LYPLAL1 Human

Lysophospholipase Like I Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to LYPLAL1 Human

LYsophospholipase-like 1 (LYPLAL1) is a human protein encoded by the LYPLAL1 gene located on chromosome 1 (1q32.1). Structurally classified as an α/β-hydrolase, it shares homology with acyl protein thioesterases but exhibits distinct substrate preferences and enzymatic activity . While initially hypothesized to function as a phospholipase or triglyceride lipase, biochemical studies confirm its role as a serine hydrolase with specificity for short-chain acyl substrates and protein depalmitoylation .

Enzymatic Activity

  • Depalmitoylation: Catalyzes removal of palmitate groups from proteins such as cGAS (cyclic GMP-AMP synthase) and KCNMA1 (calcium-activated potassium channels) .

  • No Lipase Activity: Unable to hydrolyze long-chain triglycerides or phospholipids due to steric constraints in its active site .

Metabolic Regulation

  • Adipogenesis: Genome-wide association studies (GWAS) link LYPLAL1 variants to sex-specific fat distribution and waist-to-hip ratio in humans .

    • Female Lyplal1 knockout (KO) mice show reduced adiposity, smaller adipocytes, and protection from high-fat diet-induced obesity .

    • Associated long noncoding RNA LYPLAL1-AS1 promotes adipogenic differentiation in human mesenchymal stem cells .

  • Hepatic Metabolism: Inhibition of LYPLAL1 in hepatocytes increases glucose production, suggesting a role in gluconeogenesis .

Immune Modulation

  • cGAS Regulation: Depalmitoylates cGAS, inhibiting its dimerization and downstream innate immune responses .

Sex-Specific Effects

  • Murine studies reveal female-specific phenotypes:

    • Reduced liver triglycerides and steatosis in HFHS-fed KO mice .

    • Elevated serum triglycerides despite lower adiposity .

Disease Associations

ConditionAssociationSource
ObesityGWAS links to fat distribution (female-specific)
Type 2 DiabetesLYPLAL1-DT lncRNA regulates endothelial cell autophagy and inflammation
Nonalcoholic Fatty Liver Disease (NAFLD)Reduced liver triglycerides in Lyplal1 KO mice

Therapeutic Potential

  • Inhibitors: Covalent inhibitors (e.g., compounds from Pfizer) reduce LYPLAL1 activity, enhancing cGAS-mediated immunity and glucose production .

  • Gene Therapy: Targeting LYPLAL1-AS1 or LYPLAL1-DT could modulate adipogenesis or endothelial dysfunction .

Recombinant Protein

  • Expression: Recombinant human LYPLAL1 (1–237 aa) is produced in E. coli with >95% purity, used for enzymatic assays and structural studies .

Animal Models

  • Mouse KO: CRISPR-Cas9-generated Lyplal1 KO mice show sex- and diet-dependent metabolic phenotypes .

    • Key Findings:

      • Females: Reduced body fat, adipocyte size, and liver steatosis on high-fat diets .

      • Males: No significant changes in adiposity .

Controversies and Challenges

  • Functional Redundancy: Some studies report no metabolic abnormalities in Lyplal1 KO mice under standard diets, suggesting compensatory mechanisms .

  • Mechanistic Uncertainty: The exact pathways linking LYPLAL1 to obesity and immune regulation remain unresolved .

