LCAT Human

Lecithin-Cholesterol Acyltransferase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Physiological Roles and Clinical Implications

LCAT’s primary functions include esterifying free cholesterol on HDL, enabling cholesterol transport from peripheral tissues to the liver. Deficiencies or mutations in LCAT lead to:

ConditionMechanismClinical Manifestations
HypoalphalipoproteinemiaLoss-of-function mutations (e.g., R244G, R244H) reduce HDL-cholesterol Low HDL-C, corneal opacity, renal disease, atherosclerosis
Familial LCAT DeficiencyComplete inactivation of LCATSevere HDL deficiency, anemia, renal failure, and premature cardiovascular disease

Reverse Cholesterol Transport (RCT):
LCAT converts free cholesterol to cholesteryl esters (CE), which are sequestered into HDL’s core. Mature HDL then transfers CE to LDL/VLDL via CETP or directly to the liver via SR-BI .

Recombinant Human LCAT (rhLCAT)

Therapeutic AgentDevelopment StageKey Findings
ACP-501Phase II (NCT02601560)Rapid HDL-C increase (40–50%) in CHD patients; safe in Phase I trials
MEDI6012Clinical trialsReverses HDL dysfunction in acute coronary syndrome (ACS); enhances RCT

Mechanism of Action:

  • ACP-501: Increases CE content in HDL, promoting cholesterol efflux from macrophages .

  • MEDI6012: Restores HDL functionality in patients with low LCAT activity .

DS-8190a (Daiichi Sankyo)

ParameterHuman PlasmaCynomolgus Monkey PlasmaMouse Plasma
EC₅₀0.016 μM0.012 μM>5 μM (inhibitor)
EffectDose-dependent activationActivationInhibition

Binding Site:

  • Interacts with residues in Subdomain 2 (e.g., Tyr51, Gly71) .

  • Competes with inactive analogs (e.g., ent-DS-8190a), confirming specificity .

In Vivo Efficacy:

  • Reduces atherosclerosis in Ldlr−/−× hu Lcat+/− mice .

  • Enhances RCT in nonhuman primates by lowering pre-β1-HDL levels .

Challenges and Future Directions

  1. Species-Specific Effects:

    • DS-8190a activates LCAT in humans/monkeys but inhibits it in mice, complicating preclinical models .

  2. Targeted Therapies:

    • Gene Therapy: AAV-mediated LCAT delivery rescues LCAT-deficient mice .

    • Covalent Activators: Compound A (targets Cys31) shows efficacy but risks off-target sulfhydryl interactions .

  3. Structural Insights:

    • Mutations in Arg244 (lid region) disrupt substrate binding, highlighting its role in HDL remodeling .

Product Specs

Introduction
LCAT, or lecithin-cholesterol acyltransferase, is an enzyme that plays a crucial role in cholesterol transport by esterifying cholesterol. This extracellular enzyme is essential for the metabolism of plasma lipoproteins outside of cells.
Description
Recombinant human LCAT, produced in E. coli, is a single polypeptide chain without any glycosylation. It consists of 441 amino acids, including a 25 amino acid His Tag attached to the N-terminus (amino acids 25-440), resulting in a molecular weight of 49.8 kDa. The purification process involves specialized chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The LCAT protein solution has a concentration of 0.5 mg/ml and is supplied in a buffer containing 20mM Tris-HCl (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, freezing at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity exceeding 85%.
Synonyms
Phosphatidylcholine-sterol acyltransferase, Lecithin-cholesterol acyltransferase, Phospholipid-cholesterol acyltransferase, LCAT.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMFWLLN VLFPPHTTPK AELSNHTRPV ILVPGCLGNQ LEAKLDKPDV VNWMCYRKTE DFFTIWLDLN MFLPLGVDCW IDNTRVVYNR SSGLVSNAPG VQIRVPGFGK TYSVEYLDSS KLAGYLHTLV QNLVNNGYVR DETVRAAPYD WRLEPGQQEE 
YYRKLAGLVE EMHAAYGKPV FLIGHSLGCL HLLYFLLRQP QAWKDRFIDG FISLGAPWGG SIKPMLVLAS GDNQGIPIMS SIKLKEEQRI TTTSPWMFPS RMAWPEDHVF ISTPSFNYTG RDFQRFFADL HFEEGWYMWL QSRDLLAGLP APGVEVYCLY GVGLPTPRTY IYDHGFPYTD 
PVGVLYEDGD DTVATRSTEL CGLWQGRQPQ PVHLLPLHGI QHLNMVFSNL TLEHINAILL GAYRQGPPAS PTASPEPPPP E.

