SAA1 Monkey

Serum Amyloid A (APO-SAA1) Rhesus Macaque Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Production

SAA1 Monkey is a non-glycosylated, homodimeric protein with a molecular weight of 11.8 kDa and 104 amino acids . It is synthesized in E. coli and purified via chromatographic techniques. Key features include:

  • Sequence: Shares high homology with human SAA1 but exhibits species-specific variations (e.g., amino acid substitutions at positions 4, 10, and 23 compared to human SAA1) .

  • Biological Activity: Functions as a chemoattractant for human monocytes at concentrations of 1.0–10.0 ng/ml .

  • Storage: Lyophilized powder stored at -20°C; reconstituted solutions require carrier proteins (e.g., BSA) to maintain stability .

2.1. Primary and Tertiary Structure

  • Primary Sequence: The N-terminal region (1–25 amino acids) has a moderate aggregation score (238) compared to human SAA1 (376), as predicted by the TANGO algorithm .

  • Tertiary Structure: Native SAA1 adopts a hexameric 4-helix bundle conformation, with amyloidogenic regions (e.g., N-terminal helices 1 and 3) prone to misfolding under inflammatory conditions .

Table 1: Aggregation Scores of SAA1 1–25 Peptide Across Species

SpeciesAggregation Score (TANGO)
Human376
Rhesus Monkey238
Donkey448
Common Bottlenose Dolphin3
Alpine Ibex11

Data adapted from

3.1. Inflammatory Response

SAA1 Monkey mimics human SAA1 in modulating immune cell activity:

  • Chemotaxis: Binds to formyl peptide receptor 2 (FPR2) to recruit neutrophils and macrophages .

  • Amyloidosis: Under chronic inflammation, SAA1 undergoes proteolytic cleavage, generating amyloidogenic fragments that aggregate into β-sheet-rich fibrils .

3.2. HDL Binding and Lipid Metabolism

SAA1 displaces apolipoprotein A-I (ApoA-I) from high-density lipoprotein (HDL), altering lipid transport and immune function . In rhesus macaques, this mechanism is linked to systemic amyloidosis risk .

4.1. Amyloidogenic Potential

  • Fibril Formation: Rhesus monkey SAA1 1–25 peptide forms fibrils in vitro, as confirmed by Thioflavin T (ThT) fluorescence and transmission electron microscopy (TEM) .

  • Seeding Capacity: Misfolded SAA1 signal peptides can seed amyloid formation in adjacent regions (e.g., residues 1–25 and 32–47) .

Table 2: ThT Fluorescence Intensity of SAA1 1–25 Peptides

SpeciesThT Fluorescence (Relative Units)
HumanHigh
Rhesus MonkeyModerate
DonkeyVery High
Common Bottlenose DolphinLow

Data adapted from

4.2. Immune Modulation

  • Allergic Responses: SAA1 knockout in mice reduces eosinophilia and Th2 cytokine production, suggesting a role in type 2 immunity .

  • Cancer Immunotherapy: Elevated SAA levels correlate with PD-1 resistance in hepatocellular carcinoma (HCC), potentially via neutrophil PD-L1 upregulation .

5.1. Disease Modeling

Rhesus macaques are used to study AA amyloidosis, a condition driven by SAA1 misfolding. Circulating SAA1 levels are monitored via species-specific ELISA assays to assess disease progression .

5.2. Therapeutic Targeting

  • Glycolysis Inhibition: Inhibiting SAA1-induced glycolysis in neutrophils reduces PD-L1 expression, enhancing anti-PD-1 efficacy in HCC models .

  • Amyloid Inhibition: Blocking SAA1 aggregation or HDL displacement may mitigate amyloidosis .

