SERPINA1 Human, Active

Alpha-1 Antitrypsin, Active Human Recombinant
Shipped with Ice Packs
In Stock

Description

SERPINA1 Human Recombinant produced in rice is a single, non-glycosylated polypeptide chain containing 384 amino acids and having a molecular mass of 43.1 kDa.
The SERPINA1 protein is purified by proprietary chromatographic techniques.

Product Specs

Introduction

SERPINA1, a secreted serine protease inhibitor, plays a crucial role in regulating proteolytic activity by targeting enzymes like elastase, plasmin, and collagenase. Genetic defects in SERPINA1 can lead to conditions such as emphysema or liver disease. Notably, it demonstrates in vitro inhibition of human recombinant matriptase's catalytic domain. SERPINA1 levels surge as part of the acute phase response to inflammation and tissue damage. The interplay between SERPINA1 and SLC11A1 genes significantly influences the balance of elastase produced by leukocytes during phagocytosis.

Description
Recombinant human SERPINA1, produced in rice, is a single, non-glycosylated polypeptide chain with a molecular weight of 43.1 kDa, comprising 384 amino acids. The purification process involves proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White Lyophilized (Freeze-dried) Powder
Formulation
The lyophilized SERPINA1 is prepared in a concentrated solution containing recombinant Albumin.
Solubility
For reconstitution, it is recommended to dissolve the lyophilized SERPINA1 in sterile 18M-cm H2O at a concentration not less than 100 µg/ml. This solution can then be further diluted in other aqueous solutions as needed.
Stability
While lyophilized SERPINA1 remains stable at room temperature for up to 3 weeks, it is recommended to store it desiccated below -18°C. After reconstitution, SERPINA1 should be stored at 4°C for 2-7 days. For long-term storage, freezing below -18°C is recommended. It is advisable to add a carrier protein (0.1% HSA or BSA) for long-term storage and to avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 95% by SDS-PAGE analysis.
Biological Activity
The biological activity is determined to be 3-5 mg of SERPINA1 inhibiting 1 mg of PPE, with a specific activity of 10.8 units per mg of protein.
Synonyms
Alpha-1-antitrypsin, Alpha-1 protease inhibitor, Alpha-1-antiproteinase, SERPINA1, A1AT, PI, A1A, AAT, PI1, MGC9222, PRO2275, MGC23330.
Source
Rice Grain (Oryza Sativa).

Q&A

What is SERPINA1 and what is its primary biological function?

SERPINA1 (serpin peptidase inhibitor, clade A, member 1) encodes alpha-1 antitrypsin (A1AT), a critical serine protease inhibitor primarily expressed in the liver and secreted into the bloodstream. It circulates to the lungs where it functions to neutralize lung proteases (particularly elastase) and maintain lung elasticity and function. SERPINA1 is a key inhibitor targeting multiple proteases including elastase, plasmin, collagenase, leucocytic proteases, trypsin, chymotrypsin, and plasminogen activator . Deficiency of functional SERPINA1 is associated with conditions such as emphysema and liver disease . Recent research has also revealed a novel metabolic role for SERPINA1 as a hepatokine that influences adipocyte function, energy expenditure, and glucose metabolism .

How is SERPINA1 expression regulated at the transcriptional and post-transcriptional levels?

SERPINA1 expression involves complex regulatory mechanisms at both transcriptional and post-transcriptional levels:

Transcriptional regulation:

  • SERPINA1 shows tissue-specific expression patterns, with highest expression in the liver .

  • Expression increases as an acute phase response to tissue necrosis and inflammation .

Post-transcriptional regulation:

  • The SERPINA1 gene generates 11 distinct mRNA isoforms through alternative splicing events that exclusively affect the 5′-untranslated region (5′-UTR) .

  • Each transcript variant contains a unique 5′-UTR but encodes the identical protein sequence .

  • RNA structure within the 5′-UTR significantly influences translation efficiency by regulating ribosome accessibility to start codons .

  • The NM_000295.4 isoform represents approximately 23% of total SERPINA1 mRNA in lung tissue and lacks upstream open reading frames (uORFs), potentially making it one of the most efficiently translated isoforms .