Q&A

What is the primary function of Lecithin:Cholesterol Acyltransferase in human metabolism?

Lecithin:Cholesterol Acyltransferase (LCAT) serves as a critical enzyme in lipid metabolism, catalyzing the esterification of free cholesterol to form cholesteryl esters during the process of reverse cholesterol transport . This enzymatic reaction involves transferring a fatty acid from the sn-2 position of phosphatidylcholine (lecithin) to the 3-β-hydroxyl group of cholesterol. The reaction primarily occurs on high-density lipoprotein (HDL) particles, contributing significantly to their maturation from nascent discoidal HDL to spherical HDL particles.

When investigating LCAT function, researchers typically employ enzymatic assays to measure esterification rates using radioisotope-labeled substrates or fluorescent analogs. These assays involve incubating purified LCAT enzyme with appropriate substrates under varying conditions (pH, temperature, ionic strength) to determine optimal enzymatic activity parameters. Kinetic analyses using Michaelis-Menten models help determine Km and Vmax values, providing insights into the enzyme's substrate affinity and catalytic efficiency.

The importance of LCAT in human metabolism extends beyond simple esterification. By converting free cholesterol to cholesteryl esters, LCAT facilitates the packaging of cholesterol within lipoprotein particles, enabling efficient transport through the bloodstream. This process is fundamental to cholesterol homeostasis and potential protection against atherosclerosis, though the latter relationship remains complex.

How do researchers differentiate between normal LCAT function and deficiency in laboratory settings?

Distinguishing normal LCAT function from deficiency requires multiple complementary approaches. The gold standard involves measuring LCAT activity directly using radiometric or fluorescence-based assays. In radiometric assays, researchers incubate patient plasma with radiolabeled substrates (typically [³H]cholesterol), extract lipids, and separate them by thin-layer chromatography to quantify labeled cholesteryl esters formed.

For clinical applications, researchers must establish reference ranges based on healthy control populations, considering variables such as age, sex, and ethnicity. Typical LCAT activity in healthy individuals ranges from 30-60 nmol/ml/h, with values below 10% of normal considered diagnostic for complete LCAT deficiency. Several parameters can confirm LCAT deficiency:

ParameterNormalComplete Deficiency (FLD)Partial Deficiency (FED)
LCAT activity30-60 nmol/ml/h<5% of normal10-30% of normal
Free/total cholesterol ratio<0.3>0.60.3-0.6
HDL-C levels>40 mg/dL<10 mg/dL10-30 mg/dL
Abnormal lipoprotein profileAbsentLpX presentVariable
Corneal opacitiesAbsentPresentPresent
Renal involvementAbsentPresentUsually absent

Beyond biochemical assessments, genetic testing provides definitive confirmation by identifying pathogenic variants in the LCAT gene. Modern next-generation sequencing approaches can rapidly screen for all known LCAT mutations, while functional characterization of novel variants requires expression studies in cellular systems to determine their impact on enzyme activity and secretion.

What experimental models are most effective for studying LCAT function?

Several experimental models offer complementary insights into LCAT function, each with distinct advantages and limitations:

Cell-based models provide controlled conditions for studying LCAT expression, secretion, and activity. Hepatocyte models (HepG2, Huh7, primary hepatocytes) are valuable for investigating LCAT production and regulation, while macrophage models help examine cholesterol efflux processes influenced by LCAT. Advanced three-dimensional culture systems and co-cultures better recapitulate physiological interactions between cell types involved in LCAT metabolism.

Animal models have proven invaluable for understanding LCAT's systemic roles. LCAT knockout mice demonstrate reduced HDL cholesterol levels and accumulation of unesterified cholesterol but surprisingly do not consistently show increased atherosclerosis. Transgenic mice overexpressing human LCAT show elevated HDL-C but sometimes paradoxically increased atherosclerosis, highlighting the complex relationship between LCAT and cardiovascular disease.

