectrum of neurodevelopmental and psychiatric disorders. The most immediate potential clinical application lies in the development of targeted therapies for IMPA1-associated intellectual disability (MRT59). Given that embryonic lethality in IMPA1 knockout mice can be reversed with myo-inositol supplementation , there is compelling rationale to investigate whether similar metabolic interventions might ameliorate symptoms in affected humans. Clinical trials of myo-inositol supplementation or related compounds could potentially address the inositol deficiency that appears to underlie the neuronal dysfunction in these patients.

Beyond direct treatment of IMPA1 mutations, this research has broader implications for understanding lithium's therapeutic mechanisms in bipolar disorder. As studies continue to elucidate how IMPA1 inhibition affects neuronal excitability, calcium signaling, and gene expression , this knowledge could inform the development of more targeted mood stabilizers with improved efficacy and reduced side effects compared to lithium. Compounds that selectively modulate specific aspects of the inositol signaling pathway might achieve lithium-like therapeutic benefits while avoiding lithium's narrow therapeutic window and systemic side effects.

The insights gained from studying IMPA1 may also extend to other neurodevelopmental disorders characterized by altered neurogenesis or neuronal excitability. Since IMPA1 deficiency specifically affects neural progenitor cell proliferation and differentiation , understanding these mechanisms could inform therapeutic approaches for a broader range of conditions with impaired neurogenesis.

IMPA1 research additionally offers potential for improved diagnostic and prognostic biomarkers. The quantitative EEG abnormalities identified in IMPA1 mutation carriers could potentially serve as neurophysiological biomarkers for monitoring treatment response or identifying individuals with similar pathophysiology even in the absence of identified IMPA1 mutations.

Product Science Overview

Introduction

Inositol Monophosphatase 1 (IMPA1) is an enzyme encoded by the IMPA1 gene in humans. This enzyme plays a crucial role in the phosphatidylinositol signaling pathway, which is essential for various cellular processes, including cell growth, survival, and differentiation. The recombinant form of this enzyme is produced through genetic engineering techniques, allowing for its use in research and therapeutic applications.

Structure and Function

IMPA1 is a homodimeric enzyme, meaning it consists of two identical subunits. Each subunit has a molecular weight of approximately 30 kDa. The enzyme is primarily located in the cytoplasm and cytosol of cells . It has broad substrate specificity and can act on various inositol monophosphates, including myo-inositol monophosphates, scyllo-inositol-phosphate, and D-galactose 1-phosphate .

The primary function of IMPA1 is to dephosphorylate inositol monophosphates to produce free inositol, which is required for the synthesis of phosphatidylinositol and polyphosphoinositides . These molecules are critical components of cell membranes and play a vital role in signal transduction pathways.

Biological Significance

IMPA1 is involved in several biological processes, including:

  • Inositol metabolic process
  • Inositol biosynthetic process
  • Phosphatidylinositol phosphate biosynthetic process
  • Signal transduction
  • Inositol phosphate dephosphorylation

The enzyme’s activity is regulated by various ions, including magnesium, manganese, and lithium. Lithium, in particular, is known to inhibit IMPA1 activity, which has implications for its use in the treatment of bipolar disorder .

Clinical Relevance

IMPA1 has been implicated as a pharmacological target for lithium action in the brain. Lithium is a well-known treatment for bipolar disorder, and its therapeutic effects are believed to be mediated, at least in part, by the inhibition of IMPA1 . This inhibition leads to an accumulation of inositol monophosphates, which may affect various signaling pathways involved in mood regulation.

Recombinant Production

The recombinant form of IMPA1 is produced using genetic engineering techniques. The gene encoding IMPA1 is cloned into an expression vector, which is then introduced into a host organism, such as Escherichia coli or yeast. The host organism expresses the IMPA1 protein, which can be purified and used for research or therapeutic purposes.

Recombinant IMPA1 is valuable for studying the enzyme’s structure and function, as well as for screening potential inhibitors that may have therapeutic applications. It also allows for the production of large quantities of the enzyme, which is essential for biochemical and pharmacological studies.

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.