Epigenetic regulation:

  • DNA methylation patterns in the SERPINA1 gene locus affect expression levels, with hypomethylation observed in some COPD patients .

  • Specific CpG sites (cg02181506 and cg24621042) in the SERPINA1 gene have been identified as methylation marks associated with COPD, independent of disease severity and smoking history .

What methodological approaches are recommended for studying SERPINA1 variants in clinical samples?

When investigating SERPINA1 variants in clinical contexts, researchers should follow these methodological approaches:

  • Comprehensive sequencing strategy: Complete sequencing of the SERPINA1 gene is essential, as pathogenic variants may be more frequent than previously recognized . This should include:

    • Coding regions (exons)

    • Intronic regions

    • 5' and 3' untranslated regions, as these can contain regulatory variants

  • Integrative analysis approach:

    • Combine genotyping with protein quantification (measure serum A1AT levels)

    • Correlate variants with functional outcomes (lung function tests, CT-based emphysema assessment)

    • Consider smoking history and environmental exposures as modifiers

  • Variant classification workflow:

    • Sequence identification of novel variants

    • In silico prediction of functional impact

    • Functional characterization in cell models (as performed with the seven novel variants described in Spanish patients)

    • Clinical correlation with phenotype

  • Cell-based functional characterization methods:

    • Overexpression systems (e.g., in HEK293T cells)

    • Assessment of protein expression, polymerization, degradation, and secretion

    • Analysis of antielastase activity

    • Visualization techniques: periodic acid–Schiff staining, Western blotting

    • Protein trafficking studies: pulse-chase assays

This comprehensive approach has been successfully employed to characterize novel variants such as PiSDonosti (S+Ser14Phe), PiTijarafe (Ile50Asn), and others identified in Spanish patients with A1AT deficiency .

How do epigenetic modifications influence SERPINA1 expression and what methodologies can detect these changes?

Epigenetic modifications, particularly DNA methylation, play a significant role in regulating SERPINA1 expression with important implications for lung function and disease:

Key methylation findings:

  • Array-based methylation analysis of 27,578 CpG sites identified two sites in SERPINA1 (cg02181506 and cg24621042) as the highest-ranking methylation marks associated with COPD .

  • These findings were consistent across multiple cohorts and independent of COPD severity and smoking exposure .

  • Approximately 4,800 CpG sites showed significant associations with lung function parameters (FEV1/FVC ratio and FEV1) .

Methodological approaches for detecting SERPINA1 methylation:

  • Genome-wide methylation screening:

    • Illumina Methylation arrays (enables analysis of thousands of CpG sites)

    • Whole-genome bisulfite sequencing (for comprehensive methylation landscape)

  • Targeted methylation analysis:

    • Bisulfite pyrosequencing for specific CpG sites (e.g., cg02181506)

    • Methylation-specific PCR for regions of interest

    • Next-generation sequencing of bisulfite-converted DNA

  • Integrated multi-omics approach:

    • Correlate methylation data with:

      • Gene expression (RNA-seq, qPCR)

      • Protein levels (immunoassays)

      • Lung function parameters

      • Clinical outcomes

  • Longitudinal study design:

    • Beckmeyer-Borowko et al. demonstrated the value of examining methylation as a determinant of lung function decline over 10-15 years in tobacco smoke-exposed populations .

    • This approach requires methylation analysis of the SERPINA gene cluster (covering 12 genes: PPP4R4, SERPINA10, SERPINA6, SERPINA1, SERPINA11, SERPINA9, SERPINA12, SERPINA4, SERPINA5, SERPINA3, SERPINA13, and GSC) .

These methodologies have helped establish that SERPINA1 methylation status varies between smokers and non-smokers, though with some conflicting results across studies that require further investigation .

What are the current challenges and best practices in characterizing novel SERPINA1 variants?