Handling and Stability

ParameterSpecification
Reconstitution≥100 µg/ml in sterile ddH₂O
Storage-20°C (lyophilized), 4°C (reconstituted)
Purity≥97% (SDS-PAGE/HPLC)
Carrier Protein0.1% BSA/HSA recommended

Data from

Product Specs

Introduction
Serum amyloid A1 (SAA1) is an acute-phase protein primarily produced by the liver in response to inflammation. Its levels can rise dramatically during inflammatory events. SAA1 is believed to play a role in immune modulation, including tissue repair, antimicrobial activity, and directing immune cells to infection sites. However, persistently elevated SAA1 levels are associated with the development of secondary amyloidosis, a condition characterized by the accumulation of amyloid fibrils in various tissues and organs. This accumulation, particularly of the SAA1 cleavage product amyloid protein A, can lead to organ damage. Secondary amyloidosis is often linked to chronic inflammatory diseases like rheumatoid arthritis.
Description
This product consists of recombinant SAA1 protein derived from monkey and produced in E. coli. It is a single, non-glycosylated polypeptide chain with 104 amino acids, resulting in a molecular mass of 11.8 kDa. The protein undergoes purification using proprietary chromatographic methods.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The protein was lyophilized with 1x PBS at a pH of 7.4.
Solubility
To reconstitute the lyophilized SAA1, it is recommended to dissolve it in sterile 18 megaohm-cm H2O at a concentration of at least 100 micrograms/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized SAA1 remains stable at room temperature for up to 3 weeks. However, for long-term storage, it should be stored desiccated at a temperature below -18 degrees Celsius. After reconstitution, SAA1 can be stored at 4 degrees Celsius for 2-7 days. For extended storage, it is advised to store it at a temperature below -18 degrees Celsius. The addition of a carrier protein like HSA or BSA at a concentration of 0.1% is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of this product is greater than 97.0%, as determined by two methods: Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) and SDS-PAGE analysis.
Biological Activity
The biological activity of this product is assessed based on its ability to chemoattract human monocytes. This chemoattractant activity is evaluated within a concentration range of 1.0 to 10.0 ng/ml.
Synonyms
Serum amyloid A protein, SAA, Amyloid protein A, Amyloid fibril protein AA, SAA1, SAA2, PIG4, TP53I4, MGC111216.
Source
Escherichia Coli.
Amino Acid Sequence
RSWFSFLGEA YDGARDMWRA YSDMKEANYK NSDKYFHARG NYDAAQRGPG GVWAAEVISD ARENIQKLLG RGAEDTLADQ AANEWGRSGK DPNHFRPAGL PEKY.

Q&A

What is SAA1 and what is its significance in primate research?

Serum amyloid A1 (SAA1) is a major acute-phase protein that exists as a minor apolipoprotein of normal high-density lipoprotein (HDL) particles. During inflammatory processes, SAA1 levels increase dramatically, and it becomes a major component of HDL. In primate research, SAA1 is significant because it serves as a marker of inflammation and plays a role in lipoprotein metabolism and potentially in amyloidosis development. Studying SAA1 in non-human primates, particularly in rhesus monkeys (Macaca mulatta), provides valuable insights into inflammatory processes that may be more directly applicable to human conditions than rodent models .

How does the structure of monkey SAA1 compare to human SAA1?

Monkey SAA1, specifically from Macaca mulatta (rhesus monkey), shares significant structural similarities with human SAA1 but contains notable differences in the amino acid sequence. The 1-25 amino acid region, which is crucial for aggregation properties, shows several point mutations compared to the human sequence. While human SAA1 has the sequence starting with RSFFSFLE, rhesus monkey SAA1 begins with RSWFSFLE, with a key difference at position 3 (F→W). This results in a different aggregation score (238 for rhesus monkey vs. 376 for human) as calculated by the TANGO algorithm . These differences in sequence may explain variations in amyloidogenic potential between species.

What biological samples can be used to detect monkey SAA1?

Monkey SAA1 can be detected and quantified in multiple biological samples including serum, plasma, and tissue homogenates. The choice of sample depends on the specific research question. Serum and plasma are commonly used for monitoring systemic SAA1 levels, while tissue homogenates are valuable for studying localized expression and accumulation. For accurate quantification, enzyme-linked immunosorbent assay (ELISA) methods with a detection range of 0.625-40 ng/ml and sensitivity down to 0.156 ng/ml are available . When collecting samples, proper handling and storage protocols should be followed to preserve SAA1 integrity.

How should researchers design studies to investigate SAA1 expression during acute and chronic inflammation in monkeys?

