LTF Apo Human

Lactoferrin Apo Human Recombinant
Shipped with Ice Packs
In Stock

Description

Recombinant Human Apo Lactoferrin produced in Plant is a glycosylated mature polypeptide sequence having an approximate molecular mass of 80 kDa.
The Human Apo  Lactoferrin is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Lactoferrin, a glycoprotein in the transferrin family, binds iron. Present in breast milk, tears, and other secretions, it inhibits bacterial growth by sequestering iron. Its N-terminal acts as an antimicrobial peptide, and it synergizes with lysozyme. Lactoferrin is considered an innate defense protein, offering first-line protection against pathogens. Its functions include iron binding, antibacterial, antifungal, and antiviral activities, antioxidant properties, and immunomodulatory effects. Ongoing research explores its various biological roles, making it a promising candidate for diverse applications.
Description
Recombinant Human Apo Lactoferrin, produced in plants, is a glycosylated polypeptide with a molecular weight of approximately 80 kDa. It is purified using proprietary chromatographic methods.
Physical Appearance
Lyophilized powder, pink in color.
Formulation
The lyophilized Human Apo Lactoferrin is additive-free.
Stability
While stable at room temperature for up to three weeks, Recombinant Apo Lactoferrin should ideally be stored at 2-8°C.
Solubility
For reconstitution, dissolve the lyophilized LTF Apo Human in sterile water at a concentration of 10 mg/ml. This solution can be further diluted as needed.
Purity
Purity exceeds 95% as determined by SDS-PAGE analysis.
Synonyms
Lactotransferrin, Lactoferrin, Growth-inhibiting protein 12, Talalactoferrin, LTF, GIG12, LF, HLF2, Neutrophil Lactoferrin.
Source
Rice Flour.

Q&A

What is human apo-lactoferrin and how does it differ from other forms of lactoferrin?

Human apo-lactoferrin (ApoLf) is the iron-free form of lactoferrin, a natural glycoprotein with high binding affinity for iron. Unlike holo-lactoferrin (iron-bound form), apo-lactoferrin's tertiary structure differs significantly, allowing it to efficiently chelate iron from the surrounding environment. This iron-binding capability makes it particularly relevant in conditions involving iron dysregulation, such as Alzheimer's disease, where iron accumulation contributes to oxidative stress and neurodegeneration. The distinction between apo- and holo-forms is critical because ApoLf changes its conformational structure when it binds iron, altering its functional properties . This conformational flexibility enables ApoLf to serve as both an iron scavenger in its apo form and a delivery protein in its holo form, making it particularly valuable for therapeutic applications targeting iron homeostasis.

What biological functions make apo-lactoferrin relevant to neurodegenerative disease research?

Apo-lactoferrin possesses several biological functions that make it particularly relevant to neurodegenerative disease research:

  • Metal chelation: ApoLf has high binding affinity for iron, allowing it to address the iron accumulation in the brain associated with Alzheimer's disease. This accumulation normally contributes to oxidative stress and neuronal damage .

  • Antioxidant properties: Beyond iron chelation, ApoLf provides additional antioxidant protection, potentially preventing the formation of free radicals that damage neurons in neurodegenerative conditions .

  • Anti-aggregation effects: Emerging evidence suggests ApoLf may interfere with the accumulation and aggregation of deformed beta amyloid and hyperphosphorylated tau proteins, which form senile plaques and neurofibrillary tangles characteristic of Alzheimer's disease .

  • Dual neuroprotection and neurotherapy: When conjugated with therapeutic agents like galantamine, ApoLf can potentially deliver active compounds while simultaneously removing excess iron, offering a combinatorial approach to disease management .

These properties position apo-lactoferrin as a promising multifunctional agent in the research and potential treatment of neurodegenerative disorders where metal dysregulation plays a significant pathological role.

How should researchers design experiments to evaluate apo-lactoferrin-galantamine conjugates for Alzheimer's disease?

