APOC1 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Applications in Research

APOC1 Antibody, FITC conjugated, is employed in diverse experimental contexts:

Immunofluorescence and Immunocytochemistry (IF/ICC)

  • Cellular localization: Detects APOC1 in intracellular compartments or membrane-bound lipoproteins.

  • Cancer studies: Identifies APOC1 overexpression in colorectal cancer (CRC) cells, correlating with tumor progression .

  • Protocol example:

    1. Fix and permeabilize cells.

    2. Block with BSA.

    3. Incubate with APOC1-FITC antibody (1:100–1:200).

    4. Visualize using fluorescence microscopy .

Flow Cytometry (FCM)

  • Quantification: Measures APOC1 expression on cell surfaces or in lysates.

  • Cancer research: Analyzes APOC1 levels in CRC patient-derived cells to assess prognosis .

Western Blotting (WB)

  • Protein validation: Confirms APOC1 expression in CRC cell lines (e.g., SW480, SW620) .

  • Pathway analysis: Detects downstream targets like p38 MAPK in APOC1 knockdown studies .

Research Findings: APOC1 in Colorectal Cancer (CRC)

APOC1 is a critical biomarker in CRC, with its detection via FITC-conjugated antibodies enabling mechanistic studies:

Functional Roles in CRC

ProcessAPOC1 EffectMechanism
Cell proliferationPromotes growthActivates p38 MAPK signaling
Cell cycleModulates G0/G1 arrestDownregulates cyclin B1/D1
ApoptosisLowers apoptosisIncreases Bcl-2 expression
Migration/InvasionEnhances motilitySuppresses EMT-related proteins

Key study: Silencing APOC1 in CRC cells (e.g., SW480) reduces migration by 20–40% and inhibits p38 MAPK phosphorylation .

Optimization Tips

  1. Antigen retrieval: Use TE buffer (pH 9.0) or citrate buffer (pH 6.0) for IHC .

  2. Blocking: BSA or non-fat dry milk minimizes nonspecific binding.

  3. Controls: Include secondary antibody-only controls to validate specificity.

Limitations

  • Cross-reactivity: Polyclonal antibodies may bind non-target epitopes.

  • Signal interference: FITC’s green emission (ex: 495 nm, em: 519 nm) overlaps with other fluorophores; use spectral unmixing if necessary.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the order method and location. Please consult your local distributor for specific delivery information.
Synonyms
Apolipoprotein C-I (Apo-CI) (ApoC-I) (Apolipoprotein C1) [Cleaved into: Truncated apolipoprotein C-I], APOC1
Target Names
Uniprot No.