Human studies offer the most physiologically relevant insights. These include observational studies of individuals with genetic LCAT variants, interventional studies with recombinant LCAT administration, and ex vivo experiments using patient-derived samples. Recent clinical research has demonstrated promising results from treating FLD patients with recombinant human LCAT, showing improvement in lipid profiles and potential clinical benefits .

When designing experiments, researchers should consider model-specific limitations: cell lines may not fully recapitulate the complex regulation of LCAT in vivo; mouse models lack cholesteryl ester transfer protein (CETP) and have significant differences in lipoprotein metabolism compared to humans; and human studies often face challenges in controlling for confounding variables.

How does LCAT interact with other components of the reverse cholesterol transport pathway?

LCAT functions as a critical node within the reverse cholesterol transport (RCT) pathway, interacting with multiple proteins and lipids to facilitate cholesterol movement from peripheral tissues to the liver. These interactions form a complex network that regulates HDL metabolism and cholesterol homeostasis.

The most immediate interaction partners for LCAT include apolipoprotein A-I (apoA-I), the major protein component of HDL. ApoA-I serves as both a cofactor and an activator for LCAT, increasing its enzymatic activity by approximately 5-7 fold. The C-terminal domain of apoA-I appears particularly important for LCAT activation, while the central helical domains help position LCAT on the HDL surface. Other apolipoproteins, including apoA-IV and apoE, can also activate LCAT, though less efficiently than apoA-I.

LCAT activity is modulated by additional proteins in plasma. Lipid transfer proteins, particularly cholesteryl ester transfer protein (CETP) and phospholipid transfer protein (PLTP), work in concert with LCAT to reshape HDL particles. CETP transfers LCAT-generated cholesteryl esters from HDL to apoB-containing lipoproteins, while PLTP facilitates the transfer of phospholipids to HDL, providing substrates for LCAT.

Several methodological approaches have proven valuable for studying these interactions:

  • Co-immunoprecipitation and cross-linking studies to identify direct protein-protein interactions

  • Surface plasmon resonance and isothermal titration calorimetry to measure binding affinities

  • Fluorescence resonance energy transfer (FRET) to monitor protein associations in real-time

  • Reconstituted HDL particles with defined compositions to determine how specific components affect LCAT activity

Understanding these interactions has significant implications for therapeutic development. For example, rhLCAT infusion studies in mice have demonstrated enhanced cholesterol efflux, highlighting the potential for LCAT-based therapies to influence multiple components of the RCT pathway .

What are the key structural features of the LCAT enzyme that influence its function?

LCAT's structure contains several critical domains and motifs that directly impact its enzymatic function. The mature human LCAT protein consists of 416 amino acids (excluding the 24-amino acid signal peptide) and has a molecular weight of approximately 67 kDa after glycosylation. X-ray crystallography and molecular modeling have revealed a complex architecture with distinct functional regions.

The catalytic domain contains an α/β-hydrolase fold with the catalytic triad (Ser181, Asp345, His377) positioned to facilitate the transfer of fatty acids from lecithin to cholesterol. This active site is partially covered by a flexible lid domain that regulates substrate access and specificity. The lid undergoes conformational changes upon substrate binding, similar to other lipases, creating an interfacial activation mechanism that enhances activity at lipid-water interfaces.

Several post-translational modifications significantly influence LCAT function. The enzyme contains four N-linked glycosylation sites (Asn84, Asn272, Asn384, and Asn397) that affect protein folding, secretion, and plasma half-life. Mutation of these glycosylation sites generally reduces LCAT activity, with Asn272 being particularly important. Additionally, three disulfide bridges (Cys50-Cys74, Cys313-Cys356, and Cys64-Cys97) maintain structural integrity and proper protein folding.

LCAT contains a membrane-binding region that facilitates its interaction with lipoprotein surfaces. This region includes amphipathic helices and hydrophobic residues that can partially insert into the phospholipid monolayer of lipoproteins. Mutations in this region often affect LCAT activity by altering enzyme-substrate positioning rather than directly impacting catalysis.

Researchers use various approaches to study LCAT structure-function relationships, including site-directed mutagenesis, recombinant protein expression, and assays with synthetic substrates. These methods have helped identify critical residues and regions that could serve as targets for therapeutic modulation.

How do genetic variations in LCAT affect enzyme function and clinical phenotypes?

Genetic variations in LCAT present a spectrum of functional consequences, ranging from complete loss of enzyme activity to enhanced function. These variations provide valuable natural experiments for understanding structure-function relationships and their clinical implications.

