PSAP Human

Prosaposin Human Recombinant
Shipped with Ice Packs
In Stock

Description

PSAP Human Recombinant produced in HEK cells is a single, glycosylated, polypeptide chain (a.a 17-524) containing a total of 518 amino acids, having a molecular mass of 57.7kDa (calculated), though it migrates at approximately 65kDa on SDS PAGE, the PSAP is fused to a 2 a.a N-terminal linker, a 2 a.a C-terminal linker and a 6 a.a His tag at C-Terminus.
The Human PSAP is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Prosaposin (PSAP) is a vital protein that serves as a precursor to four lysosomal saposin proteins (A, B, C, and D). These saposins play a crucial role in the breakdown of glycosphingolipids with short oligosaccharide chains. PSAP exhibits neurotrophic properties, promoting neurite outgrowth and enhancing choline acetyltransferase activity. It exists in both secretory and integral membrane protein forms and is found in human milk, cerebrospinal fluid, and seminal plasma, suggesting additional functions. Mutations in the PSAP gene are associated with disorders such as Gaucher disease, Tay-Sachs disease, and metachromatic leukodystrophy.
Description
Recombinant Human PSAP, expressed in HEK cells, is a single, glycosylated polypeptide chain comprising 518 amino acids (a.a 17-524) with a molecular weight of 57.7 kDa (calculated). It appears as a band at approximately 65 kDa on SDS-PAGE due to glycosylation. The protein construct includes a 2 a.a N-terminal linker, a 2 a.a C-terminal linker, and a C-terminal 6 a.a His tag. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile, lyophilized powder with a white color.
Formulation
The protein was sterile filtered (0.4 µm) and lyophilized from a 0.5 mg/ml solution in phosphate buffer, pH 7.4.
Solubility
To prepare a working stock solution, add deionized water to the lyophilized pellet to achieve a concentration of approximately 0.5 mg/ml. Allow for complete dissolution. Prior to use in cell culture, ensure sterility by filtering the solution through an appropriate sterile filter.
Stability
For long-term storage, keep the lyophilized protein at -20°C. After reconstitution, aliquot the protein and store at 4°C to minimize freeze/thaw cycles. Reconstituted PSAP remains stable for up to two weeks at 4°C without noticeable degradation.
Purity
The purity of this protein is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Prosaposin, Proactivator polypeptide, PSAP, GLBA, SAP1.
Source
HEK 293.
Amino Acid Sequence
ASGPVLGLKE CTRGSAVWCQ NVKTASDCGA VKHCLQTVWN KPTVKSLPCD ICKDVVTAAG DMLKDNATEE EILVYLEKTC DWLPKPNMSA SCKEIVDSYL PVILDIIKGE MSRPGEVCSA LNLCESLQKH LAELNHQKQL ESNKIPELDM TEVVAPFMAN IPLLLYPQDG PRSKPQPKDN GDVCQDCIQM VTDIQTAVRT NSTFVQALVE HVKEECDRLG PGMADICKNY ISQYSEIAIQ MMMHMQPKEI CALVGFCDEV KEMPMQTLVP AKVASKNVIP ALELVEPIKK HEVPAKSDVY CEVCEFLVKE VTKLIDNNKT EKEILDAFDK MCSKLPKSLS EECQEVVDTY GSSILSILLE EVSPELVCSM LHLCSGTRLP ALTVHVTQPK DGGFCEVCKK LVGYLDRNLE KNSTKQEILA ALEKGCSFLP DPYQKQCDQF VAEYEPVLIE ILVEVMDPSF VCLKIGACPS AHKPLLGTEK CIWGPSYWCQ NTETAAQCNA VEHCKRHVWN KLHHHHHH.

Q&A

What is Prosaposin (PSAP) and what are its primary functions in the human nervous system?

Prosaposin (PSAP) is a multifunctional protein that serves as the precursor to saposins (A, B, C, and D), which are essential for lysosomal hydrolysis of sphingolipids. Beyond this role, PSAP has demonstrated significant neuroprotective properties in cerebrovascular diseases. Research shows that PSAP can attenuate neuronal apoptosis and promote cell survival by activating specific signaling pathways. The protein appears to exert these effects through interaction with G protein-coupled receptor 37 (GPR37) and subsequently activating the PI3K/Akt/ASK1 pathway, which is critical for neuronal survival under ischemic conditions . This neuroprotective function makes PSAP a promising candidate for research in various neurological disorders, particularly those involving neuronal damage and apoptosis.