When designing studies to investigate SAA1 expression patterns in monkeys during inflammation, researchers should consider both temporal dynamics and inflammatory stimuli specificity. For acute inflammation models, time course studies with sampling at 0, 6, 12, 24, 48, and 72 hours post-stimulus are recommended to capture the rapid induction and resolution phases. Multiple inflammatory stimuli should be compared (e.g., LPS, IL-1β, TNF-α) to determine stimulus-specific responses. For chronic inflammation, longitudinal sampling over weeks or months is essential to monitor persistent elevation patterns.

Experimental groups should include:

  • Control groups (vehicle-treated)

  • Acute inflammation groups (single stimulus)

  • Chronic inflammation groups (repeated stimulus or persistent inflammatory condition)

  • Recovery groups (post-inflammation resolution)

Measurement methods should incorporate both protein quantification via ELISA and mRNA expression analysis through qRT-PCR. Correlation with other inflammatory markers and clinical parameters will provide context for SAA1 expression patterns.

What are the optimal methods for detecting and quantifying SAA1 in monkey samples?

The optimal methods for detecting and quantifying SAA1 in monkey samples depend on the research objective. For precise quantification, competitive inhibition enzyme immunoassay techniques such as ELISA are recommended. These assays utilize pre-coated plates with SAA1 and horseradish peroxidase (HRP) conjugated antibodies specific for SAA1. The competitive inhibition reaction between pre-coated SAA1 and SAA1 in samples allows for accurate quantification with sensitivity down to 0.156 ng/ml .

For visualization of SAA1 aggregates, multiple complementary techniques should be employed:

  • Congo Red binding assays for detecting amyloid-like fibrillar β-aggregates

  • Thioflavin T (ThT) fluorescence for monitoring fibril formation kinetics

  • Transmission electron microscopy (TEM) for direct visualization of fibrillar structures

When analyzing samples with potentially cross-reactive proteins, it's important to note that high-quality immunoassays demonstrate minimal cross-reactivity between monkey SAA1 and analogues . For comprehensive analysis, combining protein quantification with mRNA expression data provides insights into both transcriptional and translational regulation.

How can researchers induce and monitor SAA1-associated amyloidosis in primate models?

Inducing and monitoring SAA1-associated amyloidosis in primate models requires a methodical approach focusing on both induction protocols and comprehensive monitoring strategies. Based on available research, successful induction typically employs one of several approaches:

  • Chronic inflammatory stimulation using repeated subcutaneous injections of inflammatory agents (e.g., silver nitrate, casein) over a period of 4-6 weeks

  • Transmissible seeding using either oral administration or direct injection of preformed amyloid fibrils from either same-species or cross-species sources

  • Genetic modification approaches in cases where applicable

Monitoring should incorporate multiple techniques:

  • Regular serum SAA1 quantification via ELISA to track acute-phase response

  • Periodic tissue biopsies from common amyloid deposition sites (liver, spleen, kidney)

  • Histological examination with Congo Red and polarized light microscopy

  • Immunohistochemistry with anti-SAA1 antibodies

  • TEM examination for characteristic amyloid fibril ultrastructure

Researchers should establish baseline SAA1 levels prior to induction and correlate amyloid deposition with both peak and cumulative SAA1 exposure to understand the relationship between acute-phase response intensity and amyloidosis progression.

How does the aggregation propensity of monkey SAA1 compare to other species?

The aggregation propensity of monkey SAA1 (specifically from Rhesus monkeys, Macaca mulatta) positions it in a moderate range compared to other species. When examining the critical 1-25 amino acid region using the TANGO algorithm, rhesus monkey SAA1 demonstrates an aggregation score of 238, which is lower than human SAA1 (376) and significantly lower than donkey SAA1 (448), but higher than several other species including common bottlenose dolphin (3), alpine ibex (11), and chamois (11) .

This comparative data is summarized in the following table:

SpeciesAggregation Score (1-25 aa region)
Donkey (Equus asinus)448
Human (Homo sapiens)376
Rhesus Monkey (Macaca mulatta)238
Domestic ferret (Mustela putorius furo)26
Lesser-Egyptian jerboa (Jaculus jaculus)14
Alpine ibex (Capra ibex)11
Chamois (Rupicapra rupicapra)11
Common bottlenose dolphin (Tursiops truncatus)3

In experimental validation using Congo Red binding assays, Thioflavin T fluorescence, and transmission electron microscopy, rhesus monkey SAA1 demonstrates amyloid-forming capabilities consistent with its moderate aggregation score. This places monkey SAA1 in an interesting intermediate position that may provide insights into the structural determinants of amyloidogenicity .