Target Background

Function
Apolipoprotein C-I (apoC-I) is an inhibitor of lipoprotein binding to the low-density lipoprotein (LDL) receptor, LDL receptor-related protein, and very low-density lipoprotein (VLDL) receptor. It associates with high-density lipoproteins (HDL) and the triacylglycerol-rich lipoproteins in the plasma, comprising approximately 10% of the protein content in VLDL and 2% in HDL. ApoC-I appears to directly interfere with fatty acid uptake and serves as the primary plasma inhibitor of cholesteryl ester transfer protein (CETP). It binds free fatty acids, reducing their intracellular esterification. Furthermore, apoC-I modulates the interaction of apolipoprotein E (apoE) with beta-migrating VLDL and inhibits the binding of beta-VLDL to the LDL receptor-related protein.
Gene References Into Functions
  1. Knockdown of peroxisome proliferator-activated receptor gamma (PPARgamma) resulted in increased levels of translocase of outer mitochondrial membrane 40 (TOMM40), apolipoprotein E (APOE), and apolipoprotein C-I (APOC1) mRNAs, with the most significant impact on APOE transcript levels. PMID: 28065845
  2. Collectively, these findings suggest that apoC1 and apoE have redundant functions in hepatitis C virus (HCV) infection and morphogenesis. PMID: 30130702
  3. A study investigated the relationship between two variants of apoC1 and the risk of polycystic ovary syndrome, evaluating the genotypic effects on clinical, hormonal, and metabolic indexes, as well as plasma platelet-activating factor acetylhydrolase (PAF-AH) activity. PMID: 29636060
  4. Performance metrics were employed to select single nucleotide polymorphisms (SNPs) in stage 1, which were subsequently genotyped in a separate dataset (stage 2). Four SNPs (CPXM2 rs2362967, APOC1 rs4420638, ZNF521 rs7230380, and rs12965520) were identified for late-onset Alzheimer's disease (LOAD) by both traditional P-values (without correction for multiple tests) and performance metrics. PMID: 27805002
  5. Apolipoprotein C-I (ApoC-I) polymorphism might be a genetic factor contributing to longevity in the Bama population. The ApoC-I rs4420638 and rs584007 SNPs are associated with serum triglycerides and high-density lipoprotein-cholesterol levels in this longevous population. PMID: 28486432
  6. In white women, three SNPs (rs2075650 [TOMM40], rs4420638 [APOC1], and rs429358 [APOE]) were significantly associated with survival to 90 years after correction for multiple testing (p < .001). rs4420638 and rs429358 were also significantly associated with healthy aging (p = .02). No SNP was associated with longevity in African American women. In Hispanic women, 7 SNPs in linkage disequilibrium were associated with both longevity and healthy aging. PMID: 27707806
  7. APOC1 expression induces glomerulosclerosis, potentially by increasing the cytokine response in macrophages. PMID: 27976371
  8. Apolipoprotein C-I (apoC-I) inhibited in situ lipoprotein lipase (LPL) activity in adipocytes in both a concentration- and time-dependent manner. There was no change in postprandial white adipose tissue (WAT) apoC-I secretion. WAT apoC-I secretion may inhibit WAT LPL activity and promote delayed chylomicron clearance in overweight and obese subjects. PMID: 27040450
  9. Individuals with allelic variation in four genes related to cardiovascular diseases and metabolism were more likely to die: apolipoprotein (APO)C1 GG and AG carriers, APOE varepsilon4 carriers, insulin-degrading enzyme (IDE) TC carriers, and phosphatidylinositol 3-kinase (PI3KCB) GG carriers. PMID: 27806189
  10. A common single-nucleotide polymorphism in the APOC1/APOE region, previously found to be associated with protective levels of cholesterol and lower cardiovascular risk, may be associated with ideal health. PMID: 27179730
  11. These findings indicate that variants in the TOMM40/APOE/APOC1 region might be associated with human longevity. Further studies are needed to identify the causal genetic variants influencing human longevity. PMID: 26657933
  12. These results suggest that ApoC-I peptides may be a potential diagnostic biomarker and therapeutic approach for breast cancer. PMID: 27052600
  13. APOC1 SNP is associated with the A beta-42 levels in cerebrospinal fluid (CSF). PMID: 26576771
  14. This bioinformatics analysis explored the shared genetic etiology underlying Type 2 Diabetes and Alzheimer's Disease at the SNP level, gene level, and pathway level. Six SNPs on the APOC1 gene were identified. PMID: 26639962
  15. The endogenous retroviral promoters (LTRs) of the human endothelin B receptor (EDNRB) and apolipoprotein C1 (APOC1) genes have high sequence identity but differ in activity and tissue specificity. PMID: 12805445
  16. The ability of apoC1 to inhibit CETP activity is impaired in patients with diabetes. Glycation of apoC1 leads to a change in its electrostatic properties that might account, at least in part, for a loss of constitutive CETP inhibition and an increase in plasma CETP activity in patients with diabetes. PMID: 24574346
  17. APOE e4 allele status is associated with dementia and severity of Alzheimer's disease pathologic features in Parkinson disease. PMID: 24582705
  18. Data indicate that apolipoprotein C-I (APOC1) rs11568822 polymorphism was associated with increased Alzheimer's disease (AD) risk in Caucasians, Asians, and Caribbean Hispanics, but not in African Americans. PMID: 24498013
  19. Results suggest that peptide-lipid interactions drive alpha-helix binding to and retention on lipoproteins. PMID: 23670531
  20. Apolipoprotein C-I and apolipoprotein C-III inhibit lipolysis by displacing LPL from lipid emulsion particles. We also propose a role for these apolipoproteins in the irreversible inactivation of LPL by factors such as angiopoietin-like protein 4 (angptl4). PMID: 24121499
  21. Following regression analysis adjusted by collection center, gender, duration of diabetes, and average HbA1c, two SNPs were significantly associated with diabetic nephropathy (DN). rs4420638 in the APOC1 region and rs1532624 in cholesteryl ester transfer protein (CETP). PMID: 23555584
  22. Linkage disequilibrium between APOC1 and TOMM40 is found in patients with primary progressive aphasia but not in controls or patients with frontotemporal dementia. PMID: 22710912
  23. High concentrations of ApoCI are associated with increased triglycerides and decreased visceral fat mass in men with metabolic syndrome X. PMID: 18835943
  24. Increased white adipose tissue apoC-I secretion in obese women is associated with delayed postprandial dietary fat clearance mediated by increased triglyceride-rich apoC-I. PMID: 22995522
  25. The plasma level of apoC-I was significantly increased in obese individuals compared with healthy individuals. PMID: 22404376
  26. Apolipoprotein C1 (apoC1) as a CETP inhibitor no longer operates on cholesterol redistribution in high-risk patients with dyslipidemia. PMID: 22474067
  27. The observed increase in apoC-I interface affinity due to higher degrees of apoC-I-palmitoyloleoylphosphatidylcholine/triolein/water interactions may explain how apoC-I can displace larger apolipoproteins, such as apoE, from lipoproteins. PMID: 22264166
  28. Our approach, which is applicable to any set of interval scale traits that are heritable and exhibits evidence of phenotypic clustering, identified three new loci in or near APOC1, BRAP, and PLCG1, which were associated with multiple phenotype domains. PMID: 22022282
  29. Results describe the association of angiotensin-converting enzyme (ACE) and APOC1 gene polymorphisms with susceptibility to Alzheimer's disease and dementia in general, both alone and combined with the APOE gene. PMID: 21533863
  30. Variants in lipoprotein lipase (LPL), 2'-5'-oligoadenylate synthetase-like (OASL), and TOMM40/APOE-C1-C2-C4 genes are associated with multiple cardiovascular-related traits. PMID: 21943158
  31. Serum levels of apoC-I and apoC-III combined with other clinical parameters can serve as a basis for the formulation of a diagnostic score for stomach cancer patients. PMID: 21267442
  32. New isoforms of apoC-I were detected in a cohort of individuals with coronary artery disease using mass spectrometry, while the expected apoC-I isoforms were absent. PMID: 21187063
  33. This study examines the association between APOE/C1/C4/C2 gene cluster variation using tagging single nucleotide polymorphisms and plasma lipid concentration along with the risk of coronary heart disease in a prospective cohort. PMID: 20498921
  34. Data show that apoCI genotype is associated with serum levels of triglycerides and C-reactive protein (CRP), confirming the role of apoCI in lipid metabolism and suggesting that it also influences inflammation. PMID: 20580041
  35. From genetic association studies in Canadian Oji-Cree subjects, APOC1 T45S polymorphism may be linked to obesity, adipocyte regulation, body fat, waist circumference, hyperglycemia, and plasma leptin and apolipoprotein C-I levels. PMID: 19812053
  36. Apolipoprotein C-I reduces cholesteryl esters selective uptake from LDL and HDL by binding to HepG2 cells and lipoproteins. PMID: 19761869
  37. ApoC-I may have a significant role in glucose and lipid metabolism and may be useful for the early demonstration of metabolic abnormality in women with polycystic ovary syndrome. PMID: 19368908
  38. ApoE e4 and APOC1 A alleles have a better association with Alzheimer disease than ApoE e4 alone. PMID: 20145290
  39. Results identified haptoglobin alpha-1, apolipoprotein C-I, and apolipoprotein C-III as candidate biomarkers in papillary thyroid carcinoma (PTC). PMID: 19785722
  40. Cholesteryl ester transfer protein is the sole major determinant of cholesteryl ester transfer in normolipidemic rabbit plasma as a result of the inability of rabbit apoCI to change HDL electronegativity. PMID: 19417222
  41. APOC1 might be an additional susceptibility gene for late-onset Alzheimer disease. PMID: 11825674
  42. Regulated expression of the gene cluster in macrophages. PMID: 12032151
  43. Thermal unfolding of discoidal complexes of apolipoprotein (apo) C-1 with dimyristoyl phosphatidylcholine (DMPC) reveals a novel mechanism of lipoprotein stabilization based on kinetics rather than thermodynamics. PMID: 12044170
  44. Effects of mutations in apolipoprotein C-1 on the reconstitution and kinetic stability of discoidal lipoproteins. PMID: 12705839
  45. The effect of APOC1 genes on brain magnetic resonance imaging (MRI) measures were studied in subjects with age-associated memory impairment. The effects of APOC1 on hippocampal volumes appeared to be more robust than those of the APOE polymorphism. PMID: 12736801
  46. Overexpression of APOC1 prevents rosiglitazone-induced peripheral fatty acid uptake leading to severe hepatic steatosis. PMID: 14523051
  47. Overexpression of apoCI does not represent a suitable method for decreasing total cholesteryl ester transfer activity in CETP/apoCI transgenic mice, owing to a hyperlipidaemia-mediated effect on CETP gene expression. PMID: 15339254
  48. Apolipoprotein C-I (apoC-I) is a potent inhibitor of LPL-mediated triglyceride lipolysis. PMID: 15576844
  49. Role of apolipoprotein C1 in the infection process of hepatitis C virus. PMID: 15767578
  50. Results suggested that both apoE and apoCI on chromosome 19 were the susceptibility loci for coronary artery disease, and their linkage disequilibrium may be responsible for the development of coronary artery disease. PMID: 15793777