When designing experiments to evaluate apo-lactoferrin-galantamine (ApoLf-Gal) conjugates for Alzheimer's disease, researchers should implement a multi-tiered experimental approach:

  • Conjugate characterization studies:

    • Physical characterization of the self-assembly mechanism through which "ApoLf results in encapsulation of Gal"

    • Stability assessment under physiological conditions

    • Confirmation of maintained functional properties of both components

  • In vitro experimental design:

    MechanismAppropriate AssaysControl Groups
    Iron chelationFerrozine assay, isothermal titration calorimetryFree iron, ApoLf alone
    Cholinesterase inhibitionEllman's assay, enzyme kineticsGalantamine alone
    Antioxidant activityDPPH, ABTS, ORAC assaysVitamin E, ApoLf alone, Gal alone
    Amyloid interactionThT fluorescence, aggregation kineticsAmyloid alone, known inhibitors
    Neuronal protectionPrimary neuron/SH-SY5Y viability assaysUntreated, Aβ-exposed cells
  • Experimental design should incorporate appropriate quantitative research approaches, such as comparative or experimental designs, which allow for statistical comparison between treatment groups .

  • In vivo experimental protocols:

    • Use of transgenic mouse models of Alzheimer's disease

    • Treatment groups: vehicle, ApoLf alone, Gal alone, ApoLf-Gal conjugate

    • Multiple endpoints: cognitive (Morris water maze), biochemical (iron levels, AChE activity), and histological (amyloid plaque burden)

    • Time-course studies to assess acute versus chronic effects

  • Statistical analysis approaches should follow methodologies used in similar research: "Two-tailed Student's t-test and by ANOVA with age correction. Logarithmic transformation was used to normalise distributions" .

This comprehensive experimental design allows researchers to evaluate both the individual components and potential synergistic effects of the ApoLf-Gal conjugate as a dual neuroprotective and neurotherapeutic intervention.

What quantitative research designs are most appropriate for studying apo-lactoferrin's effects in clinical populations?

When studying apo-lactoferrin's effects in clinical populations, researchers should select quantitative research designs based on their specific research questions and constraints. According to established methodology, "There are several types of quantitative research designs, such as the experimental, comparative or predictive correlational designs" , each offering distinct advantages for ApoLf research:

  • Experimental designs:

    • Randomized controlled trials offer the strongest evidence for causal relationships between ApoLf interventions and outcomes

    • Should include carefully defined inclusion/exclusion criteria, predetermined primary and secondary endpoints, and appropriate sample size calculations

    • Particularly valuable for testing ApoLf-based therapeutic interventions

  • Comparative designs:

    • Case-control studies comparing ApoLf levels or activity between AD patients and matched controls

    • Cross-sectional studies examining relationships between ApoLf parameters and disease severity

    • Statistical approach should follow methods used in similar research: "Categorical variables were compared by chi-square or Fisher's exact test. Differences between continuous variables were evaluated by two-tailed Student's t-test and by ANOVA"

  • Correlational designs:

    • Examining relationships between ApoLf levels/function and disease biomarkers

    • Longitudinal studies tracking ApoLf parameters over disease progression

    • Analyzing genetic influences on ApoLf expression and function using approaches similar to: "Genotype distributions and allele frequencies between the study groups were compared by construction of 2 × 2 and 2 × 3 contingency tables"

  • Power and sample size considerations:

    • Studies should be "powered to allow detection with 80% and an error rate of 5%"

    • Account for potential confounding variables through statistical adjustment

    • Consider logarithmic transformation for non-normally distributed data, similar to approaches used in apolipoprotein research

The selection of appropriate quantitative research designs is critical for generating valid, reliable evidence regarding apo-lactoferrin's potential role in neurodegenerative disease management.

How can researchers develop transgenic animal models to study human apo-lactoferrin?