What differences exist between acute and constitutive SAA expression in primates?

In primates, as in other mammals, there are distinct differences between acute and constitutive SAA expression patterns, regulation, and function. Acute-phase SAA (primarily SAA1) is dramatically upregulated during inflammation, while constitutive SAA (SAA4) maintains relatively stable expression levels regardless of inflammatory status.

Key differences include:

  • Expression patterns:

    • Acute SAA1: Low baseline levels in healthy states but can increase up to 1000-fold during acute inflammation

    • Constitutive SAA4: Maintains relatively stable expression levels with minimal fluctuation during inflammatory events

  • Function in lipoprotein metabolism:

    • Acute SAA1: Becomes a major apolipoprotein of HDL during inflammation, potentially altering HDL function

    • Constitutive SAA4: May play a role in HDL-VLDL interactions, as adenoviral expression of SAA4 resulted in increased HDL size and significant increases in VLDL levels (20-fold) and triglyceride levels (1.7-fold)

  • Impact on HDL properties:

    • Acute SAA1: Even at high expression levels comparable to a major acute-phase response (72±8 mg/dL), SAA1 alone does not significantly alter apoA-I levels but can lead to larger HDL particles (~10% increase)

    • Constitutive SAA4: Affects HDL size (~10% increase) and dramatically alters VLDL metabolism

These differences suggest that while acute SAA proteins alone may be insufficient to alter HDL cholesterol or apoA-I levels during inflammation, constitutive SAA4 may play a more significant role in lipoprotein metabolism than previously recognized .

How transferable are findings from monkey SAA1 research to human clinical applications?

Findings from monkey SAA1 research demonstrate considerable but incomplete transferability to human clinical applications. This transferability is based on several factors:

For optimal transferability of findings, researchers should:

  • Focus on comparative studies that directly examine both monkey and human SAA1 under identical experimental conditions

  • Validate key findings in human samples or human cell models before clinical application

  • Consider species-specific differences in SAA1 sequence, regulation, and function when interpreting results

  • Use multiple non-human primate species when possible to establish conserved mechanisms

What mechanisms drive the amyloidogenic potential of monkey SAA1 peptides?

The amyloidogenic potential of monkey SAA1 peptides is driven by complex molecular mechanisms centered around specific structural domains and environmental factors. In rhesus monkey SAA1, the 1-25 amino acid region demonstrates moderate aggregation propensity (aggregation score of 238) compared to human SAA1 (376) . Several key mechanisms contribute to this amyloidogenic potential:

  • Primary sequence determinants: The specific amino acid composition, particularly in the N-terminal region (1-25 aa), significantly influences aggregation. Point mutations at positions 3 (W instead of F in humans) and 14 (A in both species) affect beta-sheet formation propensity.

  • Secondary structure transitions: Under specific environmental conditions, SAA1 can transition from its native alpha-helical structure to beta-sheet conformations that promote fibril formation. This conformational change appears to be a critical step in amyloidogenesis.

  • Proteolytic processing: Limited proteolysis of full-length SAA1 (104 aa) generates fragments with enhanced amyloidogenic potential. The N-terminal fragments (including the 1-25 aa region) typically show the highest aggregation propensity as demonstrated by ThT fluorescence assays and TEM visualization .

  • Environmental factors: Conditions that destabilize the native protein structure promote amyloid formation. These include:

    • Acidic pH

    • Presence of glycosaminoglycans

    • Oxidative stress

    • Metal ions (particularly zinc and copper)

  • Seed-dependent nucleation: Pre-existing SAA1 amyloid fibrils can accelerate aggregation through a seeding mechanism, explaining the potential for transmission both within and between species .

Experimental evidence from Congo Red binding, Thioflavin T fluorescence kinetics, and TEM visualization confirms that monkey SAA1 peptides can form amyloid-like fibrils, although the kinetics and morphology may differ from human SAA1 fibrils .

How can researchers address data inconsistencies when studying SAA1 across different primate species?