Show More

Hide All

Database Links

HGNC: 607

OMIM: 107710

KEGG: hsa:341

STRING: 9606.ENSP00000252491

UniGene: Hs.110675

Protein Families
Apolipoprotein C1 family
Subcellular Location
Secreted.
Tissue Specificity
Synthesized mainly in liver and to a minor degree in intestine. Also found in the lung and spleen.

Q&A

What is APOC1 and why is it important in research?

Apolipoprotein C-I (APOC1) is the smallest member of the apolipoprotein family, playing significant roles in various physiological and pathological processes. It has gained research attention due to its involvement in multiple disease conditions, particularly cancer and cardiovascular disorders. APOC1 has been identified as a key player in tumor progression, with elevated expression observed in several cancer types including glioblastoma and renal cell carcinoma. In glioblastoma, APOC1 promotes tumorigenesis by conferring resistance to ferroptosis through inhibiting KEAP1, promoting nuclear translocation of NRF2, and increasing expression of HO-1 and NQO1 . Additionally, systematic pan-cancer analysis has identified APOC1 as an immunological biomarker that regulates macrophage polarization and promotes tumor metastasis . Its detection and study using specific antibodies, such as FITC-conjugated variants, therefore provides valuable insights into disease mechanisms and potential therapeutic approaches.

What are the key characteristics of APOC1 antibody with FITC conjugation?