Developing transgenic animal models to study human apo-lactoferrin requires strategic genetic and experimental approaches. Drawing from methodologies used in apolipoprotein research, researchers should consider:

  • Gene construct design:

    • Human LTF gene with appropriate regulatory elements to ensure physiologically relevant expression

    • Consideration of incorporating methods similar to those used for apolipoprotein research: "LPA-YAC transgenic mice" , which utilize yeast artificial chromosomes for large gene constructs

    • Inclusion of reporter elements to track expression patterns

  • Transgenic model validation:

    Validation StepMethodologyExpected Outcome
    Gene integrationPCR, Southern blotConfirmation of transgene presence
    mRNA expressionRT-PCR, RNA-seqTissue-specific expression pattern
    Protein expressionWestern blot, ELISADetection of human ApoLf
    Functional validationIron binding assaysConfirmation of chelation activity
    Phenotypic analysisBehavioral, biochemical testsAssessment of functional effects
  • Experimental approaches should include:

    • Dietary manipulations similar to those used in apolipoprotein studies: "Feeding of a high-cholesterol/high-fat (HCHF) diet" to assess nutritional regulation

    • Crossbreeding with disease models, similar to approaches where "LPA-YAC transgenic and mice transgenic for the human APOB (hAPOB) gene were crossed"

    • Tissue-specific expression analysis to confirm expected distribution patterns

  • Ensure ethical compliance:
    "The Norwegian Animal Research Authority approved the experiments and all animal experiments were performed in accordance with the Norwegian Gene Technology Act" . Similarly, all LTF transgenic studies must adhere to appropriate institutional and regulatory approvals.

  • Specialized transgenic approaches may include:

    • Inducible expression systems to control timing of human LTF expression

    • Tissue-specific promoters to target expression to relevant organs

    • Knockin models to replace mouse Ltf with human LTF

    • CRISPR/Cas9 technology for precise genetic modifications

These approaches allow researchers to investigate human apo-lactoferrin's biological functions, regulatory mechanisms, and therapeutic potential in physiologically relevant in vivo systems.

What statistical methods should be used to analyze genetic influences on apo-lactoferrin expression and function?

When analyzing genetic influences on apo-lactoferrin expression and function, researchers should employ rigorous statistical methods similar to those used in apolipoprotein research:

  • Heritability estimation:
    Methods similar to those used in apolipoprotein studies are recommended: "Weighted midparent–offspring regression estimated a heritability h²=0.76. The correlation of Lp(a) levels associated with LPA alleles identical by descent (IBD) resulted in a heritability estimate of 0.801" . These approaches can determine the genetic contribution to ApoLf variation.

  • Genetic association analysis:

    • For comparing genotype distributions: "Genotype distributions and allele frequencies between the study groups were compared by construction of 2 × 2 and 2 × 3 contingency tables and chi-square analysis"

    • For estimating disease risk: "To estimate the risk of CAD and the progression of CAD associated with gene variants, odds ratios were calculated by multiple regression analysis, after adjustment for other modulators"

  • Data transformation and normalization:
    "Logarithmic transformation was used to normalise distributions of BMI, plasma total and HDL cholesterol, triglycerides and Lp(a) values" . Similar transformations may be necessary for ApoLf data to meet statistical test assumptions.

  • Genotype-phenotype correlations:
    "The relation between genotypes and concomitant variables were evaluated by ANOVA after age-standardisation" . This approach should be applied to analyze relationships between LTF variants and ApoLf expression or function.

  • Multiple testing correction:
    Given the potential for numerous genetic variants and phenotypic measures, appropriate correction for multiple testing is essential to avoid false positive associations.

  • Power considerations:
    Studies should be "powered to allow detection with 80% and an error rate of 5% for differences in allele frequencies of 11%" . Similar power calculations should be performed for LTF genetic studies.

These statistical approaches enable researchers to rigorously assess genetic influences on apo-lactoferrin, providing insights into the hereditary components of its expression, structure, and function.

How should researchers interpret contradictory findings in apo-lactoferrin research?

When researchers encounter contradictory findings in apo-lactoferrin research, a systematic approach to interpretation and reconciliation is essential:

By systematically analyzing contradictory findings through these approaches, researchers can advance understanding of apo-lactoferrin biology despite initial inconsistencies in the literature.

How can researchers effectively measure and analyze apo-lactoferrin's conformational changes upon iron binding?