Product Specs

Introduction
Lysophospholipase-like protein 1 (LYPLAL1) is a member of the AB hydrolase 2 family. It does not exhibit phospholipase or triacylglycerol lipase activity and can only hydrolyze short-chain substrates due to its shallow active site.
Description
Recombinant human LYPLAL1, expressed in E. coli, is a single polypeptide chain with a molecular weight of 28.9 kDa. It comprises 261 amino acids, including a 24 amino acid His-tag at the N-terminus (amino acids 1-237). Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The LYPLAL1 solution is provided at a concentration of 0.25 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For longer storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
Purity is determined to be greater than 95% by SDS-PAGE analysis.
Synonyms
Lysophospholipase-like protein 1, LYPLAL1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMAAASG SVLQRCIVSP AGRHSASLIF LHGSGDSGQG LRMWIKQVLN QDLTFQHIKI IYPTAPPRSY TPMKGGISNV WFDRFKITND CPEHLESIDV MCQVLTDLID EEVKSGIKKN RILIGGFSMG GCMAMHLAYR NHQDVAGVFA LSSFLNKASA VYQALQKSNG VLPELFQCHG TADELVLHSW AEETNSMLKS LGVTTKFHSF PNVYHELSKT ELDILKLWIL TKLPGEMEKQ K.

Q&A

What is LYPLAL1 and what is its molecular structure?

LYPLAL1 (Lysophospholipase-like protein 1) is a protein encoded by the LYPLAL1 gene in humans. Structurally, it belongs to the α/β-hydrolase family of proteins . The protein has been categorized in several databases including Pfam (PF02230), InterPro (IPR029058), and CATH (3u0v) . Its three-dimensional structure reveals critical features that distinguish it from related proteins, particularly in the substrate binding region which influences its enzymatic specificity and function .

How does LYPLAL1 differ from acyl-protein thioesterases?

Despite sharing sequence conservation and structural homology with acyl-protein thioesterases (APTs), LYPLAL1 exhibits a critical structural difference: its hydrophobic substrate binding tunnel is closed due to a different loop conformation, whereas this tunnel is open in human acyl-protein thioesterases 1 and 2, as well as in Zea mays acyl-protein thioesterase 2 . This structural difference fundamentally alters the enzyme's substrate specificity, restricting it to short acyl chains rather than the longer chains preferred by APTs . Notably, while APTs can depalmitoylate the oncogene Ras, LYPLAL1 has been demonstrated to be incapable of performing this function . These structural and functional differences distinguish LYPLAL1 from canonical acyl-protein thioesterases despite their evolutionary relationship.

What is LYPLAL1-AS1 and how does it relate to LYPLAL1?

LYPLAL1-AS1 (LYPLAL1-antisense RNA1) is a long non-coding RNA that is dramatically upregulated during adipogenic differentiation of human adipose-derived mesenchymal stem cells (hAMSCs) . The full-length LYPLAL1-AS1 is 523 nucleotides as determined by 5′ and 3′ rapid amplification of cDNA ends assays . As an antisense RNA, it is transcribed from the opposite strand of the DNA encoding the LYPLAL1 gene. While LYPLAL1 itself is associated with fat distribution and metabolic traits, LYPLAL1-AS1 plays a regulatory role in adipogenic differentiation by targeting desmoplakin (DSP) and inhibiting the Wnt/β-catenin pathway . Cell fractionation studies have demonstrated that LYPLAL1-AS1 is predominantly located in the cytoplasm (>70%), consistent with its role in post-transcriptional regulation .

What are the key structural features of LYPLAL1?

As a member of the α/β-hydrolase family, LYPLAL1 possesses the characteristic fold consisting of a central β-sheet surrounded by α-helices . The most distinguishing structural feature of LYPLAL1 is its substrate binding region, particularly the hydrophobic substrate binding tunnel. Unlike related enzymes such as acyl-protein thioesterases, this tunnel is closed in LYPLAL1 due to a different loop conformation . This structural difference is functionally significant as it restricts LYPLAL1's substrate specificity to short acyl chains . The active site architecture of LYPLAL1 is consistent with its function as an esterase rather than a lipase, with a preference for acetyl groups as substrates .

What are the best methods to study LYPLAL1 function in adipose tissue?

Studying LYPLAL1 function in adipose tissue requires a multi-faceted approach:

  • Genetic Manipulation Models: CRISPR-Cas9 technology has proven effective for creating LYPLAL1 knockout models in mice, as demonstrated by studies where a single base pair deletion was introduced in exon 1 of the murine Lyplal1 gene . These models allow for in vivo assessment of LYPLAL1's role in adipose tissue development and function.