APOC1 antibody with FITC (Fluorescein isothiocyanate) conjugation is a specialized immunological tool designed for fluorescence-based detection methods. The FITC conjugation enables direct visualization of APOC1 protein in various applications. Key characteristics include:

  • Antibody type: Available in both polyclonal and monoclonal formats, with polyclonal being more common

  • Host species: Typically raised in rabbit or mouse against human, rat, or mouse APOC1

  • Conjugate properties: The FITC fluorophore absorbs blue light (approximately 495 nm) and emits green fluorescence (approximately 519 nm)

  • Format: Often available as IgG-FITC conjugate, typically supplied lyophilized from PBS pH 7.4 with stabilizers like BSA and trehalose

  • Storage requirements: Most require storage at 2-8°C and should not be frozen to maintain conjugate stability

  • Applications: Primarily used in fluorescence-based techniques including immunofluorescence (IF), immunocytochemistry (ICC), immunohistochemistry (IHC), and flow cytometry (FACS)

The direct conjugation eliminates the need for secondary antibodies in fluorescence applications, streamlining experimental workflows and reducing background signal in multi-color experiments.

What are the validated applications for APOC1 antibody with FITC conjugation?

APOC1 antibody with FITC conjugation has been validated for multiple research applications, each requiring specific methodological considerations:

  • Immunofluorescence (IF): Used to visualize APOC1 localization in fixed tissue sections or cell cultures

    • Optimal dilution typically ranges from 1:50 to 1:200 depending on antibody concentration and sample type

    • Compatible with both paraffin-embedded and frozen sections

  • Flow cytometry (FACS): For quantitative assessment of APOC1 expression in cell populations

    • Typical working dilutions range from 1:20 to 1:100

    • Particularly useful for analyzing APOC1 expression in immune cells, especially macrophages

  • Immunocytochemistry (ICC): For cellular localization studies in cultured cells

    • Effective for studying APOC1's subcellular distribution and expression patterns

    • Often used in cancer cell lines to assess APOC1 upregulation

  • Immunohistochemistry (IHC): For examining APOC1 expression patterns in tissue contexts

    • Can be used on frozen and paraffin-embedded sections with appropriate antigen retrieval

Each application requires optimization of fixation conditions, antibody concentration, incubation times, and appropriate controls to ensure specific and reproducible results.

How should researchers design experiments to study APOC1's role in tumor microenvironment using FITC-conjugated antibodies?

Designing experiments to investigate APOC1's role in the tumor microenvironment using FITC-conjugated antibodies requires careful methodological planning:

  • Multi-color immunofluorescence approach:

    • Combine APOC1-FITC antibody with markers for different cell types (e.g., CD68 for macrophages, CD3 for T cells)

    • Use complementary fluorophores (e.g., TRITC, Cy5) that don't overlap with FITC spectrum

    • Include nuclear counterstains like DAPI to facilitate cellular localization

  • Flow cytometry for immune cell profiling:

    • Design panels including APOC1-FITC alongside lineage markers for macrophages (particularly focusing on M1/M2 polarization markers like CD163 and CD206)

    • Include appropriate FMO (Fluorescence Minus One) controls to set accurate gates

    • Consider cell sorting to isolate APOC1-positive populations for further functional studies

  • Co-culture experimental design:

    • Establish co-culture systems with tumor cells and macrophages to study APOC1's role in macrophage polarization

    • Monitor APOC1 expression changes using the FITC-conjugated antibody in flow cytometry or live-cell imaging

    • Include appropriate knockdown or overexpression controls to establish causality

  • In vivo tumor models:

    • Design experiments to analyze tumor sections using APOC1-FITC antibodies alongside other markers

    • Consider using fresh frozen tissue to preserve FITC signal

    • Implement appropriate positive and negative controls

Based on recent research, particular attention should be given to macrophage populations, as APOC1 has been shown to regulate macrophage polarization toward the M2 phenotype and promote tumor metastasis through mechanisms involving CCL5 secretion .

What are the critical parameters for optimizing immunofluorescence protocols with APOC1-FITC antibodies?

Optimizing immunofluorescence protocols with APOC1-FITC antibodies requires attention to several critical parameters:

  • Fixation method:

    • Paraformaldehyde (4%) is generally recommended for preserving both protein structure and FITC fluorescence

    • Avoid methanol fixation which can diminish FITC signal intensity

    • Fixation time should be optimized (typically 10-20 minutes) to balance structural preservation and epitope accessibility

  • Permeabilization conditions:

    • For intracellular APOC1 detection, use 0.1-0.3% Triton X-100 or 0.1% saponin

    • Excessive permeabilization can lead to loss of cellular architecture and increased background

    • Duration should be optimized based on cell/tissue type (typically 5-15 minutes)

  • Blocking parameters:

    • Use 3-5% BSA or 5-10% normal serum from the same species as the secondary antibody (if using)

    • Include 0.1% Tween-20 to reduce non-specific binding

    • Block for at least 30-60 minutes at room temperature

  • Antibody dilution and incubation:

    • Start with manufacturer's recommended dilution (typically 1:50 to 1:200)

    • Perform titration experiments to determine optimal concentration

    • Consider temperature (4°C overnight versus room temperature for 1-2 hours)

  • Anti-photobleaching measures:

    • Minimize exposure to light during all protocol steps

    • Use anti-fade mounting media containing DAPI for nuclear counterstaining

    • Store slides at 4°C in the dark and image promptly

  • Controls:

    • Include negative controls (omitting primary antibody)

    • Use tissues or cells known to express APOC1 as positive controls

    • Consider competing peptide controls to verify specificity

By systematically optimizing these parameters, researchers can achieve specific staining with minimal background and preserve FITC signal intensity for accurate APOC1 detection.