Effectively measuring and analyzing apo-lactoferrin's conformational changes upon iron binding requires sophisticated biophysical techniques and careful experimental design:

  • Spectroscopic techniques:

    TechniqueApplicationAnalytical Considerations
    Circular dichroismSecondary structure changesRequires baseline correction and buffer matching
    Intrinsic fluorescenceTertiary structure alterationsAccounts for aromatic amino acid environments
    FTIR spectroscopyBond vibrations reflecting structureRequires careful water subtraction
    NMR spectroscopyAtomic-level structural informationProvides detailed but complex data
  • Experimental design considerations:

    • Titration experiments with increasing iron concentrations

    • Time-resolved measurements to capture kinetics of conformational change

    • Comparison of different iron species (Fe²⁺ vs. Fe³⁺)

    • Control experiments with other metals to assess specificity

  • Data analysis approaches:

    • Differential analysis between apo and holo states

    • Calculation of binding constants from spectroscopic changes

    • Principal component analysis to identify major conformational states

    • Correlation of spectroscopic data with functional outcomes

  • Integration with functional assays:

    • Since "ApoLf changes its conformational structure in the presence of iron" , correlate structural changes with:

      • Iron binding capacity

      • Antioxidant activity

      • Interaction with therapeutic conjugates like galantamine

      • Ability to interfere with amyloid aggregation

  • Advanced structural characterization:

    • X-ray crystallography of apo and holo forms

    • Cryo-electron microscopy for larger complexes

    • Molecular dynamics simulations to model transition states

    • Hydrogen-deuterium exchange mass spectrometry for dynamics

These approaches allow researchers to comprehensively characterize the conformational plasticity of apo-lactoferrin upon iron binding, which is crucial for understanding its biological function and therapeutic potential, particularly in the context of neurodegenerative disease management.

What experimental approaches can assess apo-lactoferrin's potential in Alzheimer's disease management?

Assessing apo-lactoferrin's potential in Alzheimer's disease management requires multifaceted experimental approaches that evaluate both its mechanistic actions and therapeutic efficacy:

  • Mechanistic studies for iron chelation:

    • Quantitative assessment of iron binding capacity using isothermal titration calorimetry

    • Evaluation of iron removal from amyloid-β aggregates

    • Measurement of reduced oxidative stress markers in neuronal models

    • Assessment of ApoLf's ability to "pick up excess iron when in contact with iron"

  • Amyloid interaction studies:

    • Thioflavin T fluorescence assays to measure effects on amyloid aggregation

    • Surface plasmon resonance to quantify binding to amyloid species

    • Electron microscopy to visualize effects on fibril morphology

    • Cell-based assays measuring protection against amyloid toxicity

  • Combinatorial therapeutic evaluation:

    • Assessment of ApoLf-Gal conjugates that represent "a combinatorial antioxidant-metal-chelator approach"

    • Comparison of efficacy between single agents and combination therapy

    • Analysis of synergistic effects on multiple AD pathological pathways

    • Evaluation of drug delivery and targeting capabilities

  • Preclinical in vivo studies:

    • Cognitive assessment in AD model animals using validated behavioral tests

    • Biomarker analysis including amyloid load, tau phosphorylation, and oxidative stress

    • Brain iron quantification using techniques like MRI T2* mapping

    • Histopathological evaluation of treated versus untreated animals

  • Statistical approaches:

    • Implementation of appropriate quantitative research designs for intervention studies

    • Statistical analysis following established methods: "Two-tailed Student's t-test and by ANOVA with age correction"

    • Data normalization through "logarithmic transformation" when necessary

    • Correlation analysis between biomarkers and functional outcomes

These experimental approaches provide comprehensive evidence regarding apo-lactoferrin's potential efficacy in Alzheimer's disease management, particularly through its dual role in iron chelation and direct interaction with disease-specific pathological processes.

How can researchers evaluate the pharmacokinetics of apo-lactoferrin-galantamine conjugates?