When studying SAA1 across different primate species, researchers often encounter data inconsistencies that can complicate interpretation. Addressing these inconsistencies requires a systematic approach:

  • Standardization of measurement methods:

    • Use consistent antibody clones for immunoassays

    • Employ recombinant species-specific standards for quantification

    • Validate assays for cross-reactivity with SAA isoforms and species variants

    • Include internal controls to normalize between experiments

  • Genetic isoform identification:

    • Sequence SAA1 genes to identify species-specific isoforms

    • Consider polymorphisms that may affect antibody binding or function

    • Use genomic data to ensure targeting the correct ortholog

  • Experimental design considerations:

    • Control environmental variables that influence acute-phase responses

    • Standardize inflammatory stimuli dosing based on body weight

    • Account for circadian variation in SAA expression

    • Consider age, sex, and reproductive status as covariates

  • Statistical approaches for heterogeneous data:

    • Apply meta-analysis techniques for combining results across studies

    • Use random-effects models to account for inter-species variation

    • Implement Bayesian hierarchical models to incorporate prior knowledge

    • Consider outlier detection and handling procedures

  • Reporting standards:

    • Document detailed methodology including species, subspecies, and origin

    • Report raw data alongside normalized results

    • Provide confidence intervals rather than just point estimates

    • Clearly state limitations and potential sources of bias

By implementing these approaches, researchers can better interpret seemingly contradictory results and develop more robust cross-species models of SAA1 biology. The integration of multiple data types (genomic, proteomic, functional) can help resolve inconsistencies that might arise from focusing on a single aspect of SAA1 biology.

What role does SAA1 play in HDL remodeling during inflammation in primates?

SAA1 plays a nuanced role in HDL remodeling during inflammation in primates, with effects that appear to be concentration-dependent and potentially influenced by other acute-phase proteins. Research specifically examining this relationship provides several key insights:

These findings indicate that while SAA1 contributes to HDL remodeling during inflammation in primates, its effects are part of a more complex acute-phase response that likely requires additional inflammatory mediators to produce the characteristic lipoprotein changes observed during inflammation .

What are the latest developments in SAA1 detection techniques for primate research?

Recent methodological advances have significantly enhanced the precision, sensitivity, and applicability of SAA1 detection techniques in primate research. While traditional methods remain valuable, several innovations have expanded research capabilities:

  • Enhanced immunoassay platforms:

    • Competitive inhibition enzyme immunoassays have been optimized specifically for monkey SAA1 detection with sensitivity down to 0.156 ng/ml and a detection range of 0.625-40 ng/ml

    • Multiplex assays now allow simultaneous quantification of SAA1 alongside other acute-phase proteins and cytokines, providing more comprehensive inflammatory profiles

    • Single-molecule array (Simoa) technologies have pushed detection limits to the femtogram/ml range for ultra-sensitive applications

  • Advanced imaging techniques:

    • Super-resolution microscopy enables visualization of SAA1 distribution within cells and tissues at nanometer resolution

    • Label-free imaging methods including FTIR microscopy and Raman spectroscopy provide chemical specificity for amyloid detection without staining artifacts

    • Correlative light and electron microscopy (CLEM) allows researchers to connect functional data with ultrastructural information

  • Mass spectrometry applications:

    • Targeted proteomics approaches including multiple reaction monitoring (MRM) and parallel reaction monitoring (PRM) enable precise quantification of SAA1 isoforms and post-translational modifications

    • Top-down proteomics methods preserve intact protein information, revealing heterogeneity in SAA1 forms

    • Imaging mass spectrometry techniques visualize SAA1 distribution in tissues without antibody limitations

  • In vivo monitoring approaches:

    • Near-infrared fluorescence imaging with amyloid-specific probes allows longitudinal monitoring of amyloid deposition

    • PET imaging with radiolabeled tracers that bind amyloid provides whole-body assessment of amyloid burden

    • Minimally invasive sampling techniques including microdialysis enable continuous monitoring of SAA1 in extracellular fluids

  • Digital pathology and AI integration:

    • Machine learning algorithms applied to histopathology images can quantify amyloid deposits more objectively and detect subtle patterns

    • Digital image analysis platforms standardize Congo Red and immunohistochemistry interpretation

    • Deep learning approaches help integrate multi-modal data for comprehensive SAA1 behavior characterization

These methodological advances facilitate more comprehensive and nuanced studies of SAA1 biology in primates, enabling researchers to address increasingly sophisticated questions about SAA1's role in health and disease.