How can researchers troubleshoot weak or non-specific signals when using APOC1-FITC antibodies?

When encountering weak or non-specific signals with APOC1-FITC antibodies, researchers should implement the following troubleshooting strategies:

For weak signal problems:

  • Antibody concentration:

    • Increase antibody concentration incrementally (e.g., from 1:200 to 1:100 to 1:50)

    • Extend incubation time (overnight at 4°C instead of 1-2 hours at room temperature)

  • Antigen retrieval enhancement:

    • For paraffin sections, optimize antigen retrieval methods (citrate buffer pH 6.0 or EDTA buffer pH 9.0)

    • Extend retrieval time or adjust temperature parameters

    • Consider enzymatic retrieval for certain tissue types

  • Detection system amplification:

    • Use biotin-streptavidin amplification systems if signal remains weak

    • Consider tyramide signal amplification (TSA) for very low abundance targets

  • Storage and handling:

    • Ensure antibody hasn't been subjected to freeze-thaw cycles which can diminish FITC activity

    • Store in small aliquots protected from light at recommended temperature (2-8°C)

For non-specific signal problems:

  • Blocking optimization:

    • Increase blocking reagent concentration (5-10% BSA or normal serum)

    • Extend blocking time to 2 hours or overnight at 4°C

    • Add 0.1-0.3% Triton X-100 to blocking buffer to reduce hydrophobic interactions

  • Washing procedure enhancement:

    • Increase number of washes (5-6 times for 5 minutes each)

    • Use TBS-T instead of PBS-T if phosphate interferes with antibody binding

    • Include gentle agitation during washing steps

  • Antibody specificity verification:

    • Perform peptide competition assays using human APOC1 purified protein

    • Test antibody on known negative tissue/cells

    • Verify results with a different APOC1 antibody clone

  • Autofluorescence reduction:

    • Treat sections with sodium borohydride (0.1% for 2 minutes) to reduce fixative-induced autofluorescence

    • Use Sudan Black B (0.1-0.3% in 70% ethanol) for 5-10 minutes to block lipofuscin autofluorescence

    • Consider spectral unmixing during image acquisition if available

Systematic implementation of these strategies while changing one parameter at a time will help identify and resolve issues with APOC1-FITC antibody staining protocols.

How can APOC1-FITC antibodies be utilized to investigate the mechanistic relationship between APOC1 and ferroptosis in cancer?

Recent research has uncovered a critical relationship between APOC1 and ferroptosis resistance in cancer, particularly in glioblastoma . FITC-conjugated APOC1 antibodies can be instrumental in investigating this relationship through several advanced approaches:

  • Multiparameter imaging analysis:

    • Co-stain tissue sections or cells with APOC1-FITC antibody and markers of ferroptosis (e.g., ACSL4, GPX4)

    • Evaluate nuclear translocation of NRF2 in relation to APOC1 expression using confocal microscopy

    • Quantify co-localization coefficients between APOC1 and KEAP1 to study inhibitory interactions

  • Flow cytometry-based mechanistic studies:

    • Measure lipid ROS (using C11-BODIPY) in conjunction with APOC1-FITC staining

    • Assess ferroptosis sensitivity in sorted APOC1-high versus APOC1-low cell populations

    • Monitor changes in APOC1 expression following treatments with ferroptosis inducers (e.g., erastin, RSL3)

  • Live-cell imaging approaches:

    • Combine APOC1-FITC antibody labeling with membrane-permeable probes for glutathione (GSH) or lipid peroxidation

    • Track temporal changes in APOC1 expression during ferroptosis induction

    • Correlate APOC1 levels with cellular antioxidant responses

  • Molecular pathway analysis:

    • Design experiments to track the APOC1-KEAP1-NRF2 axis using APOC1-FITC alongside other antibodies

    • Investigate trans-sulfuration pathway components (particularly CBS) in relation to APOC1 expression

    • Examine how APOC1 knockdown affects GPX4 expression using combinatorial antibody approaches

When investigating APOC1's role in ferroptosis, researchers should particularly focus on:

  • The relationship between APOC1 and NRF2 nuclear translocation

  • Effects on HO-1 and NQO1 expression as downstream targets

  • Changes in CBS expression and GSH synthesis

  • The impact on GPX4 levels and subsequent lipid ROS regulation

These approaches leverage the specificity and fluorescent properties of APOC1-FITC antibodies to dissect the complex mechanisms by which APOC1 confers ferroptosis resistance, potentially leading to novel therapeutic strategies targeting this pathway in cancer .

What are the considerations for using APOC1-FITC antibodies in studying the immunomodulatory functions of APOC1 in the tumor microenvironment?