  • Diet Challenges: As LYPLAL1's effects appear to be diet-responsive, utilizing appropriate dietary challenges is crucial. Studies have shown that high-fat, high-sucrose (HFHS) diets better reveal the phenotypic effects of LYPLAL1 knockout compared to standard high-fat diets alone . Experimental protocols typically involve 23-week feeding periods to allow adequate time for phenotype development .

  • Tissue-Specific Analyses: Comprehensive assessment should include measurement of whole-body fat percentage, analysis of specific fat depots (visceral vs. subcutaneous), histological examination of adipocyte size and morphology, triglyceride quantification in adipose tissue and liver, and liver histology for steatosis assessment .

  • Sex-Specific Analysis: Given the known sex-specific effects of LYPLAL1, experiments should always include both male and female subjects with sex-stratified analyses .

How can LYPLAL1 knockout models be generated and validated?

Generation and validation of LYPLAL1 knockout models involves several key steps:

  • CRISPR-Cas9 Gene Editing: Design guide RNAs targeting the first coding exon of LYPLAL1 and introduce a frameshift mutation (e.g., a single base pair deletion) to cause nonsense-mediated mRNA decay .

  • Validation of Knockout Efficiency: Confirm absence of LYPLAL1 mRNA using quantitative RT-PCR and verify complete protein knockout using Western blot analysis with specific antibodies against LYPLAL1 . Include appropriate tissue controls where LYPLAL1 is known to be expressed (e.g., kidney tissue) .

  • Breeding Strategy: Establish heterozygous breeding pairs to generate wild-type, heterozygous, and homozygous knockout littermates. This approach allows for direct comparison between genotypes while controlling for genetic background.

  • Phenotypic Characterization: Compare baseline characteristics between genotypes and subject animals to appropriate challenges (e.g., high-fat, high-sucrose diet) to reveal phenotypes . Include both sexes in all analyses to detect sex-specific effects.

  • Controls and Experimental Design: Include appropriate diet controls (e.g., regular chow vs. HFHS diet), use littermate controls whenever possible, and plan for sufficient sample sizes to detect sex-specific differences .

What techniques are most effective for measuring LYPLAL1 enzymatic activity?

Measuring LYPLAL1 enzymatic activity presents challenges due to its somewhat ambiguous substrate specificity. Based on the current understanding of LYPLAL1 as an esterase with preference for short-chain substrates, several techniques can be employed:

  • Recombinant Protein Expression and Purification: Express LYPLAL1 in bacterial or mammalian expression systems and purify using affinity chromatography (e.g., His-tag purification).

  • Esterase Activity Assays: Use fluorogenic or chromogenic ester substrates (p-nitrophenyl acetate is appropriate for short-chain esterase activity) and monitor substrate hydrolysis spectrophotometrically.

  • Substrate Specificity Profiling: Test activity against a panel of substrates with varying acyl chain lengths to compare with acyl-protein thioesterases and highlight functional differences .

  • Cellular Deacetylation Assays: Given that structural studies suggest LYPLAL1 might function as a protein deacetylase, techniques to specifically assess this activity should be considered, though experimental validation is still needed .

How can the subcellular localization of LYPLAL1 be determined?

Determining the subcellular localization of LYPLAL1 is essential for understanding its biological function. Several complementary techniques can be employed:

  • Cell Fractionation and Western Blotting: Separate cellular components (nucleus, cytoplasm, membrane fractions) and perform Western blot analysis using LYPLAL1-specific antibodies. Include markers for different cellular compartments as controls.

  • Immunofluorescence Microscopy: Fix and permeabilize cells, stain with specific antibodies against LYPLAL1, and counterstain with markers for cellular compartments. Analyze using confocal microscopy for precise localization.