How is endogenous PSAP expression regulated following neurological injury?

Following neurological injury, such as ischemic stroke, endogenous PSAP expression undergoes significant temporal regulation. Experimental data from middle cerebral artery occlusion (MCAO) models indicate that both PSAP and its receptor GPR37 show increased expression after injury . This upregulation follows a specific time course, with expression changes observable at 6, 12, 24, and 72 hours post-injury. The temporal pattern suggests that PSAP upregulation represents an endogenous neuroprotective response to injury. Understanding this natural regulatory mechanism provides valuable insights for researchers designing interventions that might enhance or mimic this protective response in clinical settings. Knockdown studies further support PSAP's protective role, as reduction of endogenous PSAP through siRNA techniques has been shown to aggravate neurological deficits following injury .

What experimental methods are most effective for detecting PSAP expression in neural tissues?

For researchers studying PSAP in neural tissues, several complementary techniques have proven effective. Western blot analysis remains the gold standard for quantifying PSAP protein expression and can effectively track temporal changes following experimental interventions. Immunofluorescence staining provides valuable information about the spatial distribution of PSAP and its colocalization with other proteins such as GPR37. This technique is particularly useful for determining whether PSAP is expressed in neurons, glia, or other cell types within the nervous system . For functional studies examining PSAP's effects on neuronal degeneration and apoptosis, Fluoro-Jade C (FJC) staining can identify degenerating neurons, while Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining effectively detects apoptotic cells. These techniques have successfully demonstrated PSAP's protective effects against neuronal apoptosis in experimental models .

How should dose-response studies for PSAP be designed to determine optimal therapeutic concentrations?

Designing effective dose-response studies for PSAP requires careful consideration of multiple dosage levels and appropriate outcome measures. Based on successful experimental approaches, researchers should test at least three concentration levels (e.g., 1μg/kg, 3μg/kg, and 9μg/kg as used in MCAO studies) . Primary outcome measures should include both structural and functional assessments. Structurally, infarct volume measurements (using techniques such as TTC staining) provide quantitative data on tissue preservation. Functionally, both short-term and long-term neurobehavioral assessments are essential, including tests such as modified Garcia scores, beam walking tests, rotarod performance, and cognitive assessments like the Morris water maze . Researchers should also include molecular analyses to determine dose-dependent effects on relevant signaling pathways. Importantly, separate cohorts should be established for different time points (24 hours, 72 hours, and longer-term assessments) to evaluate both immediate and sustained effects of different dosages.

What considerations are important when preparing recombinant PSAP for experimental use?

When preparing recombinant PSAP for experimental applications, researchers must address several critical considerations to ensure experimental validity and reproducibility. Purification protocols must maintain the protein's natural conformation and biological activity. Validation of the recombinant protein should include confirmation of its ability to activate the same downstream signaling pathways as endogenous PSAP, particularly the GPR37/PI3K/Akt/ASK1 pathway . Vehicle selection is also crucial—physiological solutions such as normal saline have been used successfully in experimental models . Additionally, researchers should verify protein stability under experimental storage and delivery conditions. When administering rPSAP intranasally, factors such as volume, concentration, and administration technique must be standardized to ensure consistent delivery across experimental subjects. Finally, researchers should include experiments to confirm the effective delivery of the protein to target tissues, as demonstrated in previous studies using Western blot analysis to verify increased PSAP levels in brain tissue following intranasal administration .

How does PSAP interact with GPR37 to initiate neuroprotective signaling cascades?

PSAP initiates its neuroprotective signaling cascade through interaction with G protein-coupled receptor 37 (GPR37), an orphan receptor expressed in neural tissues. This interaction serves as the initial trigger for downstream neuroprotective pathways. In experimental models, double immunohistochemistry staining has confirmed the colocalization of PSAP and GPR37 in neurons, supporting their functional interaction . Upon binding to GPR37, PSAP activates the PI3K/Akt signaling pathway, which represents a critical survival mechanism in neurons exposed to ischemic conditions. The functional significance of this interaction has been validated through knockdown experiments where GPR37 siRNA administration abolished the anti-apoptotic effects of recombinant PSAP treatment following MCAO . These findings demonstrate that GPR37 is not merely correlated with PSAP function but is mechanistically essential for its neuroprotective effects. Understanding this receptor-ligand interaction provides researchers with a specific target for investigating the initial step in PSAP's neuroprotective signaling cascade.