APOC1 has emerged as an important immunomodulatory molecule, particularly in its interaction with macrophages in the tumor microenvironment . When using APOC1-FITC antibodies to study these functions, researchers should consider:

  • Experimental design for macrophage polarization studies:

    • Develop multi-color flow cytometry panels combining APOC1-FITC with markers for M1 (CD80, CD86, MHC-II) and M2 (CD163, CD206) macrophage phenotypes

    • Use appropriate compensation controls to account for spectral overlap with other fluorophores

    • Design time-course experiments to track APOC1 expression changes during macrophage polarization

  • Co-culture systems optimization:

    • Establish tumor cell-macrophage co-culture systems with varying ratios to assess APOC1's role

    • Use transwell systems to distinguish between contact-dependent and secreted factor effects

    • Monitor APOC1 transfer between cell populations using FITC-labeled antibodies

  • Receptor interaction studies:

    • Design experiments to investigate APOC1's interaction with CD163 and CD206 receptors on macrophages

    • Use proximity ligation assays in combination with APOC1-FITC antibodies to visualize molecular interactions

    • Consider competitive binding assays to characterize binding kinetics

  • Cytokine/chemokine profiling correlation:

    • Correlate APOC1 expression (detected via FITC-antibody) with CCL5 secretion levels

    • Implement Luminex or cytokine array approaches alongside flow cytometry for comprehensive profiling

    • Design inhibition experiments to establish causality in APOC1-induced cytokine production

  • In vivo tumor model considerations:

    • Develop protocols for analyzing APOC1 expression in tumor-associated macrophages from fresh tissue

    • Implement appropriate tissue processing methods to preserve both antigenicity and fluorescence

    • Consider intravital imaging approaches for dynamic studies of APOC1 in the tumor microenvironment

  • Technical optimizations for immune cell analysis:

    • Adjust fixation protocols to maintain surface marker expression alongside APOC1 detection

    • Implement gentle cell isolation procedures to preserve fragile myeloid populations

    • Consider using cell-sorting approaches to isolate specific APOC1-expressing immune populations for functional assays

When investigating APOC1's immunomodulatory functions, particular attention should be given to macrophage populations since research has shown that APOC1 promotes M2 polarization of macrophages through interactions with CD163 and CD206, subsequently enhancing tumor metastasis through CCL5 secretion .

How do results from FITC-conjugated APOC1 antibodies compare with other detection methods for APOC1 expression analysis?

Researchers should understand the comparative advantages and limitations of FITC-conjugated APOC1 antibodies versus other detection methods for comprehensive experimental planning:

Detection MethodAdvantagesLimitationsBest Applications
FITC-conjugated APOC1 antibody- Direct detection without secondary antibody
- Suitable for multicolor IF and flow cytometry
- Good for live cell applications
- Rapid protocols with fewer steps
- Potential photobleaching
- No signal amplification
- Limited shelf-life
- Sensitivity to pH changes
- Flow cytometry
- Immunofluorescence
- Live cell imaging
- Multi-color applications
HRP-conjugated APOC1 antibody- Enzymatic amplification increases sensitivity
- Stable signal that doesn't photobleach
- Permanent staining for long-term storage
- Compatible with brightfield microscopy
- Not suitable for live cell imaging
- Limited multiplexing capability
- Potential for diffusion artifacts
- Endogenous peroxidase interference
- IHC in paraffin sections
- Chromogenic Western blotting
- Applications requiring signal amplification
- Long-term archival studies
Unconjugated APOC1 antibody with fluorescent secondary- Flexible choice of secondary antibody fluorophores
- Signal amplification (multiple secondaries per primary)
- Primary antibody concentration can be reduced
- Compatible with other detection systems
- Additional incubation steps
- Potential cross-reactivity of secondary antibodies
- Higher background in some applications
- More complex protocols
- Multiplexed immunofluorescence
- Applications requiring signal amplification
- When antibody concentration is limited
- When flexible detection systems are needed
APOC1 qPCR- Quantitative assessment of mRNA expression
- High sensitivity for low abundance transcripts
- No antibody-related artifacts
- High-throughput compatible
- Does not measure protein levels
- Cannot determine protein localization
- Requires RNA isolation
- Post-transcriptional regulation not captured
- Expression level screening
- Transcript variant analysis
- When protein antibodies are unavailable
- Highly quantitative applications

When selecting between these methods, researchers should consider:

  • Research question alignment: FITC-conjugated antibodies excel in cellular localization and co-expression studies, particularly in immune cell populations where APOC1 shows variable expression .

  • Data integration approaches: Combining multiple methods provides complementary insights:

    • Verify FITC-antibody protein detection results with mRNA expression data

    • Confirm localization patterns with alternative detection methods

    • Use different methodologies for screening versus detailed mechanistic studies

  • Result interpretation considerations:

    • FITC signal intensity may not perfectly correlate with expression levels due to quenching effects

    • Different detection methods may yield slightly different patterns due to epitope accessibility

    • Quantification approaches must be tailored to the specific detection method

By understanding these comparative aspects, researchers can select the optimal approach for their specific APOC1 research questions and interpret results appropriately across different experimental platforms.

What methodological approaches should be considered when analyzing contradictory data between APOC1 expression and clinical outcomes in cancer research?