Familial LCAT Deficiency (FLD) results from mutations causing complete loss of enzyme activity against both HDL and LDL substrates (α-LCAT and β-LCAT activities). Common pathogenic variants include frameshift mutations, nonsense mutations causing premature termination, and missense mutations affecting the catalytic triad or disulfide bridges. FLD patients typically present with corneal opacities, anemia, proteinuria, and progressive renal failure, accompanied by extremely low HDL levels and the presence of abnormal lipoprotein X (LpX) particles.

Fish-Eye Disease (FED) represents a partial LCAT deficiency, with mutations selectively impairing α-LCAT activity while preserving some β-LCAT function. These mutations often affect regions involved in HDL binding or substrate recognition. FED patients typically show corneal opacities and low HDL levels but do not develop severe renal disease, suggesting that residual LCAT activity provides some protection against renal involvement.

Gain-of-function variants are less common but provide interesting insights. The p.Val114Met variant has been associated with increased HDL levels and potentially enhanced cholesterol efflux capacity. Such variants might offer protection against cardiovascular disease, though this relationship remains complex and incompletely understood.

Common polymorphisms, including rs5923 and rs4986970, have been associated with modest variations in plasma HDL levels in population studies. While individual effects are small, these polymorphisms may contribute to the polygenic determination of lipid profiles and potentially modify cardiovascular risk in conjunction with other genetic and environmental factors.

Methodologically, researchers investigate LCAT variants through several complementary approaches:

  • Recombinant expression systems to determine functional effects on enzyme activity, substrate specificity, and protein stability

  • Patient-derived samples to correlate genotypes with biochemical phenotypes

  • Population studies to assess frequency and associations with lipid profiles and disease outcomes

  • Animal models expressing human variants to examine systemic effects

What methodological approaches can resolve discrepancies between in vitro and in vivo LCAT studies?

Researchers frequently encounter discrepancies between in vitro and in vivo LCAT studies, presenting significant challenges for data interpretation and translation. These discrepancies stem from differences in experimental conditions, model systems, and the complex regulatory networks present in living organisms. Several methodological strategies can help bridge this gap.

Standardizing assay conditions represents a critical first step. Researchers should use physiologically relevant substrates and concentrations, maintain consistent temperature and pH, and carefully control enzyme sources. For in vitro studies, reconstituted HDL particles with defined compositions better approximate physiological substrates than synthetic detergent-based systems. Standardized reporting of assay conditions enables more effective comparison across studies.

Bridging approaches provide intermediate complexity between purified systems and whole organisms. Ex vivo studies using fresh plasma or serum preserve the natural milieu of LCAT while allowing controlled experimental manipulation. Whole blood assays maintain cellular interactions that may influence LCAT activity. Perfused organ systems, particularly isolated liver preparations, preserve tissue architecture and cell-cell communications while allowing defined experimental interventions.

Advanced in vivo methodologies offer more sophisticated insights. Stable isotope kinetic studies can track cholesterol movement through different body compartments, providing dynamic information about LCAT's role in cholesterol esterification and transport. Tissue-specific conditional knockout models allow temporal and spatial control of LCAT expression, helping to dissect organ-specific functions. Humanized animal models expressing human LCAT, CETP, and apolipoproteins better recapitulate human lipoprotein metabolism.

Systematic validation approaches enhance confidence in research findings. Testing hypotheses across multiple model systems of increasing complexity can identify specific factors causing discrepancies. Dose-response experiments in both in vitro and in vivo systems help establish physiologically relevant concentration ranges. Combining pharmacological inhibition with genetic approaches provides complementary evidence for LCAT's specific roles.

By implementing these methodological strategies, researchers can improve concordance between different experimental systems and enhance the translational relevance of LCAT research.

How can advanced imaging techniques enhance the study of LCAT in disease models?

Advanced imaging techniques have revolutionized LCAT research by enabling visualization of dynamic processes in living systems at multiple scales, from molecular interactions to whole-organism effects. These methodologies provide unique insights into LCAT's role in lipid metabolism and disease progression.