  • Fluorescent Protein Tagging: Generate GFP or other fluorescent protein fusions with LYPLAL1, express in relevant cell types, and visualize in live cells to avoid fixation artifacts.

For LYPLAL1-AS1, cell fractionation followed by quantitative RT-PCR has been successfully used to demonstrate its predominantly cytoplasmic distribution (>70%) . RNA fluorescent in situ hybridization (FISH) assays have also confirmed this cytoplasmic localization, with appropriate controls such as 18S rRNA and U6 .

What are the methodological challenges in studying LYPLAL1-AS1?

Studying LYPLAL1-AS1, like other long non-coding RNAs, presents several methodological challenges:

  • RNA Stability and Handling: Long non-coding RNAs often have lower expression levels than mRNAs, making RNA degradation during isolation a significant concern.

  • Full-Length Characterization: Determining the complete sequence requires specialized techniques such as 5′ and 3′ rapid amplification of cDNA ends (RACE) assays, which were essential for defining the exact transcript boundaries of LYPLAL1-AS1 (523 nt) .

  • Functional Analysis: Knockdown approaches using siRNA or shRNA may have off-target effects, while CRISPR-based approaches for lncRNA knockout need to avoid disrupting the overlapping LYPLAL1 gene. Lentiviral overexpression systems have been successfully used for LYPLAL1-AS1 functional studies .

  • Localization Studies: RNA fluorescent in situ hybridization (FISH) requires carefully designed probes, while subcellular fractionation followed by qRT-PCR should include appropriate controls (e.g., GAPDH for cytoplasm, U1 for nucleus) .

  • Target Identification: Identifying molecular targets of LYPLAL1-AS1 requires specialized techniques such as RNA pulldown assays to identify protein binding partners.

What explains the sex-specific effects of LYPLAL1 on fat distribution?

The sex-specific effects of LYPLAL1 on fat distribution represent one of the most intriguing aspects of this gene's biology. Multiple genome-wide association studies have found that variants near LYPLAL1 have stronger associations with fat distribution metrics in females than males, including visceral/subcutaneous adipose tissue ratio (VAT/SAT) , waist-to-hip ratio (WHR) , and WHR adjusted for BMI . Several mechanisms may explain these sex-specific effects:

  • Experimental Evidence from Mouse Models: Lyplal1 knockout (KO) female mice on high-fat, high-sucrose (HFHS) diet showed significant protection against weight gain and fat accumulation compared to wild-type mice, an effect not observed in male Lyplal1 KO mice . Female KO mice specifically demonstrated reduced body fat percentage, white fat mass, and adipocyte diameter without changes in metabolic rate .

  • Sex-Specific Metabolic Effects: Female, but not male, Lyplal1 KO mice exhibited increased serum triglycerides and decreased liver enzymes (aspartate and alanine aminotransferases) , suggesting sex-specific roles in lipid metabolism and liver function.

  • Gene-by-Sex-by-Diet Interaction: The phenotypic differences become apparent only under specific dietary conditions (HFHS diet), indicating a complex interaction where LYPLAL1's role in metabolism is context-dependent and influenced by both sex and environmental factors .

Further research using tissue-specific knockout models and mechanistic studies focusing on hormonal regulation may help elucidate the molecular basis of these sex-specific effects.

How does LYPLAL1 interact with the insulin signaling pathway?

The relationship between LYPLAL1 and insulin signaling is supported by multiple genome-wide association studies that have linked variants near LYPLAL1 to insulin-related traits:

  • Genetic Association Studies: Variants near LYPLAL1 have been associated with insulin clearance and insulin resistance , suggesting LYPLAL1 may influence insulin metabolism or signaling.

  • Potential Molecular Mechanisms: As a potential acyl protein thioesterase, LYPLAL1 might regulate the acylation status of proteins involved in insulin signaling. Protein acylation (particularly palmitoylation) affects membrane localization and function of many signaling proteins.