Characterizing novel SERPINA1 variants presents several challenges that require systematic approaches:

Current challenges:

  • High polymorphism (>100 variants described)

  • Variants of uncertain significance requiring functional validation

  • Variable penetrance and expressivity

  • Complex genotype-phenotype correlations

  • Interactions with environmental factors (particularly smoking)

Best practices for variant characterization workflow:

  • Discovery phase:

    • Complete gene sequencing (not just common variants)

    • Population screening in diverse ethnic groups

    • Family studies to track inheritance patterns

  • Bioinformatic analysis:

    • In silico prediction tools

    • Structural modeling

    • Conservation analysis across species

    • Population frequency assessment

  • Functional characterization in cell models:

    • Overexpression systems (such as HEK293T cells)

    • Analysis of:

      • Intracellular polymer formation

      • Protein secretion

      • Protein degradation patterns

      • Antielastase activity

  • Specific experimental methods:

    • Periodic acid–Schiff staining for polymer detection

    • Western blotting for expression and secretion

    • Pulse-chase assays for trafficking defects

    • Elastase inhibition assays for functional assessment

  • Clinical correlation:

    • Measure serum levels in variant carriers

    • Assess lung function parameters

    • Evaluate for emphysema using CT imaging

    • Document liver involvement

This comprehensive approach has successfully characterized seven novel variants in Spanish patients: PiSDonosti (S+Ser14Phe), PiTijarafe (Ile50Asn), PiSevilla (Ala58Asp), PiCadiz (Glu151Lys), PiTarragona (Phe227Cys), PiPuerto Real (Thr249Ala), and PiValencia (Lys328Glu) . These variants exhibited different molecular behaviors, with five forming intracellular polymers and failing to secrete protein, while two (E151K and K328E) secreted protein but showed other functional abnormalities .

How can researchers distinguish between the effects of different SERPINA1 isoforms in experimental systems?

Distinguishing between SERPINA1 isoforms presents methodological challenges due to their identical protein-coding sequences but different 5'-UTR regions. Here are recommended approaches:

Isoform identification and quantification:

  • Transcript-specific RT-PCR:

    • Design primers targeting unique 5'-UTR sequences of each isoform

    • Use quantitative RT-PCR to measure relative abundance

    • Digital droplet PCR for absolute quantification

  • RNA-Seq with specialized analysis:

    • Long-read sequencing technologies (PacBio, Oxford Nanopore)

    • Bioinformatic pipelines designed to distinguish isoforms (e.g., Kallisto, RSEM)

    • Junction analysis to identify splice variants

  • Isoform-specific reporter constructs:

    • Clone individual 5'-UTRs into reporter gene constructs

    • Assess relative translation efficiency

    • Evaluate the impact of RNA structure on translation

Functional analysis of isoform-specific effects:

  • Structure-function studies:

    • RNA structure prediction software to analyze 5'-UTR folding

    • SHAPE (Selective 2′-hydroxyl acylation analyzed by primer extension) analysis to experimentally validate RNA structures

    • Ribosome profiling to assess translation efficiency of different isoforms

  • Experimental manipulation:

    • CRISPR-based approaches to selectively modify specific isoforms

    • Antisense oligonucleotides to block specific splice variants

    • Overexpression of individual isoforms to assess differential effects

  • Context-dependent expression analysis:

    • Tissue-specific expression patterns (e.g., NM_000295.4 represents 23% of total SERPINA1 mRNA in lung)

    • Response to inflammatory stimuli

    • Changes during development or disease progression

This approach is particularly important given that the NM_000295.4 isoform lacks upstream open reading frames (uORFs) and is likely one of the most efficiently translated SERPINA1 mRNAs, making it an important model system for understanding post-transcriptional regulation .

What is the current understanding of SERPINA1's role in metabolic regulation and how can researchers study these functions?

Recent research has revealed unexpected roles for SERPINA1 in metabolic regulation beyond its classic function as a protease inhibitor:

SERPINA1's metabolic functions:

  • Adipocyte regulation:

    • Promotes proliferation of white and brown preadipocytes

    • Increases expression of uncoupling protein 1 (UCP1)

    • Enhances mitochondrial activation in mature adipocytes

  • Energy metabolism effects:

    • Liver-specific SERPINA1 transgenic mice show increased browning of adipose tissues

    • Enhanced energy expenditure

    • Reduced adiposity

    • Improved glucose tolerance

  • Molecular mechanism:

    • Forms a complex with Eph receptor B2

    • Regulates downstream signaling in adipocytes

Methodological approaches to study metabolic functions:

  • Animal models:

    • Liver-specific SERPINA1 transgenic mice

    • SERPINA1 knockout mice

    • Diet-induced obesity models to assess metabolic protection

  • Metabolic phenotyping:

    • Indirect calorimetry to measure energy expenditure

    • Glucose tolerance tests

    • Insulin sensitivity assays

    • Body composition analysis

  • Adipose tissue analysis:

    • Histological assessment of white and brown adipose tissues

    • UCP1 expression quantification (qPCR, Western blot)

    • Mitochondrial function assays (oxygen consumption, ATP production)

    • Thermogenesis assessment (cold challenge experiments)

  • Mechanistic studies:

    • Co-immunoprecipitation to detect SERPINA1-EphB2 complex formation

    • Signaling pathway analysis (Western blot, phosphorylation assays)

    • Cell-based assays for adipocyte differentiation and function

  • Translational approaches:

    • Correlate circulating SERPINA1 levels with metabolic parameters in humans

    • Assess metabolic phenotypes in patients with SERPINA1 variants

    • Investigate therapeutic potential for obesity and metabolic syndrome

These findings highlight SERPINA1 as "an important hepatokine that improves obesity, energy expenditure and glucose metabolism" with therapeutic potential for metabolic disorders .

What are the recommended methods for producing and handling recombinant SERPINA1 for functional studies?

Producing high-quality recombinant SERPINA1 protein is essential for functional studies. Based on established protocols, researchers should consider the following:

Production systems:

  • Rice grain expression system (Oryza Sativa):

    • Produces non-glycosylated single polypeptide chain SERPINA1

    • Molecular mass of approximately 43.1 kDa

    • Purified through proprietary chromatographic techniques

  • Mammalian expression systems:

    • HEK293T cells commonly used for expression of wild-type and variant forms

    • Enable post-translational modifications similar to native protein

    • Useful for studying variants and their trafficking

Handling and storage recommendations:

  • Reconstitution protocol:

    • Reconstitute lyophilized SERPINA1 in sterile 18M-cm H2O

    • Maintain concentration not less than 100μg/ml

    • Can be further diluted in other aqueous solutions as needed

  • Storage conditions:

    • Store lyophilized protein desiccated below -18°C (stable at room temperature for up to 3 weeks)

    • Store reconstituted protein at 4°C for short-term use (2-7 days)

    • For long-term storage, keep below -18°C

    • Add carrier protein (0.1% HSA or BSA) for enhanced stability

    • Avoid freeze-thaw cycles

  • Quality control measures:

    • Verify protein purity by SDS-PAGE

    • Confirm identity by mass spectrometry

    • Validate functional activity through elastase inhibition assays

    • Assess polymerization tendency using native PAGE

These technical considerations are crucial for maintaining protein activity and ensuring reliable experimental results when working with recombinant SERPINA1.

What are the most sensitive and specific methods for assessing SERPINA1 genetic variants in large-scale studies?

For large-scale studies investigating SERPINA1 genetic variants, researchers should consider these methodological approaches:

Screening and discovery methods:

  • Next-generation sequencing approaches:

    • Targeted sequencing of 16.9 kB of SERPINA1 gene region

    • Whole exome sequencing with focused analysis of SERPINA1

    • Capture-based enrichment strategies for high-throughput screening

  • Variant detection in diverse populations:

    • Include multiple ethnic groups (as in studies with 1,693 non-Hispanic whites, 385 African Americans, and 90 Hispanics)

    • Focus on populations with significant exposure to risk factors (e.g., ≥20 pack-years smoking)

    • Consider family-based designs for rare variant discovery

Analytical approaches:

  • Integrated phenotyping:

    • Combine genetic data with:

      • Alpha-1 antitrypsin serum concentrations

      • Lung function parameters (FEV1, FEV1/FVC, FEF25-75%)

      • CT-based emphysema quantification

      • Clinical characteristics

  • Variant classification strategy:

    • Define rare variants (allele frequency < 0.05)

    • Classify variants by predicted effect (PI Z, S, and additional rare variants)

    • Group variants by functional impact for statistical power

    • Assess compound heterozygotes separately (e.g., ZS/ZVR)

  • Statistical approaches for rare variant analysis:

    • Burden tests

    • Sequence kernel association tests

    • Mixed models adjusting for covariates

    • Meta-analysis techniques for combining cohorts

This comprehensive approach has enabled the identification of significant associations between PI Z heterozygotes and lower post-bronchodilator FEV1, FEV1/FVC, and greater emphysema compared to individuals without rare variants. Additionally, it has revealed the effects of compound heterozygotes and the impact of multiple SERPINA1 variants on alpha-1 antitrypsin deficiency, lung function, and emphysema .