When researchers encounter contradictory data regarding APOC1 expression and clinical outcomes in cancer research, several methodological approaches should be implemented to resolve discrepancies:

  • Multivariate analysis frameworks:

    • Implement Cox proportional hazards models that include APOC1 expression alongside established prognostic factors

    • Perform stratified analyses based on cancer subtypes, stage, and molecular characteristics

    • Consider interaction terms between APOC1 and other biomarkers to identify context-dependent effects

  • Methodological triangulation:

    • Compare results across multiple APOC1 detection methods (FITC-antibody IHC, qPCR, proteomics)

    • Harmonize scoring systems for APOC1 positivity across studies

    • Evaluate both continuous and categorical approaches to APOC1 expression analysis

  • Integrative -omics approaches:

    • Correlate APOC1 protein expression with RNA sequencing and DNA methylation data

    • Analyze APOC1 in the context of relevant pathway alterations

    • Apply network analysis to position APOC1 within functional modules that may explain contextual effects

  • Temporal and spatial consideration:

    • Evaluate APOC1 expression at different disease stages and treatment timepoints

    • Distinguish between APOC1 expression in tumor cells versus stromal/immune compartments

    • Assess potential changes in APOC1 function during disease progression

  • Functional validation experiments:

    • Design in vitro and in vivo models with varying levels of APOC1 expression

    • Implement APOC1 knockdown/overexpression in different genetic backgrounds

    • Evaluate phenotypic outcomes in models that recapitulate specific clinical scenarios

  • Meta-analytical approaches:

    • Conduct systematic reviews with pre-specified inclusion criteria and quality assessment

    • Implement random-effects models to account for between-study heterogeneity

    • Perform sensitivity analyses excluding studies with methodological limitations

Recent research has highlighted potentially context-dependent roles of APOC1 in different cancers. In glioblastoma, APOC1 promotes tumorigenesis through ferroptosis resistance mechanisms , while pan-cancer analysis suggests APOC1 functions as an immunological biomarker regulating macrophage polarization . These seemingly contradictory findings might be reconciled by considering tissue-specific effects, the tumor immune microenvironment, and the impact of specific molecular alterations that co-occur with APOC1 dysregulation.

What emerging applications for APOC1-FITC antibodies should researchers consider for advancing cancer and cardiovascular disease research?

Based on recent advances in understanding APOC1 biology, several emerging applications for APOC1-FITC antibodies warrant exploration:

  • Single-cell analysis technologies:

    • Integrate APOC1-FITC antibodies into CyTOF/mass cytometry panels for high-dimensional immune profiling

    • Develop protocols for APOC1 detection in single-cell RNA-seq with protein (CITE-seq) approaches

    • Apply spatial transcriptomics combined with APOC1 immunofluorescence for tissue-level expression mapping

  • Liquid biopsy applications:

    • Explore APOC1 detection in circulating tumor cells using FITC-conjugated antibodies

    • Develop flow cytometry protocols for detecting APOC1-positive extracellular vesicles

    • Investigate APOC1 as a marker for tumor-educated platelets or leukocytes in peripheral blood

  • Therapeutic response monitoring:

    • Track changes in APOC1 expression during immunotherapy treatment

    • Correlate APOC1 levels with response to ferroptosis-inducing therapies

    • Develop companion diagnostic approaches using APOC1-FITC antibodies for patient stratification

  • Intravital imaging approaches:

    • Adapt APOC1-FITC antibodies for intravital microscopy to study real-time dynamics

    • Develop methods for in vivo tracking of APOC1-expressing cells in tumor models

    • Implement APOC1 detection in cleared tissue samples for whole-organ expression mapping

  • Nanoscale imaging technologies:

    • Apply super-resolution microscopy (STORM, PALM) to map APOC1 distribution at nanoscale resolution

    • Implement correlative light and electron microscopy (CLEM) to study APOC1 subcellular localization

    • Utilize expansion microscopy to resolve APOC1 interactions with binding partners

  • Artificial intelligence integration:

    • Develop machine learning algorithms for automated quantification of APOC1 expression patterns

    • Implement computer vision approaches for analyzing APOC1 distribution in spatial context

    • Create predictive models connecting APOC1 expression patterns with clinical outcomes

These emerging applications leverage the specificity and fluorescence properties of APOC1-FITC antibodies to address key knowledge gaps, particularly in understanding APOC1's role in tumor-immune interactions and its potential as a therapeutic target in both cancer and cardiovascular disease contexts.

How might researchers design longitudinal studies to track APOC1 expression changes during disease progression using FITC-conjugated antibodies?

Designing rigorous longitudinal studies to track APOC1 expression changes during disease progression requires careful methodological planning:

  • Cohort design considerations:

    • Establish well-defined patient cohorts with standardized sampling timepoints

    • Include pre-disease, early-stage, advanced-stage, and post-treatment samples when possible

    • Implement appropriate power calculations to determine sample size requirements

    • Include control groups matched for relevant demographic and clinical variables

  • Sample collection standardization:

    • Develop standard operating procedures for consistent tissue acquisition and processing

    • Implement rapid fixation protocols to preserve APOC1 antigenicity

    • Create tissue microarrays (TMAs) from longitudinal samples for batch processing

    • Consider establishing living biobanks (patient-derived xenografts, organoids) for functional studies

  • Multimodal tracking approaches:

    • Design flow cytometry panels for APOC1-FITC combined with lineage markers for immune monitoring

    • Implement serial liquid biopsy protocols for minimally invasive APOC1 monitoring

    • Develop quantitative imaging workflows for consistent APOC1 assessment across timepoints

    • Consider companion animal studies for more frequent sampling possibilities

  • Quantification and normalization strategies:

    • Establish quantitative metrics for APOC1 expression (mean fluorescence intensity, H-score, etc.)