  • Adiponectin Regulation: Variants near LYPLAL1 have been associated with levels of adiponectin , an adipose-derived hormone that improves insulin sensitivity, suggesting LYPLAL1 may indirectly influence insulin signaling through effects on adipokine production.

The precise molecular mechanisms linking LYPLAL1 to insulin signaling remain to be elucidated. Future research should focus on determining if LYPLAL1 directly deacylates components of the insulin signaling pathway and investigating tissue-specific effects of LYPLAL1 on insulin sensitivity.

What is the relationship between LYPLAL1 and non-alcoholic fatty liver disease?

The relationship between LYPLAL1 and non-alcoholic fatty liver disease (NAFLD) is supported by both human genetic studies and experimental animal models:

  • Genetic Association Studies: Genome-wide association studies have identified variants near LYPLAL1 that associate with NAFLD , suggesting LYPLAL1 may play a role in hepatic lipid metabolism or liver disease pathogenesis.

  • Experimental Evidence from Mouse Models: Lyplal1 knockout (KO) mice of both sexes showed reduced liver triglycerides and decreased hepatic steatosis when fed a high-fat, high-sucrose (HFHS) diet . This protective effect against liver fat accumulation occurred in both male and female KO mice, unlike the sex-specific effects on body weight and adiposity. Female, but not male, KO mice exhibited decreased serum levels of liver enzymes , suggesting reduced liver injury.

  • Clinical Implications: The protective effect of Lyplal1 knockout against liver steatosis suggests that LYPLAL1 inhibition might be a therapeutic strategy for NAFLD, with potentially sex-specific benefits for liver health.

Future studies should focus on liver-specific knockout models and detailed analyses of hepatic lipid metabolism pathways to better understand LYPLAL1's role in NAFLD pathogenesis.

How does LYPLAL1-AS1 regulate adipogenic differentiation at the molecular level?

LYPLAL1-AS1, a long non-coding RNA, regulates adipogenic differentiation through a specific molecular mechanism that has been characterized in human adipose-derived mesenchymal stem cells (hAMSCs):

  • Expression Pattern: LYPLAL1-AS1 is dramatically upregulated during adipogenic differentiation of hAMSCs , suggesting a role in promoting the adipogenic program.

  • Functional Effects: Knockdown of LYPLAL1-AS1 decreases adipogenic differentiation of hAMSCs, while overexpression enhances it . These gain- and loss-of-function experiments demonstrate that LYPLAL1-AS1 is both necessary and sufficient to promote adipogenesis.

  • Molecular Target: Desmoplakin (DSP) has been identified as a direct target of LYPLAL1-AS1 . Knockdown of DSP enhances adipogenic differentiation and rescues the adipogenic differentiation defect caused by LYPLAL1-AS1 depletion .

  • Mechanism of Action: LYPLAL1-AS1 modulates DSP protein stability, possibly via proteasome degradation . This post-translational regulation inhibits the Wnt/β-catenin pathway during adipogenic differentiation . Since Wnt/β-catenin signaling is a known negative regulator of adipogenesis, LYPLAL1-AS1 promotes adipogenic differentiation by suppressing this inhibitory pathway.

This molecular mechanism represents a novel regulatory axis in adipogenic differentiation and suggests LYPLAL1-AS1 could be a therapeutic target for disorders related to abnormal adipogenesis .

What contradictions exist in the literature regarding LYPLAL1's enzymatic activity?

The enzymatic activity of LYPLAL1 has been a subject of contradictory findings in the scientific literature:

These contradictions highlight the need for further biochemical and structural studies to definitively characterize LYPLAL1's enzymatic activity and biological function.

How do LYPLAL1 genetic variants contribute to obesity and metabolic syndrome?