How can researchers effectively study the interaction between SERPINA1 genetic variants and environmental factors?

Studying gene-environment interactions for SERPINA1 requires specialized methodological approaches:

Study design considerations:

  • Cohort stratification:

    • Stratify by smoking history (pack-years)

    • Account for occupational exposures

    • Consider air pollution exposure levels

    • Categorize by different SERPINA1 variant types

  • Statistical approaches:

    • Interaction term analysis in regression models

    • Case-only designs for interaction assessment

    • Propensity score matching to control for confounding

    • Sensitivity analysis for threshold effects

  • Longitudinal assessment:

    • Track lung function decline over time (10-15 years)

    • Relate to cumulative environmental exposures

    • Assess time-varying covariates

    • Determine critical exposure windows

Molecular approaches to detect interactions:

  • Epigenetic analysis:

    • Assess methylation changes in response to environmental exposures

    • Compare methylation patterns across genotypes

    • Integrate with gene expression data

    • Focus on key CpG sites (e.g., cg02181506)

  • Functional validation:

    • In vitro exposure models using patient-derived cells

    • Organoid systems to model tissue-specific effects

    • Controlled exposure experiments in animal models

    • Proteomics to detect post-translational modifications

  • Biomarker development:

    • Identify markers of environmental damage

    • Correlate with SERPINA1 levels and function

    • Develop panels for early detection of susceptibility

    • Validate across different populations

This integrated approach is particularly important given that environmental factors, especially cigarette smoking, significantly modify the clinical presentation and progression of SERPINA1-related diseases .

How is SERPINA1 research contributing to our understanding of personalized medicine approaches for lung and liver diseases?

SERPINA1 research is advancing personalized medicine in several key ways:

Precision diagnostics:

  • Comprehensive variant profiling:

    • Beyond traditional PIZ and PIS testing

    • Deep sequencing reveals clinically significant rare variants

    • 5' UTR variants can affect translation efficiency and disease risk

  • Risk stratification:

    • Different genotypes confer variable disease risks:

      • PI Z heterozygotes show lower FEV1, FEV1/FVC, and greater emphysema

      • Compound heterozygotes (ZS/ZVR) demonstrate significant lung function impairment

      • Novel variants like those found in Spanish patients have distinct functional impacts

  • Integrated biomarker approach:

    • Combining:

      • Genetic variant profiles

      • Alpha-1 antitrypsin serum levels

      • Methylation patterns at key CpG sites

      • Functional assays (elastase inhibition capacity)

Therapeutic implications:

  • Variant-specific treatment approaches:

    • Polymerization inhibitors for variants that form intracellular polymers

    • RNA-targeted therapies for variants affecting splicing or translation

    • Augmentation therapy optimization based on specific deficiencies

  • Environmental modification strategies:

    • Personalized recommendations for exposure avoidance

    • Tailored monitoring schedules based on genetic risk

    • Smoking cessation programs designed for specific genetic backgrounds

  • Emerging therapeutic directions:

    • Enhancing autophagy for variants causing accumulation

    • Chaperone therapies to assist protein folding

    • Gene editing approaches for correction of specific mutations

    • Targeting RNA structures to enhance translation of functional protein

This personalized approach recognizes that "significant differences were observed within matched populations of severe AATD patients regarding risk of complications, age at onset, and disease course, including the dynamics of lung function decline" , necessitating individualized clinical management based on comprehensive genetic and functional assessments.

What novel roles for SERPINA1 beyond lung and liver disease are emerging from recent research?