Evaluating the pharmacokinetics of apo-lactoferrin-galantamine conjugates requires specialized approaches that account for the unique properties of this proteo-alkaloid combination:

  • Bioanalytical method development:

    • Development of specific assays to quantify both ApoLf and galantamine components

    • Validation of methods for detecting the intact conjugate versus individual components

    • Establishment of appropriate internal standards and quality controls

    • Consideration of matrix effects in biological samples

  • Absorption and distribution studies:

    ParameterMethodologyKey Considerations
    Absorption profileBlood sampling after administrationDifferent routes (oral, parenteral)
    Tissue distributionQuantification in multiple organsSpecial focus on brain penetration
    Blood-brain barrier penetrationCSF sampling, brain microdialysisCritical for AD applications
    Cellular uptakeFluorescently-labeled conjugate trackingCell type-specific differences
  • Metabolism and excretion analysis:

    • Identification of metabolic products using LC-MS/MS

    • Assessment of galantamine release kinetics from the conjugate

    • Evaluation of how "ApoLf changes its conformational structure in the presence of iron" affects drug release

    • Determination of elimination routes and half-life

  • Special considerations for iron interaction:

    • Analysis of how conjugate pharmacokinetics change with iron binding

    • Evaluation of whether the conjugate "pick[s] up excess iron when in contact with iron"

    • Assessment of how iron binding affects biodistribution and drug release

  • Comparison with individual components:

    • Parallel assessment of unconjugated ApoLf and galantamine

    • Evaluation of whether conjugation alters the pharmacokinetic profile of either component

    • Analysis of potential advantages in terms of half-life, target tissue delivery, or blood-brain barrier penetration

  • Statistical analysis:

    • Application of non-compartmental and compartmental modeling

    • Use of appropriate data transformations for non-normal distributions similar to "logarithmic transformation" approaches in related research

    • Correlation of pharmacokinetic parameters with pharmacodynamic outcomes

This comprehensive approach to pharmacokinetic evaluation provides critical information for optimizing dosing regimens, predicting therapeutic efficacy, and anticipating potential drug interactions of apo-lactoferrin-galantamine conjugates.

What techniques should researchers use to optimize apo-lactoferrin-galantamine conjugate formulation?

Optimizing apo-lactoferrin-galantamine conjugate formulation requires systematic application of pharmaceutical and biophysical techniques to achieve maximum therapeutic efficacy:

  • Conjugation method optimization:

    • Refinement of the self-assembly process through which "conjugation is achieved through self-assembly of ApoLf which results in encapsulation of Gal"

    • Evaluation of different conjugation chemistries (if applicable)

    • Assessment of conjugation efficiency under varying conditions

    • Characterization of conjugate stoichiometry and structure

  • Physical characterization:

    PropertyAnalytical TechniqueSignificance
    Particle sizeDynamic light scatteringAffects biodistribution
    Surface chargeZeta potential measurementInfluences stability and cell interaction
    MorphologyTransmission electron microscopyStructural organization
    Thermal stabilityDifferential scanning calorimetryFormulation stability
    Drug loadingHPLC, UV spectroscopyTherapeutic content
  • Functional characterization:

    • Iron binding capacity assays to confirm ApoLf functionality

    • Acetylcholinesterase inhibition assays to verify galantamine activity

    • Conformational analysis to ensure "ApoLf changes its conformational structure in the presence of iron"

    • Antioxidant activity measurement to confirm dual functionality

  • Stability optimization:

    • Accelerated stability studies under various storage conditions

    • Evaluation of excipients to enhance stability

    • Lyophilization studies for improved shelf-life

    • Assessment of stability in biological fluids

  • Drug release characteristics:

    • In vitro release kinetics of galantamine from the conjugate

    • Effect of iron presence on drug release profile

    • Development of controlled-release formulations if desired

    • Correlation between in vitro release and in vivo performance

  • Scale-up considerations:

    • Process parameter optimization for reproducible manufacturing

    • Critical quality attribute identification and control

    • Method transfer for analytical procedures

    • Batch-to-batch consistency evaluation

These techniques enable researchers to develop optimized apo-lactoferrin-galantamine conjugate formulations with appropriate physiochemical properties, functional characteristics, and stability profiles to support further preclinical and potential clinical development for Alzheimer's disease management.

How can dietary interventions be designed to study apo-lactoferrin regulation?