What is the role of the PI3K/Akt/ASK1 pathway in mediating PSAP's anti-apoptotic effects?

The PI3K/Akt/ASK1 pathway serves as the central mediator of PSAP's anti-apoptotic effects in neurons. Following GPR37 activation by PSAP, this pathway initiates a sequential signaling cascade that ultimately inhibits neuronal apoptosis. Mechanistically, PSAP treatment increases PI3K expression, which subsequently enhances Akt phosphorylation (p-Akt). Activated Akt then phosphorylates Apoptosis Signal-regulating Kinase 1 (ASK1), inhibiting its pro-apoptotic function . This inhibition leads to decreased levels of phosphorylated-JNK (p-JNK), a downstream target of ASK1 that normally promotes apoptosis when activated. The pathway culminates in changes to apoptosis regulators, with increased expression of anti-apoptotic Bcl-2 and decreased levels of pro-apoptotic Bax . The critical nature of this pathway has been confirmed through pharmacological inhibition studies, where the PI3K inhibitor LY294002 abolished PSAP's neuroprotective effects, demonstrating that PI3K activity is essential rather than merely associated with PSAP's function .

How can researchers effectively investigate the temporal dynamics of PSAP-mediated signaling?

Investigating the temporal dynamics of PSAP-mediated signaling requires a carefully designed experimental approach that captures the sequential activation of pathway components. Researchers should establish multiple time points for analysis (e.g., 6h, 12h, 24h, and 72h post-intervention) to track the temporal expression patterns of both PSAP and its downstream effectors . Western blot analysis should be performed to quantify changes in key signaling molecules, including PI3K, phosphorylated-Akt, phosphorylated-ASK1, phosphorylated-JNK, Bcl-2, and Bax, across these time points. Parallel immunohistochemistry studies at each time point can provide spatial information about where these signaling events occur within neural tissues. Researchers should also implement intervention studies at different time points following injury to determine critical windows for therapeutic effectiveness. An effective experimental design would include both gain-of-function approaches (rPSAP administration) and loss-of-function approaches (PSAP siRNA, GPR37 siRNA, or PI3K inhibitors like LY294002) to fully characterize the necessity and sufficiency of each pathway component at different time points .

What controls and experimental groups are essential for rigorous PSAP functional studies?

Rigorous PSAP functional studies require a comprehensive set of control and experimental groups to establish causality and mechanism. At minimum, researchers should include: (1) Sham group (surgical procedure without the experimental injury); (2) Injury+Vehicle group (e.g., MCAO+Normal Saline); (3) Injury+rPSAP treatment group at the optimized dose; (4) Injury+rPSAP+Scramble siRNA (control for siRNA effects); and (5) Injury+rPSAP+Target siRNA (e.g., GPR37 siRNA) to demonstrate pathway specificity . For pharmacological inhibitor studies, additional groups are necessary: (6) Injury+rPSAP+Vehicle (e.g., DMSO) and (7) Injury+rPSAP+Inhibitor (e.g., LY294002) . To validate siRNA or inhibitor effectiveness, separate naive animals should be included to demonstrate knockdown efficiency without the confounding effects of injury. To control for potential sex differences, both male and female subjects should be included, with separate analyses to identify any sexually dimorphic responses to PSAP treatment . This comprehensive approach ensures that observed effects can be specifically attributed to PSAP's activity through the proposed mechanistic pathway.

How should researchers design studies to evaluate both short-term and long-term effects of PSAP?

A comprehensive evaluation of both short-term and long-term effects of PSAP requires a multi-timepoint, multi-outcome experimental design. For short-term assessments, researchers should evaluate outcomes at acute time points (24 and 72 hours post-intervention), focusing on infarct volume measurements, neurological deficit scores (such as modified Garcia and beam walking scores), and molecular analyses of apoptotic markers (using Western blot, FJC, and TUNEL staining) . For long-term assessments, separate experimental cohorts should be maintained for at least 3-4 weeks post-intervention. These long-term studies should include sensorimotor coordination tests (such as rotarod at multiple speeds) performed at weekly intervals (days 7, 14, and 21) to track recovery progression . Cognitive function should be evaluated using tests such as the Morris water maze, with assessments of both learning (escape latency and swim distance over multiple trial blocks) and memory (probe trial performance) . This comprehensive temporal assessment allows researchers to determine whether PSAP produces sustained benefits beyond the acute phase and affects both motor and cognitive domains of recovery.