LYPLAL1 genetic variants have been consistently associated with obesity-related traits and metabolic syndrome components through genome-wide association studies:

  • Associations with Body Fat Distribution: Single nucleotide polymorphisms (SNPs) near LYPLAL1 have been repeatedly associated with waist-to-hip ratio (WHR) and visceral/subcutaneous adipose tissue ratio (VAT/SAT) . These associations indicate LYPLAL1 variants influence how fat is distributed in the body, with preferential association with central adiposity, which is more strongly linked to metabolic disease risk.

  • Sex-Specific Effects: The associations between LYPLAL1 variants and fat distribution metrics are more significant in females than males , including stronger associations with VAT/SAT ratio , WHR , and WHR adjusted for BMI .

  • Metabolic Trait Associations: LYPLAL1 variants have been linked to insulin clearance , insulin resistance , fasting serum triglyceride levels , and adiponectin levels .

  • Liver-Related Phenotypes: Variants near LYPLAL1 associate with non-alcoholic fatty liver disease (NAFLD) , consistent with the reduced liver triglycerides and steatosis observed in Lyplal1 knockout mice .

  • Experimental Evidence Supporting Causality: Mouse models with Lyplal1 knockout demonstrate phenotypes that parallel human genetic associations, suggesting the genetic associations may be causal rather than merely correlative .

While these associations and experimental findings strongly implicate LYPLAL1 in obesity and metabolic syndrome pathophysiology, the precise molecular mechanisms remain to be fully elucidated.

Could LYPLAL1 or LYPLAL1-AS1 serve as therapeutic targets for obesity?

Both LYPLAL1 and LYPLAL1-AS1 show potential as therapeutic targets for obesity, supported by experimental evidence:

  • LYPLAL1 as a Therapeutic Target:

    • Female Lyplal1 knockout mice show protection against diet-induced obesity with reduced body fat percentage, white fat mass, and adipocyte diameter

    • Both male and female knockout mice display reduced liver triglycerides and hepatic steatosis

    • Potential therapeutic strategies include small molecule inhibitors targeting LYPLAL1's enzymatic activity or antisense approaches to reduce expression

    • Sex-specific effects suggest therapies might be more effective in females

  • LYPLAL1-AS1 as a Therapeutic Target:

    • Knockdown of LYPLAL1-AS1 decreases adipogenic differentiation of human mesenchymal stem cells

    • LYPLAL1-AS1 has been explicitly suggested as a novel therapeutic target for preventing and combating diseases related to abnormal adipogenesis

    • Potential approaches include antisense oligonucleotides targeting LYPLAL1-AS1 or molecules that disrupt LYPLAL1-AS1/DSP interaction

Further research is needed to fully evaluate the therapeutic potential of these targets, including development of specific, effective targeting strategies and assessment of potential off-target effects.

What is the significance of LYPLAL1 in personalized medicine approaches to metabolic diseases?

LYPLAL1 holds significant potential for personalized medicine approaches to metabolic diseases:

  • Sex-Specific Genetic Associations: Variants near LYPLAL1 show stronger associations with metabolic traits in females than males , suggesting genetic testing might help predict which female patients are at higher risk for central obesity and related complications.

  • Gene-by-Diet Interactions: Studies in mouse models reveal that Lyplal1 knockout effects become apparent primarily under high-fat, high-sucrose dietary conditions . This gene-by-diet interaction suggests LYPLAL1 genotyping could help identify individuals who would particularly benefit from specific dietary interventions.

  • Stratification for Targeted Therapies: The complex gene-by-sex-by-diet interactions observed with LYPLAL1 provide a basis for stratifying patients for personalized interventions. Female patients with specific LYPLAL1 variants might benefit most from LYPLAL1-targeted therapies.

  • Potential Biomarker Applications: LYPLAL1 genetic variants could serve as biomarkers for predicting risk of central obesity, identifying individuals susceptible to diet-induced metabolic disease, and assessing risk for non-alcoholic fatty liver disease progression.