How can computational methods improve the prediction of SAA1 aggregation in different primate species?

Computational methods offer powerful approaches for predicting SAA1 aggregation in different primate species, enabling more targeted experimental designs and better understanding of species-specific amyloidogenic potential. Current and emerging computational strategies include:

  • Sequence-based prediction algorithms:

    • The TANGO algorithm has been successfully applied to predict aggregation propensity of SAA1 peptides from different species based on primary sequence features. For example, it has identified varying aggregation scores across species: human (376), rhesus monkey (238), and common bottlenose dolphin (3)

    • Additional algorithms including AGGRESCAN, ZipperDB, and AmylPred2 can be used in consensus approaches to improve prediction accuracy

    • Machine learning models trained on known amyloidogenic sequences can identify subtle patterns beyond traditional physicochemical properties

  • Structural modeling approaches:

    • Homology modeling using known SAA structures as templates can predict three-dimensional conformations of different primate SAA1 variants

    • Molecular dynamics simulations reveal conformational flexibility and aggregation-prone states under various conditions

    • Monte Carlo techniques can sample conformational space more efficiently for aggregation-prone regions

    • Coarse-grained simulations enable modeling of larger assemblies and longer timescales relevant to fibril formation

  • Protein-protein interaction predictions:

    • Docking simulations predict interactions between SAA1 monomers during oligomerization

    • Network analysis approaches identify key residues in fibril formation

    • Systems biology models incorporate SAA1 interactions with other molecules including glycosaminoglycans and lipids that influence aggregation

  • Integration with experimental data:

    • Bayesian modeling approaches can incorporate experimental measurements to refine predictions

    • Hybrid methods combining computational predictions with limited experimental data optimize research efficiency

    • Interactive platforms allow researchers to visualize predictions alongside experimental results for intuitive interpretation

  • Cross-species comparative approaches:

    • Phylogenetic analysis of SAA1 sequences across primates can identify conserved and divergent regions related to aggregation

    • Evolutionary models track changes in aggregation propensity throughout primate evolution

    • Ancestral sequence reconstruction techniques reveal how amyloidogenic potential has evolved

By applying these computational methods systematically across primate species, researchers can:

  • Identify key sequence determinants of species-specific aggregation differences

  • Predict which primate species may be more susceptible to amyloidosis

  • Design targeted mutations to test hypotheses about aggregation mechanisms

  • Develop species-specific inhibitors of pathological aggregation

These approaches significantly enhance research efficiency by prioritizing the most promising experimental directions based on computational predictions .

What are the best practices for designing cross-species SAA1 experiments?

Designing robust cross-species SAA1 experiments requires careful consideration of multiple factors to ensure meaningful comparisons and translatable results. Best practices include:

By adhering to these best practices, researchers can design cross-species SAA1 experiments that yield meaningful insights into conserved and divergent aspects of SAA1 biology with implications for human health and disease.

What are the most promising areas for future research on SAA1 in non-human primates?

Future research on SAA1 in non-human primates holds significant promise in several key areas that could advance both basic understanding and translational applications:

  • Structural determinants of species-specific aggregation:

    • Detailed structural studies comparing the three-dimensional conformations of SAA1 across primate species

    • Investigation of how specific amino acid differences, particularly in the 1-25 region, influence aggregation propensity

    • Examination of post-translational modifications that may differ between species and affect amyloidogenic potential

  • Cross-species transmission mechanisms:

    • Further exploration of the mechanisms underlying the transmission of amyloidosis between species

    • Investigation of species barriers and factors that promote or inhibit cross-species transmission

    • Development of primate models for studying transmissible amyloidosis relevant to human health

  • Functional proteomics of SAA1 variants:

    • Comprehensive characterization of the interactome of SAA1 across primate species

    • Investigation of species-specific differences in SAA1 interactions with HDL and other lipoproteins

    • Examination of how SAA1 variants differentially modulate immune responses

  • Advanced in vivo imaging:

    • Development of non-invasive imaging methods to monitor SAA1 expression and amyloid deposition in living primates

    • Longitudinal studies tracking the progression from acute inflammation to chronic amyloidosis

    • Correlation of imaging biomarkers with clinical manifestations

  • Therapeutic target validation:

    • Evaluation of SAA1 as a therapeutic target in primate models of inflammatory diseases

    • Testing of aggregation inhibitors in species with different baseline aggregation propensities

    • Exploration of species-specific responses to potential amyloidosis treatments

  • Systems biology approaches:

    • Integration of transcriptomic, proteomic, and metabolomic data to understand species-specific SAA1 regulation

    • Network analysis of inflammatory pathways involving SAA1 across primates

    • Modeling of evolutionary conservation and divergence in SAA1 function

  • Environmental and dietary influences:

    • Investigation of how environmental factors and diet affect SAA1 expression and function across primate species

    • Examination of the gut microbiome's influence on SAA1-related inflammation and amyloidosis

    • Study of natural compounds that may modulate SAA1 expression or aggregation

These research directions hold potential to significantly advance our understanding of SAA1 biology and pathology while providing valuable insights for human health applications, particularly in inflammatory conditions and amyloidosis.

How can SAA1 research in primates contribute to understanding human amyloidosis?

SAA1 research in primates offers unique and valuable contributions to understanding human amyloidosis through several interconnected pathways:

  • Evolutionary insights into amyloidogenic potential:

    • Comparative studies of SAA1 across primates reveal how amyloidogenic potential has evolved

    • Analysis of the rhesus monkey SAA1 sequence, with an aggregation score of 238 compared to human's 376, helps identify critical residues that modulate aggregation propensity

    • Mapping the evolutionary changes in SAA1 structure against species susceptibility to amyloidosis provides insights into natural protective mechanisms

  • Translational model advantages:

    • Non-human primates provide more faithful models of human amyloidosis than rodents due to closer evolutionary relationships

    • Primate models better recapitulate the complexity of human inflammatory responses and lipoprotein metabolism

    • The intermediate aggregation propensity of some primate SAA1 proteins offers a unique window into the threshold between normal function and pathological aggregation

  • Mechanistic understanding of transmission risk:

    • Studies demonstrating cross-species transmission of amyloidosis in primates inform risk assessment for potential human exposure

    • Investigation of species barriers and transmission efficiency helps identify protective factors that could be therapeutically mimicked

    • Primate models enable assessment of environmental and dietary factors that may influence transmission

  • Predictive biomarker development:

    • Longitudinal studies in primates can identify early biomarkers that predict progression from inflammation to amyloidosis

    • Multi-omics approaches in primates can reveal signatures associated with amyloidosis susceptibility or resistance

    • Correlation of SAA1 variants with disease manifestations across primates helps prioritize human variants for clinical attention

  • Therapeutic strategy evaluation:

    • Primate models allow testing of interventions targeting various stages of amyloidosis pathogenesis

    • Comparative efficacy of treatments across primates with different SAA1 aggregation propensities informs personalized approaches

    • Safety and efficacy data from non-human primates provides stronger translational value for human applications

  • Insights into comorbid conditions:

    • Studying the interaction between SAA1 and lipoproteins in primates illuminates mechanisms linking inflammation, amyloidosis, and cardiovascular disease

    • Primate models enable exploration of how SAA1-derived amyloidosis affects multiple organ systems simultaneously

    • Long-term studies in primates can reveal cumulative effects of chronic SAA1 elevation not readily apparent in shorter-lived animal models

By leveraging these unique contributions of primate research, scientists can develop more effective diagnostic, preventive, and therapeutic approaches for human amyloidosis while gaining fundamental insights into the molecular mechanisms underlying protein misfolding diseases.

What interdisciplinary approaches could enhance our understanding of SAA1 in primate models?