What evidence supports the potential therapeutic application of PSAP in human ischemic stroke?

Several lines of evidence support PSAP's potential therapeutic application in human ischemic stroke. Preclinical studies have demonstrated that intranasal administration of recombinant PSAP significantly reduces brain infarction and neuronal apoptosis while improving both short-term and long-term neurological function in experimental stroke models . The protein's mechanism of action through the GPR37/PI3K/Akt/ASK1 pathway represents a well-characterized neuroprotective mechanism that is conserved across mammalian species . PSAP's ability to attenuate multiple aspects of ischemic injury—including reduction of neuronal apoptosis, decrease in neurodegeneration (as evidenced by FJC staining), and improvement in functional outcomes—suggests potential for comprehensive therapeutic benefits . The effectiveness of intranasal delivery provides a clinically feasible administration route that could be implemented in acute stroke settings. Additionally, the fact that PSAP treatment remains effective when administered after the onset of injury (1 hour after reperfusion in experimental models) aligns with the clinical reality of stroke treatment timelines . These findings collectively support further exploration of PSAP as a potential therapeutic agent for human ischemic stroke.

How might sex-specific differences influence PSAP efficacy in clinical applications?

Sex-specific differences may significantly influence PSAP efficacy in clinical applications, necessitating careful consideration in both preclinical and clinical research designs. Experimental studies have included both male and female subjects, specifically investigating whether PSAP treatment produces sexually dimorphic outcomes in neurological recovery following ischemic injury . This approach acknowledges the growing recognition that stroke pathophysiology and treatment response may differ between males and females due to hormonal, genetic, and physiological factors. When designing clinical trials, researchers should ensure adequate representation of both male and female participants and plan for sex-stratified analyses of efficacy outcomes. Potential mechanisms for sex-specific differences might include variations in PSAP receptor expression, differences in downstream signaling pathway activity, or interactions with sex hormones that could enhance or diminish PSAP's neuroprotective effects. Understanding these sex-specific factors will be crucial for optimizing PSAP-based therapies and potentially tailoring dosing strategies for male and female patients to maximize therapeutic benefits while minimizing adverse effects.

What potential challenges might arise in translating PSAP therapy from animal models to human clinical trials?

Translating PSAP therapy from animal models to human clinical trials faces several significant challenges that researchers must address. Species differences in PSAP signaling and receptor distribution may affect therapeutic efficacy, requiring careful validation of pathway conservation between experimental models and humans. Pharmacokinetic considerations present another challenge, as optimal dosing, timing, and administration routes established in animal models may require substantial adjustment for human applications . The heterogeneity of human stroke presents additional complexity—unlike standardized experimental models like MCAO, human strokes vary widely in location, severity, mechanism, and comorbidities, potentially affecting PSAP's therapeutic efficacy across different patient populations . The therapeutic window for intervention remains uncertain in humans and will require careful investigation, as the 1-hour post-reperfusion timepoint used in animal studies may not directly translate to clinical scenarios where treatment delays are common . Finally, researchers must develop appropriate biomarkers to monitor PSAP activity and treatment response in human subjects, which might include neuroimaging techniques, blood-based biomarkers, or functional assessments that differ from those used in experimental models. Addressing these translational challenges will be essential for successful development of PSAP-based therapies for human neurological conditions.

What are the long-term consequences of PSAP administration on neurogenesis and neural circuit remodeling?

Understanding PSAP's effects on neurogenesis and neural circuit remodeling represents a critical frontier in advanced PSAP research. While current studies have demonstrated PSAP's ability to improve long-term functional outcomes (up to 27 days post-injury in experimental models) , the cellular and circuit-level mechanisms underlying these sustained benefits remain incompletely characterized. Future research should investigate whether PSAP treatment influences adult neurogenesis in regions such as the subventricular zone and hippocampus following injury. Studies should examine whether PSAP affects neural progenitor proliferation, migration, differentiation, or survival, potentially contributing to replacement of lost neurons. Additionally, researchers should explore PSAP's effects on axonal sprouting, dendritic remodeling, and synaptogenesis using techniques such as targeted tract tracing, high-resolution imaging, and electrophysiological recordings. Understanding whether PSAP modulates neuroplasticity-related genes and proteins would provide mechanistic insights into these potential effects. These investigations would determine whether PSAP's benefits extend beyond acute neuroprotection to include enhancement of endogenous repair mechanisms, which could significantly impact its therapeutic potential for promoting long-term recovery from neurological injuries.