Designing dietary interventions to study apo-lactoferrin regulation requires careful consideration of experimental parameters, drawing from approaches used in related research:

  • Diet formulation and experimental design:

    • Development of defined diets with controlled nutrient composition, similar to studies using "high-cholesterol/high-fat (HCHF) diet" and "AIN-76 diet" as control

    • Implementation of appropriate feeding protocols with measured food intake

    • Inclusion of relevant experimental groups to isolate dietary effects

    • Consideration of both acute and chronic dietary exposures

  • Comprehensive assessment parameters:

    ParameterMethodologyResearch Relevance
    ApoLf gene expressionRT-qPCR, similar to measuring "hepatic apo(a) mRNA level" Transcriptional regulation
    Protein levelsELISA, Western blot for "serum total apo(a) level" Translational effects
    Functional assaysIron binding capacity, conformational analysisFunctional impacts
    Related biomarkersLipid profile, iron status, oxidative stress markersContextual changes
  • Time-course considerations:

    • Collection of baseline measurements before dietary intervention

    • Multiple sampling timepoints to capture dynamic responses

    • Sufficient intervention duration to observe chronic adaptations

    • Recovery period to assess reversibility of effects

  • Statistical approaches:

    • Implementation of appropriate statistical methods similar to those used in apolipoprotein research

    • Sample size determination based on expected effect sizes

    • Consideration of individual variability and potential need for logarithmic transformation of data

    • Analysis of correlations between dietary components and ApoLf parameters

  • Mechanistic investigations:

    • Analysis of signaling pathways mediating dietary effects

    • Evaluation of epigenetic modifications influencing gene expression

    • Assessment of post-translational modifications affecting protein function

    • Identification of dietary components directly interacting with ApoLf

By implementing these methodological approaches, researchers can systematically investigate how dietary factors influence apo-lactoferrin expression, secretion, and function, potentially identifying nutritional strategies to modulate its biological activities in health and disease.

What methods should be used to study the interaction between apo-lactoferrin and the blood-brain barrier?

Studying the interaction between apo-lactoferrin and the blood-brain barrier (BBB) requires specialized methodological approaches that can evaluate both transport mechanisms and therapeutic implications:

  • In vitro BBB models:

    • Establishment of endothelial cell monolayers (hCMEC/D3, primary brain endothelial cells)

    • Development of co-culture systems incorporating astrocytes and pericytes

    • Microfluidic "organ-on-chip" platforms mimicking BBB physiological conditions

    • Measurement of transendothelial electrical resistance (TEER) to verify barrier integrity

  • Transport studies:

    ApproachMethodologyOutcome Measure
    Transwell assaysLabeled ApoLf transport across cell layersPermeability coefficients
    Receptor identificationCompetition studies with known ligandsTransport mechanism
    Vesicular traffickingConfocal microscopy with trafficking markersIntracellular pathway
    Iron dependencyVarying iron conditions to assess "ApoLf changes its conformational structure in the presence of iron" Conformation-dependent transport
  • In vivo BBB penetration studies:

    • Biodistribution studies with labeled ApoLf or ApoLf-Gal conjugates

    • Cerebrospinal fluid sampling at multiple timepoints

    • Brain microdialysis for direct measurement in extracellular fluid

    • Ex vivo brain slice techniques to assess regional distribution

  • Mechanistic investigations:

    • Studies of potential receptor-mediated transport (LRP1, transferrin receptor)

    • Assessment of adsorptive-mediated transcytosis

    • Evaluation of how iron binding affects BBB transport

    • Investigation of ApoLf effects on BBB integrity and function

  • Therapeutic delivery applications:

    • Assessment of ApoLf as a delivery vector for drugs like galantamine

    • Comparison of brain uptake between free galantamine and ApoLf-Gal conjugates

    • Evaluation of whether conjugates can "deliver Gal and pick up excess iron" across the BBB

    • Development of strategies to enhance BBB penetration

  • Statistical analysis:

    • Appropriate data transformation for non-normal distributions

    • Comparative analysis between different experimental conditions

    • Correlation between in vitro and in vivo findings

    • Pharmacokinetic modeling of brain exposure

These methodological approaches provide comprehensive insights into apo-lactoferrin's interaction with the blood-brain barrier, critical information for developing effective neurotherapeutic interventions for conditions like Alzheimer's disease.