The multifaceted role of LYPLAL1 in metabolism, combined with its sex-specific and diet-dependent effects, makes it a promising candidate for inclusion in personalized medicine approaches to obesity and metabolic diseases.

How do LYPLAL1-related phenotypes differ between sexes in human populations?

LYPLAL1-related phenotypes show consistent and significant differences between sexes in human populations:

  • Fat Distribution Phenotypes: Variants near LYPLAL1 show stronger associations with waist-to-hip ratio (WHR) in females compared to males . The association with visceral/subcutaneous adipose tissue ratio (VAT/SAT) is more significant in females , and WHR adjusted for BMI shows sex-specific effects favoring stronger associations in females .

  • Strength and Consistency of Sex Differences: The sex-specific effects have been replicated across multiple independent studies by different research groups and persist across different ethnic populations .

  • Parallel Findings in Animal Models: Female Lyplal1 knockout mice show protection against diet-induced obesity, with effects not observed in male knockout mice . Female knockout mice also show altered serum profiles (increased triglycerides, decreased liver enzymes) not seen in males . These animal model findings remarkably parallel the human genetic association data.

Understanding these sex-specific effects is crucial for translating LYPLAL1 research into clinical applications, as interventions targeting LYPLAL1 may need to be tailored differently for male and female patients.

Is there a correlation between LYPLAL1 expression and response to diet interventions?

The relationship between LYPLAL1 expression and response to dietary interventions represents an emerging area with important implications:

  • Evidence from Animal Models: Lyplal1 knockout mice show phenotypic differences compared to wild-type mice primarily when challenged with a high-fat, high-sucrose (HFHS) diet . Previous studies using only high-fat diet without high sucrose failed to reveal phenotypes in Lyplal1 knockout mice . Female Lyplal1 knockout mice were protected against HFHS diet-induced obesity, indicating that LYPLAL1 expression may influence the response to obesogenic diets in a sex-specific manner .

  • Diet-Responsive Expression: Evidence suggests that Lyplal1 mRNA expression is at least partly diet-responsive , indicating that dietary composition may directly influence LYPLAL1 expression levels.

  • Gene-by-Diet Interaction: The phenotypic effects of Lyplal1 knockout becoming apparent only under specific dietary conditions represents a clear gene-by-diet interaction , suggesting that individuals with different LYPLAL1 variants or expression levels might respond differently to various dietary interventions.

The evidence for LYPLAL1 as a diet-responsive gene that influences metabolic outcomes in a sex-specific manner suggests it could be an important factor in personalized nutrition approaches for obesity and metabolic disease management.

Product Science Overview

Gene and Protein Structure

The LYPLAL1 gene is located on chromosome 1q41 . The protein shares structural similarities with acyl protein thioesterases (APTs), featuring a typical alpha/beta hydrolase fold and a classical catalytic triad of serine, aspartate, and histidine . However, unlike other APT family members, LYPLAL1’s hydrophobic tunnel is closed, and it functions as a monomer rather than a dimer .

Biochemical Features

Using an in vitro assay of recombinant human LYPLAL1 expressed in Escherichia coli, researchers found that LYPLAL1 showed no phospholipase activity. Instead, it hydrolyzed short-chain substrates such as 4-nitrophenyl acetate . The crystal structure of human LYPLAL1 at 1.72-angstrom resolution revealed its functional plasticity despite its close relationship to acyl protein thioesterases .

Functional Insights

LYPLAL1’s primary function is to hydrolyze short-chain substrates, which distinguishes it from other members of the AB hydrolase 2 family . This unique functionality is attributed to its shallow active site, which limits its activity to short-chain substrates .

Research and Applications

The study of LYPLAL1 has provided valuable insights into the functional diversity of the AB hydrolase 2 family. Understanding its structure and function can aid in the development of targeted therapies and biotechnological applications. For instance, the recombinant expression of LYPLAL1 in E. coli has facilitated detailed biochemical studies and potential industrial applications .

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.