    • Implement internal controls and reference standards for cross-timepoint normalization

    • Use digital pathology approaches for objective quantification

    • Apply mixed-effects statistical models designed for longitudinal data analysis

  • Integration with clinical parameters:

    • Correlate APOC1 expression changes with disease-specific clinical markers

    • Track response to therapeutic interventions alongside APOC1 expression

    • Implement multivariate analysis approaches to identify predictive patterns

    • Develop predictive models for disease progression based on APOC1 dynamics

  • Technical innovations for longitudinal tracking:

    • Consider window chamber models for repeated intravital imaging of APOC1 in preclinical studies

    • Develop multiplexed approaches to simultaneously track APOC1 and related markers

    • Implement machine learning algorithms for pattern recognition across timepoints

    • Integrate with other longitudinal -omics data for systems-level analysis

When designing such studies, researchers should pay particular attention to APOC1's role in macrophage polarization and ferroptosis resistance , as these mechanisms may evolve during disease progression. For example, tracking changes in APOC1 expression alongside macrophage polarization markers (CD163, CD206) and ferroptosis indicators could provide insight into how these processes contribute to disease advancement and treatment response.

What are the key methodological considerations researchers should prioritize when working with APOC1-FITC antibodies?

When working with APOC1-FITC antibodies, researchers should prioritize the following methodological considerations to ensure robust and reproducible results:

  • Validation and controls:

    • Verify antibody specificity through appropriate positive and negative controls

    • Perform peptide competition assays to confirm binding specificity

    • Include isotype controls to assess non-specific binding

    • Validate findings using complementary detection methods

  • Technical optimization:

    • Determine optimal fixation and permeabilization conditions for your specific sample type

    • Titrate antibody concentration to achieve optimal signal-to-noise ratio

    • Implement rigorous protection from photobleaching at all protocol stages

    • Establish consistent image acquisition parameters across experiments

  • Experimental design:

    • Include appropriate biological replicates (minimum n=3) for statistical validity

    • Design experiments with appropriate power to detect biologically meaningful differences

    • Consider potential confounding variables in your experimental system

    • Implement blinding procedures for analysis where appropriate

  • Data analysis and interpretation:

    • Apply quantitative approaches with clearly defined metrics for APOC1 expression

    • Consider both intensity and distribution patterns in image analysis

    • Implement appropriate statistical tests based on data distribution

    • Acknowledge limitations of the detection method in result interpretation

  • Reporting standards:

    • Provide detailed methodological descriptions including antibody catalog number, lot, dilution

    • Report all image acquisition parameters (exposure times, gain settings, etc.)

    • Include representative images showing both positive and negative staining

    • Make raw data available when possible for transparency

By prioritizing these methodological considerations, researchers can generate high-quality data on APOC1 expression patterns that contribute meaningfully to understanding its role in disease processes, particularly in cancer and cardiovascular research contexts.

How should researchers interpret APOC1 expression data in the context of its diverse biological functions?

Interpreting APOC1 expression data requires nuanced consideration of its diverse biological functions across different physiological and pathological contexts:

  • Context-dependent interpretation frameworks:

    • Consider tissue-specific baseline expression levels when interpreting changes

    • Evaluate APOC1 expression relative to relevant pathway components (e.g., NRF2 pathway in ferroptosis contexts )

    • Interpret findings in light of the specific disease state being studied

    • Account for potential post-translational modifications that may affect antibody binding

  • Cellular source considerations:

    • Distinguish between APOC1 expressed by tumor cells versus stromal/immune cells

    • Pay particular attention to macrophage expression given APOC1's role in macrophage polarization

    • Consider paracrine versus autocrine signaling contexts

    • Evaluate spatial relationships between APOC1-expressing and responding cells

  • Functional correlation approaches:

    • Correlate APOC1 expression with functional readouts of relevant pathways

    • In cancer contexts, assess relationships with ferroptosis markers and macrophage polarization

    • In cardiovascular disease, evaluate lipid metabolism parameters

    • Consider correlation with patient outcomes and treatment responses

  • Integrative analysis strategies:

    • Implement multivariate analyses incorporating clinical and molecular variables

    • Position APOC1 within relevant signaling networks based on co-expression patterns

    • Consider genetic and epigenetic regulators of APOC1 expression

    • Apply causal inference methods to distinguish drivers from passengers

  • Translational relevance assessment:

    • Evaluate potential as biomarker for disease diagnosis, prognosis, or treatment response

    • Consider implications for therapeutic targeting based on expression patterns

    • Assess potential off-target effects based on expression in non-target tissues

    • Develop predictive models incorporating APOC1 expression data

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.