How should genetic studies be designed to understand variants in the human LTF gene?

Designing genetic studies to understand variants in the human LTF gene requires rigorous methodological approaches similar to those employed in apolipoprotein research:

  • Study population selection:

    • Definition of appropriate case and control groups

    • Consideration of population stratification and ancestry

    • Sample size determination: studies should be "powered to allow detection with 80% and an error rate of 5% for differences in allele frequencies"

    • Family-based designs for heritability estimation, similar to approaches yielding "heritability estimate of 0.801"

  • Genetic analysis strategies:

    ApproachMethodologyApplication
    Candidate variant analysisPCR, TaqMan assaysSpecific known variants
    Haplotype mappingMultiple marker genotypingExtended genetic regions
    Next-generation sequencingTargeted, exome, or whole-genomeNovel variant discovery
    Functional genomicsReporter assays, EMSARegulatory variant effects
  • Statistical genetic methods:

    • Allele and genotype frequency comparisons: "Genotype distributions and allele frequencies between the study groups were compared by construction of 2 × 2 and 2 × 3 contingency tables and chi-square analysis"

    • Risk assessment: "odds ratios were calculated by multiple regression analysis, after adjustment for other modulators"

    • Heritability estimation using "weighted midparent–offspring regression" approaches

    • Appropriate transformation of phenotypic data: "Logarithmic transformation was used to normalise distributions"

  • Genotype-phenotype correlation:

    • Analysis of how LTF variants affect ApoLf levels and function

    • Assessment of variants in relation to disease phenotypes

    • Evaluation of gene-environment interactions, particularly dietary effects similar to HCHF diet studies

    • Integration with expression data (eQTL analysis)

  • Advanced genetic approaches:

    • Genome-wide association studies for LTF-related traits

    • Polygenic risk score development incorporating LTF variants

    • Mendelian randomization to assess causality

    • Trans-ethnic genetic studies to identify conserved mechanisms

  • Functional validation:

    • Creation of cell models expressing variant forms of LTF

    • Development of transgenic animal models similar to approaches for apolipoprotein genes where "LPA-YAC transgenic and mice transgenic for the human APOB (hAPOB) gene were crossed"

    • Structural biology to assess variant effects on protein function

These comprehensive genetic study approaches enable researchers to systematically investigate how genetic variation in the LTF gene influences apo-lactoferrin expression, structure, function, and relationship to disease phenotypes.

Product Science Overview

Introduction

Lactoferrin Apo, also known as Apolactoferrin, is a form of lactoferrin that is devoid of iron. Lactoferrin itself is a multifunctional glycoprotein found in various secretory fluids, including milk, saliva, tears, and nasal secretions. It plays a crucial role in the immune system, exhibiting antibacterial, antiviral, antifungal, and anti-inflammatory properties .

Discovery and Structure

Lactoferrin was first isolated from human milk by scientist Johanson Bengt in 1960 . It is composed of 703 amino acids and has a molecular weight of approximately 80 kDa . The protein consists of two lobes, each capable of binding one iron ion, which is essential for its biological functions .

Functions and Mechanisms

Lactoferrin’s primary function is to bind and transport iron, which is vital for various physiological processes. By sequestering iron, lactoferrin limits the availability of this essential nutrient to pathogenic microorganisms, thereby inhibiting their growth . Additionally, lactoferrin interacts with cellular receptors, modulating immune responses and promoting the growth of beneficial bacteria in the gut .

Recombinant Production

Recombinant human lactoferrin is produced using genetic engineering techniques. The gene encoding human lactoferrin is inserted into host cells, such as bacteria, yeast, or mammalian cells, which then express the protein. This method allows for large-scale production of lactoferrin with similar biological activity to the natural protein .

Applications

Lactoferrin Apo has numerous applications in healthcare and biotechnology. It is used in dietary supplements, infant formula, and functional foods due to its immune-boosting properties . Additionally, lactoferrin is explored as a potential therapeutic agent for various conditions, including infections, inflammation, and cancer .

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.