Recent research has uncovered unexpected functions of SERPINA1 beyond its classical role in lung and liver protection:

Metabolic regulation:

  • Adipose tissue effects:

    • Functions as a hepatokine affecting adipocyte biology

    • Promotes proliferation of white and brown preadipocytes

    • Increases UCP1 expression and mitochondrial activation

    • Enhances energy expenditure and improves glucose tolerance

  • Molecular mechanisms:

    • Forms complex with Eph receptor B2

    • Regulates downstream signaling pathways in adipocytes

    • Promotes browning of white adipose tissue

    • May represent a liver-adipose tissue communication axis

  • Phenotypic consequences:

    • SERPINA1 transgenic mice show reduced adiposity

    • SERPINA1 knockout mice exhibit:

      • Decreased adipocyte mitochondrial function

      • Impaired thermogenesis

      • Obesity development

      • Systemic insulin resistance

Other emerging functions:

  • Inflammation modulation:

    • Beyond simple protease inhibition

    • Potential immunomodulatory effects

    • Interaction with inflammatory signaling pathways

  • Cancer biology connections:

    • Altered expression in certain cancer types

    • Potential role in tumor microenvironment

    • Interaction with matrix remodeling processes

  • Vascular biology:

    • Protection against elastase-mediated vascular damage

    • Potential role in atherosclerosis development

    • Interaction with endothelial function

These discoveries open new research directions and potential therapeutic applications for SERPINA1 beyond traditional alpha-1 antitrypsin deficiency treatment, particularly in metabolic diseases like obesity and type 2 diabetes .

How can integrated multi-omics approaches enhance our understanding of SERPINA1 biology and pathophysiology?

Integrated multi-omics approaches provide powerful tools for comprehensively understanding SERPINA1 biology:

Multi-omics integration strategies:

  • Genomic-transcriptomic-proteomic integration:

    • Correlate SERPINA1 genetic variants with:

      • Transcript isoform expression (11 different mRNA variants)

      • RNA structural variations

      • Protein levels and functional activity

      • Clinical phenotypes

  • Epigenomic-transcriptomic analysis:

    • Link methylation patterns to expression levels

    • Assess impact of smoking on methylation status

    • Identify regulatory networks controlling SERPINA1 expression

    • Map tissue-specific epigenetic landscapes

  • Functional genomics approaches:

    • CRISPR-based screens to identify regulators

    • RNA structure mapping techniques

    • Ribosome profiling to assess translation efficiency

    • Interactome studies to identify protein partners (e.g., EphB2)

Data integration frameworks:

  • Clinical-molecular data integration:

    • Connect genetic and molecular data with:

      • Lung function parameters

      • CT-based emphysema quantification

      • Metabolic measurements

      • Longitudinal disease progression

  • Systems biology modeling:

    • Network analysis of SERPINA gene cluster interactions

    • Pathway enrichment to identify biological processes

    • Machine learning approaches for predictive modeling

    • Causal inference methods for mechanism discovery

  • Visualization and analysis tools:

    • Multi-omics data visualization platforms

    • Integrative clustering approaches

    • Knowledge graphs connecting molecular and clinical data

    • Interactive dashboards for hypothesis generation

This integrated approach could help resolve current research gaps, such as conflicting results regarding SERPINA1 hypomethylation in smokers and COPD patients, and better characterize the complex effects of rare variants on disease risk and progression .

What are the most important quality control measures when working with SERPINA1 in experimental systems?

Ensuring experimental rigor when working with SERPINA1 requires attention to several quality control measures:

Protein quality control:

  • Recombinant protein validation:

    • Verify molecular mass (expected 43.1 kDa)

    • Confirm purity through chromatographic techniques

    • Assess functional activity through elastase inhibition assays

    • Check for aggregation/polymerization tendency

  • Storage and handling:

    • Maintain proper storage conditions (desiccated below -18°C for lyophilized protein)

    • Use appropriate reconstitution protocols (sterile 18M-cm H2O)

    • Add carrier protein (0.1% HSA or BSA) for long-term storage

    • Avoid freeze-thaw cycles

  • Functional integrity assessment:

    • Regular testing of protease inhibitory activity

    • Monitoring for conformational changes

    • Verifying cellular uptake and trafficking

    • Assessing polymerization status

Genetic and expression system controls:

  • Cell model validation:

    • Verify expression vector sequence integrity

    • Confirm cellular expression through immunoblotting

    • Assess proper cellular localization through imaging

    • Monitor secretion rates through pulse-chase experiments

  • Variant characterization controls:

    • Include wild-type controls in all experiments

    • Use characterized variants as positive controls

    • Implement appropriate negative controls

    • Consider allelic dosage effects in heterozygous models

  • Expression analysis standardization:

    • Use validated housekeeping genes for normalization

    • Include isoform-specific controls

    • Account for tissue-specific expression patterns

    • Consider environmental influences on expression

These quality control measures are essential for generating reliable and reproducible data when investigating SERPINA1 biology and pathology, especially when characterizing novel variants or studying complex disease mechanisms .