At the molecular and cellular level, super-resolution microscopy techniques overcome the diffraction limit of conventional light microscopy to visualize LCAT interactions with lipoprotein particles and cell membranes. Techniques such as stochastic optical reconstruction microscopy (STORM) and stimulated emission depletion microscopy (STED) can achieve resolutions of 20-50 nm, suitable for studying LCAT localization on HDL particles (8-12 nm diameter). When combined with fluorescently labeled LCAT and lipoproteins, these approaches can track enzyme-substrate interactions in real-time.

For tissue-level analysis, intravital microscopy enables real-time visualization of LCAT-mediated processes in living animals. Two-photon microscopy can penetrate several hundred micrometers into tissues, allowing researchers to observe labeled LCAT and lipoproteins in organs like liver and kidney. This approach has particular value for studying FLD, where understanding the progression of renal pathology remains a critical research goal.

Whole-body imaging provides system-level insights. Positron emission tomography (PET) with radiolabeled cholesterol analogs can track cholesterol movement through the RCT pathway, while tagged LCAT can monitor enzyme biodistribution and tissue uptake. These approaches are particularly valuable for evaluating rhLCAT therapy, as they can determine optimal dosing regimens and monitor tissue-specific effects.

For atherosclerosis research, multimodal imaging approaches offer comprehensive assessment of plaque development and composition. Intravascular ultrasound, optical coherence tomography, and near-infrared spectroscopy can characterize plaque morphology and lipid content, while PET with specialized tracers can quantify inflammation and metabolic activity. These techniques allow researchers to assess how LCAT modulation affects not just plaque size but also stability and composition, which may have greater clinical relevance.

Effective implementation of these advanced imaging approaches requires careful experimental design, including appropriate controls, validation with complementary techniques, and quantitative image analysis protocols to extract meaningful data from complex datasets.

What are the mechanisms by which recombinant human LCAT (rhLCAT) affects lipid metabolism in therapeutic contexts?

Recombinant human LCAT (rhLCAT) represents a promising therapeutic approach for LCAT deficiency and potentially for broader applications in cardiovascular disease. Understanding its precise mechanisms of action is crucial for optimizing therapeutic protocols and predicting clinical outcomes.

The primary mechanism of rhLCAT therapy involves direct enzymatic effects on plasma lipoproteins. Upon infusion, rhLCAT rapidly associates with HDL particles, converting free cholesterol to cholesteryl esters. This process normalizes the free/total cholesterol ratio, which is typically elevated in LCAT deficiency. In FLD patients, rhLCAT administration has shown promising results in normalizing lipoprotein profiles and potentially improving clinical outcomes .

Beyond direct esterification, rhLCAT stimulates cholesterol efflux from peripheral tissues, including macrophages in atherosclerotic plaques . This effect occurs through several pathways: by maintaining a favorable concentration gradient for free cholesterol movement from cells to HDL; by converting pre-β HDL (minimal acceptors) to mature α-HDL (efficient acceptors); and by modulating the expression of cellular cholesterol transporters like ABCA1 and ABCG1.

The tissue-specific effects of rhLCAT extend to several organ systems. In the kidney, rhLCAT may reduce lipid accumulation in glomeruli and tubular cells, potentially slowing the progression of renal disease in FLD patients. In the vasculature, rhLCAT could modify plaque composition by enhancing cholesterol removal from foam cells, though the net effect on atherosclerosis progression remains uncertain. In the liver, rhLCAT may influence hepatic lipid metabolism and bile acid synthesis, affecting systemic cholesterol homeostasis.

Several methodological approaches have proven valuable for studying rhLCAT mechanisms:

  • Pharmacokinetic/pharmacodynamic modeling to determine optimal dosing regimens

  • Stable isotope studies to track cholesterol movement between compartments

  • Lipoprotein subfraction analysis to characterize changes in particle composition and distribution

  • Gene expression profiling to identify regulated pathways in target tissues

  • Proteomics approaches to identify proteins interacting with rhLCAT in plasma

Emerging research suggests that rhLCAT infusion can enhance cholesterol efflux in mice, including potential enhancement from macrophages, though translation to human physiology requires careful consideration of species differences in lipoprotein metabolism .

What experimental designs can best resolve the paradoxical findings regarding LCAT and atherosclerosis?

The relationship between LCAT and atherosclerosis presents one of the most perplexing paradoxes in lipoprotein metabolism research. Despite LCAT's role in generating mature HDL particles, studies have yielded contradictory findings regarding its impact on atherosclerosis. Resolving these paradoxes requires sophisticated experimental designs that address the complexity of lipoprotein metabolism and atherogenesis.