How might genetic variations in PSAP or its receptors influence individual responsiveness to PSAP-based therapies?

The influence of genetic variations on PSAP efficacy represents an important consideration for personalized medicine approaches to PSAP-based therapies. Future research should investigate how polymorphisms in the PSAP gene itself might affect protein structure, stability, or receptor binding, potentially altering therapeutic efficacy. Similarly, genetic variations in GPR37 or other PSAP receptors could influence receptor expression, binding affinity, or downstream signaling efficiency . Research approaches should include: (1) genome-wide association studies correlating genetic variants with outcomes in PSAP treatment cohorts; (2) in vitro studies examining how specific variants affect PSAP-receptor interactions and signaling pathway activation; and (3) development of personalized medicine approaches that might tailor PSAP treatment strategies based on individual genetic profiles. Additionally, epigenetic modifications affecting PSAP or receptor expression should be investigated as potential markers of treatment responsiveness. This pharmacogenomic approach would help identify which patient populations might benefit most from PSAP-based therapies and allow for personalized dosing strategies that maximize therapeutic benefits while minimizing potential adverse effects in genetically diverse patient populations.

Research AreaKey MethodsPrimary OutcomesChallenges
PSAP Expression AnalysisWestern blot, ImmunofluorescenceProtein levels, Cellular localizationAntibody specificity, Sample preparation
Functional AssessmentModified Garcia score, Beam walking, Rotarod, Morris water mazeSensorimotor function, Cognitive performanceStandardization, Experimenter bias
Pathway AnalysissiRNA knockdown, Pharmacological inhibitors, Phospho-protein detectionSignaling molecule activation, Pathway necessityOff-target effects, Temporal dynamics
Apoptosis EvaluationTUNEL staining, FJC staining, Bcl-2/Bax ratioNeuronal death, NeuroprotectionTissue processing, Quantification methods
Translational StudiesIntranasal delivery, Dose optimization, Sex-comparisonTherapeutic efficacy, Safety profileSpecies differences, Clinical relevance

Product Science Overview

Structure and Function

Prosaposin is composed of 524 amino acids and has a molecular mass of approximately 57.9 kDa . The protein is synthesized as a single polypeptide chain and undergoes post-translational modifications, including glycosylation. The four saposins derived from prosaposin each have distinct roles in lipid metabolism:

  • Saposin A and C: These stimulate the hydrolysis of glucosylceramide and galactosylceramide by their respective enzymes .
  • Saposin B: This facilitates the hydrolysis of galactocerebroside sulfate, GM1 gangliosides, and globotriaosylceramide .
  • Saposin D: This acts as a specific activator of sphingomyelin phosphodiesterase .

Prosaposin itself functions as a myelinotrophic and neurotrophic factor, mediating its effects through G-protein-coupled receptors, GPR37 and GPR37L1 .

Genetic and Clinical Significance

Mutations in the PSAP gene can lead to various lysosomal storage disorders. For instance, mutations affecting the saposin B domain cause a rare variant of metachromatic leukodystrophy (MLD), where arylsulfatase A activity remains normal . Additionally, variants in the saposin D domain have been linked to Parkinson’s disease .

Recombinant Prosaposin

Recombinant human prosaposin is produced using DNA sequences encoding the human PSAP gene, typically expressed in HEK293 cells . The recombinant protein is often tagged with polyhistidine for purification purposes and is verified for purity and activity through methods such as SDS-PAGE and HPLC .

Recombinant prosaposin is used in research to study its biological functions and potential therapeutic applications, particularly in the context of neurodegenerative diseases and lysosomal storage disorders.

Storage and Stability

Recombinant prosaposin is usually provided as a lyophilized powder and should be stored under sterile conditions at -20°C to -80°C. It is recommended to avoid repeated freeze-thaw cycles to maintain its stability and activity .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.