What are the key considerations when designing studies to investigate rare SERPINA1 variants in diverse populations?

Designing studies to investigate rare SERPINA1 variants across diverse populations requires careful methodological planning:

Study design considerations:

  • Population selection and characterization:

    • Include multiple ethnic groups (e.g., non-Hispanic white, African American, Hispanic)

    • Consider populations with varying exposure profiles

    • Account for genetic admixture and population structure

    • Implement matched controls from the same populations

  • Sample size and power calculations:

    • Adjust for the rarity of variants

    • Consider variant aggregation approaches

    • Plan for subgroup analyses (e.g., by smoking status)

    • Account for varying effect sizes across populations

  • Phenotyping strategy:

    • Standardize lung function testing

    • Implement consistent CT imaging protocols

    • Measure serum alpha-1 antitrypsin levels

    • Document environmental exposures comprehensively

Analytical approaches:

  • Variant detection methodology:

    • Deep resequencing of the entire 16.9 kB SERPINA1 region

    • Coverage requirements for reliable rare variant detection

    • Validation of novel variants through orthogonal methods

    • Functional prediction of variant effects

  • Statistical analysis plan:

    • Rare variant association methods

    • Population-specific variant frequency adjustments

    • Methods for detecting gene-environment interactions

    • Multiple testing correction strategies

  • Functional validation pathway:

    • Prioritization strategy for functional studies

    • Cell-based assays appropriate for population-specific variants

    • Consideration of genetic background effects

    • Translation to clinical implications

This approach has successfully identified population-specific variants with significant clinical implications, such as a 5′ untranslated region insertion (rs568223361) in African Americans associated with lower alpha-1 antitrypsin levels and functional small airway disease .

Product Science Overview

Structure and Function

AAT is a glycoprotein primarily produced in the liver and released into the bloodstream. It belongs to the serpin (serine protease inhibitor) superfamily, which includes proteins that inhibit proteases like trypsin, cathepsin G, thrombin, and tissue kallikrein . The primary function of AAT is to protect the lungs from neutrophil elastase, an enzyme that can degrade elastin and other structural proteins in the lung tissue .

Alpha-1 Antitrypsin Deficiency (AATD)

AAT deficiency (AATD) is a genetic disorder characterized by low levels of AAT in the blood. This deficiency allows neutrophil elastase to destroy alveolar walls, leading to chronic obstructive pulmonary disease (COPD) and emphysema . Approximately 1-2% of COPD cases are attributed to AATD .

Recombinant Alpha-1 Antitrypsin

Recombinant Alpha-1 Antitrypsin (rAAT) is produced using yeast or other expression systems to create a biologically active form of AAT that is structurally similar to the natural protein found in human blood . This recombinant form eliminates the risk of blood-borne infectious agents associated with plasma-derived AAT and allows for increased manufacturing efficiency .

Therapeutic Applications

Recombinant AAT is used in augmentation therapy for individuals with AATD. This therapy involves regular infusions of AAT to maintain protective levels in the blood and lungs, thereby reducing the risk of lung damage . Studies have shown that recombinant AAT can effectively prevent or attenuate elastase- and cigarette smoke-induced models of emphysema, providing greater protection against alveolar enlargement, lung dysfunction, and airway inflammation compared to plasma-derived AAT .

Production and Optimization

The production of recombinant AAT involves optimizing the expression system to maximize yield. For instance, using the methylotrophic yeast Pichia pastoris as a host has proven effective for producing and secreting biologically active AAT . Strategies such as optimizing codon usage, using effective signal sequences, and adjusting gene dosage have been employed to enhance the production levels of recombinant AAT .

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.