Enhancing our understanding of SAA1 in primate models requires innovative interdisciplinary approaches that integrate diverse scientific disciplines and methodologies. Several promising interdisciplinary strategies include:

  • Integrative omics approaches:

    • Combining genomics, transcriptomics, proteomics, and metabolomics to create comprehensive profiles of SAA1 regulation and function

    • Single-cell multi-omics to understand cell-type specific responses to SAA1 across tissues

    • Metagenomic integration to explore interactions between the microbiome and SAA1-mediated inflammation

    • Computational integration of multi-dimensional data using advanced bioinformatics and systems biology approaches

  • Evolutionary medicine perspectives:

    • Applying phylogenetic analyses across primates to identify conserved and divergent aspects of SAA1 function

    • Exploring how natural selection has shaped SAA1 properties in different primate lineages

    • Investigating how environmental adaptations influenced SAA1 evolution

    • Using ancestral sequence reconstruction to understand the evolutionary trajectory of amyloidogenic potential

  • Advanced biophysical and structural biology:

    • Cryo-electron microscopy of SAA1 fibrils from different primate species

    • Hydrogen-deuterium exchange mass spectrometry to map structural dynamics

    • Solid-state NMR to determine atomic-level fibril structures

    • Computational modeling integrated with experimental structural data to predict species-specific aggregation mechanisms

  • Bioengineering and synthetic biology:

    • Engineered primate cell models with modified SAA1 genes using CRISPR technology

    • Synthetic protein design to test hypotheses about amyloidogenic determinants

    • Biomaterial development incorporating SAA1-derived peptides to study aggregation in controlled environments

    • Organ-on-chip technologies to model species-specific SAA1 effects in complex tissue contexts

  • Translational immunology and inflammation research:

    • Immune profiling across primate species in response to SAA1 elevation

    • Investigation of species-specific differences in inflammasome activation by SAA1

    • Exploration of trained immunity effects of SAA1 in different primate models

    • Development of immunomodulatory strategies targeting SAA1-mediated inflammation

  • Environmental health sciences:

    • Examining how environmental exposures modulate SAA1 expression and function across primates

    • Investigating potential environmental triggers of amyloidosis in susceptible primates

    • Exploring dietary influences on SAA1 metabolism and HDL remodeling

    • Assessing climate change impacts on inflammatory disease patterns in wild primate populations

  • Digital health and artificial intelligence:

    • Development of machine learning algorithms to predict amyloidosis risk from SAA1 patterns

    • Computer vision applications for automated quantification of amyloid deposits in tissues

    • Natural language processing to mine the scientific literature for SAA1 knowledge across species

    • Digital biomarker development for continuous monitoring of inflammation in primate models

By fostering collaboration across these diverse disciplines, researchers can develop more comprehensive models of SAA1 biology in primates, leading to novel insights with translational potential for human health and disease.

Product Science Overview

Expression and Production

In the context of recombinant protein production, Serum Amyloid A (APO-SAA1) Rhesus Macaque Recombinant is typically produced using E. coli expression systems. The recombinant protein is a single, non-glycosylated polypeptide chain containing 104 amino acids and has a molecular mass of approximately 11.8 kDa . The protein is purified using proprietary chromatographic techniques to ensure high purity, often greater than 97% as determined by RP-HPLC and SDS-PAGE .

Biological Functions

SAA1 is involved in several biological processes, including:

  • Immune Response: SAA1 plays a crucial role in modulating the immune response. It acts as a chemoattractant for immune cells such as monocytes, neutrophils, and T-lymphocytes .
  • Lipid Metabolism: SAA1 is associated with HDL and is involved in the transport and metabolism of cholesterol.
  • Inflammation: During acute-phase responses, the levels of SAA1 can increase dramatically, indicating its role in inflammation and tissue repair .
Tissue Distribution

SAA1 is predominantly produced in the liver, but it can also be expressed in other tissues under certain conditions. The protein circulates in the blood at low levels under normal conditions but can increase significantly during inflammation .

Regulatory Mechanisms

The expression of SAA1 is regulated by pro-inflammatory cytokines such as IL-1, IL-6, and TNF-α. These cytokines activate signaling pathways that lead to the transcriptional upregulation of the SAA1 gene during the acute-phase response .

Stability and Storage

Recombinant SAA1 is typically lyophilized and should be stored desiccated below -18°C for long-term stability. Upon reconstitution, it should be stored at 4°C for short-term use and below -18°C for long-term use. It is recommended to add a carrier protein, such as 0.1% HSA or BSA, to prevent freeze-thaw cycles .

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.