Comprehensive animal models that better recapitulate human lipoprotein metabolism represent a critical approach. Traditional LCAT knockout mice lack CETP and have fundamentally different lipoprotein profiles compared to humans. More sophisticated models include:

  • "Humanized" mice expressing human LCAT, CETP, apoA-I, and apoB

  • LCAT transgenic or knockout models on backgrounds prone to atherosclerosis (ApoE-/- or LDLR-/-)

  • Tissue-specific and inducible LCAT expression systems to distinguish developmental from adult effects

  • Models combining LCAT modification with other HDL-modifying genes to capture pathway interactions

Detailed plaque characterization beyond simple area measurements is essential. Advanced approaches include:

  • Compositional analysis of plaques (lipid core size, cellular content, collagen, calcification)

  • Inflammatory marker assessment within plaques using immunohistochemistry or laser capture microdissection

  • Assessment of plaque stability features (fibrous cap thickness, neovascularization)

  • Functional vascular studies (endothelial function, arterial stiffness)

Molecular pathway analysis can help identify mechanisms underlying paradoxical findings:

  • Cholesterol flux studies using labeled cholesterol to quantify actual reverse cholesterol transport rates

  • Investigation of inflammation-modifying effects of LCAT independent of cholesterol esterification

  • Analysis of specific HDL subpopulations and their functional properties

  • Examination of potential pro-oxidant or pro-inflammatory effects of LCAT-modified HDL under certain conditions

Temporal considerations may resolve some paradoxes. The effect of LCAT on atherosclerosis might differ at various disease stages, with intervention studies testing LCAT modulation at different timepoints providing valuable insights. Similarly, the duration of LCAT alteration may influence outcomes, with acute versus chronic LCAT changes potentially having opposing effects.

As ongoing research continues to explore these complex relationships, the current consensus acknowledges that while LCAT is essential for HDL maturation, its role in atherosclerosis remains elusive, requiring continued investigation with increasingly sophisticated experimental approaches .

Product Science Overview

Structure and Function

LCAT is a member of the phospholipid:diacylglycerol acyltransferases family. It is primarily bound to HDL particles in the blood plasma, where it catalyzes the transfer of fatty acids from lecithin (phosphatidylcholine) to free cholesterol, forming cholesteryl ester and lysolecithin . This reaction is critical for the conversion of nascent discoidal HDL particles into mature spherical HDL particles, which are more efficient in cholesterol transport.

Clinical Significance

LCAT deficiency can lead to several health issues, including impaired vision due to cholesterol corneal opacities, anemia, and kidney damage . The enzyme’s role in HDL metabolism and RCT makes it a potential therapeutic target for cardiovascular diseases, particularly atherosclerosis. Increasing LCAT activity has been shown to enhance HDL functionality and promote cholesterol efflux from macrophages, reducing the risk of plaque formation in arteries .

Recombinant Human LCAT

Recombinant human LCAT (rhLCAT) has been developed to study its therapeutic potential. In clinical studies, rhLCAT has demonstrated the ability to increase HDL cholesterol levels and improve HDL functionality. For instance, in a Phase 2a study, multiple ascending doses of rhLCAT (MEDI6012) were administered to patients with stable atherosclerosis. The results showed significant dose-related increases in HDL cholesterol, HDL cholesterol ester, and total cholesterol ester levels . Additionally, rhLCAT has been shown to rescue defective HDL-mediated endothelial protection in patients with acute coronary syndrome, suggesting its potential in restoring HDL functionality and promoting vascular health .

Research and Development

Ongoing research aims to further understand the mechanisms by which LCAT influences lipid metabolism and to explore its therapeutic applications. Studies have shown that rhLCAT can restore HDL-mediated nitric oxide production in endothelial cells, which is crucial for maintaining vascular tone and preventing inflammation . These findings highlight the enzyme’s potential in treating cardiovascular diseases and improving overall cardiovascular health.

In conclusion, Lecithin-Cholesterol Acyltransferase (Human Recombinant) is a vital enzyme in lipid metabolism with significant therapeutic potential. Its role in HDL maturation and reverse cholesterol transport makes it a promising target for cardiovascular disease treatment and prevention.

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.