CD36 is a 472-amino-acid protein with a molecular weight of ~53 kDa that increases to ~88 kDa after glycosylation . Its structure includes:
N-terminal cytosolic domain (residues 1–7)
N-terminal transmembrane domain (residues 8–29)
Extracellular domain (residues 30–439)
C-terminal transmembrane domain (residues 440–461)
The extracellular domain forms an antiparallel β-barrel core with hydrophobic channels for ligand binding, including two entrances for fatty acids . Disulfide bonds stabilize the extracellular loop, while palmitoylation and phosphorylation modify intracellular regions .
CD36 serves dual roles as a scavenger receptor and signaling molecule:
Insulin resistance: CD36 deficiency correlates with impaired fatty acid uptake in muscle and liver, contributing to glucose intolerance .
Atherosclerosis: CD36 mediates oxidized LDL uptake in macrophages, promoting foam cell formation .
Nonalcoholic fatty liver disease (NAFLD): Overexpression exacerbates lipid accumulation .
Hepatocellular carcinoma (HCC): CD36 enhances fatty acid uptake, fueling tumor growth and metastasis .
Breast/colorectal cancer: Elevated CD36 expression associates with epithelial-mesenchymal transition .
Two phenotypes are recognized:
Type I deficiency: Absent CD36 on platelets and monocytes due to homozygous mutations (e.g., c.329_330delAC) .
Type II deficiency: Tissue-specific loss from heterozygous mutations (e.g., c.-132 A>C in promoter region) .
Over 60 polymorphisms in CD36 are linked to clinical conditions:
Current research focuses on:
Platelet glycoprotein 4 isoform 1, BDPLT10, CHDS7, FAT, GP3B, GP4, GPIV, PASIV, SCARB3, Glycoprotein IIIb, GPIIIB, Leukocyte differentiation antigen CD36, PAS-4, Platelet collagen receptor, Platelet glycoprotein IV, Thrombospondin receptor.
ADLGDLLIQK TIKKQVVLEE GTIAFKNWVK TGTEVYRQFW IFDVQNPQEV MMNSSNIQVK QRGPYTYRVR FLAKENVTQD AEDNTVSFLQ PNGAIFEPSL SVGTEADNFT VLNLAVAAAS HIYQNQFVQM ILNSLINKSK SSMFQVRTLR ELLWGYRDPF LSLVPYPVTT TVGLFYPYNN
TADGVYKVFN GKDNISKVAI IDTYKGKRNL SYWESHCDMI NGTDAASFPP FVEKSQVLQF FSSDICRSIY AVFESDVNLK GIPVYRFVLP SKAFASPVEN PDNYCFCTEK IISKNCTSYG VLDISKCKEG RPVYISLPHF LYASPDVSEP IDGLNPNEEE HRTYLDIEPI TGFTLQFAKR
LQVNLLVKPS EKIQVLKNLK RNYIVPILWL NETGTIGDEK ANMFRSQVTG KINLEPKSCD KTHTCPPCPA PELLGGPSVF LFPPKPKDTL MISRTPEVTC VVVDVSHEDP EVKFNWYVDG VEVHNAKTKP REEQYNSTYR VVSVLTVLHQ DWLNGKEYKC KVSNKALPAP IEKTISKAKG
QPREPQVYTL PPSRDELTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY KTTPPVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGKHHHH HH.
CD36 exhibits biased expression patterns across human tissues, with notably higher expression in breast, adipose, heart, colon, and white blood cells compared to other tissues . For accurate quantification, researchers should consider:
Transcriptional analysis: High-throughput sequencing methods such as those employed by Illumina have proven effective for tissue-specific transcription profiling . This approach allows for comparative analysis across multiple tissues simultaneously.
Immunohistochemistry: For protein-level detection, immunostaining of biopsied tissue samples provides spatial information on CD36 localization, which is particularly important as CD36 function differs based on its membrane localization versus cytoplasmic expression . This method has successfully demonstrated that in nonalcoholic steatosis (NAS) patients, CD36 predominantly localizes to the plasma membrane of hepatocytes in aged patients compared to younger patients .
Flow cytometry: For blood cells and isolated primary cells, flow cytometry provides quantitative data on CD36 surface expression at the single-cell level, allowing for identification of type I versus type II CD36 deficiency phenotypes .
CD36 expression exhibits significant age-dependent changes that correlate with metabolic disease susceptibility:
Age-related increase: CD36 expression is positively associated with age in individuals with histologically normal livers, but this correlation is disrupted in patients with nonalcoholic steatosis (NAS) .
Subcellular localization shift: In aged NAS patients, CD36 predominantly localizes to the plasma membrane of hepatocytes compared to younger patients, suggesting age-specific regulation of protein trafficking .
Experimental models: Studies using C57BL/6J mice at different ages (young/12-week vs. middle-aged/52-week) fed with various diets (chow, low-fat, high-fat) have demonstrated that aging affects CD36 expression patterns and correlates with enhanced susceptibility to nonalcoholic fatty liver disease .
CD36 is expressed in numerous cell types, and experimental design should account for this diversity:
Immune cells: Monocytes, macrophages, dendritic cells, and platelets all express CD36, making it relevant for immunity and thrombosis studies .
Metabolic tissues: Adipocytes, skeletal and cardiac myocytes, and hepatocytes express CD36, important for metabolic disease research .
Other significant expressions: Epithelial cells, vascular endothelium, microglia, pancreatic β-cells, erythroid precursors, kidney glomeruli and tubules cells, retinal pericytes, and pigment epithelium cells all express CD36 .
When designing tissue-specific studies, researchers should consider potential cross-talk between these cell types and account for the integrated nature of CD36 signaling across different tissues.
CD36 functions as both a signaling receptor and a fatty acid transporter, creating a unique integration point between metabolism and immunity:
Signaling mechanisms: CD36 activates distinct but analogous downstream kinase cascades depending on cellular context. In all cases, signaling involves recruitment and activation of Src family nonreceptor protein tyrosine kinases and serine/threonine kinases of the MAPK family .
Cell-specific signaling components:
Integration with metabolism: As highlighted in recent immunometabolism research, CD36-mediated fatty acid uptake affects cellular metabolic profiles, which in turn influence immune cell differentiation and activation. Metabolic intermediates themselves can act as critical signaling molecules leading to either pro- or anti-inflammatory responses .
Influence on cell fate: Through these integrated pathways, CD36 helps determine immune cell activation states and ultimately cell fate decisions, contributing to disease pathogenesis including atherosclerosis and tumor progression .
Researchers face challenges separating CD36's dual functions, but several methodological approaches have proven effective:
Radio-labeled fatty acid uptake studies: Using PET/CT with [11C]palmitate provides quantitative in vivo assessment of fatty acid uptake in multiple tissues simultaneously, as demonstrated in studies of CD36-deficient individuals .
Fatty acid uptake inhibition: Selective inhibition of CD36-mediated fatty acid transport without affecting signaling can be achieved with specific small molecule inhibitors, allowing functional separation.
Signaling pathway analysis: Phosphorylation of downstream targets like Src family kinases and MAPKs can be measured to assess signaling activation independent of fatty acid transport using phospho-specific antibodies and kinase activity assays .
Mutational analysis: Studies using CD36 variants with selective disruption of either signaling or fatty acid transport functions help distinguish these roles, particularly in cell culture systems.
CD36 involvement in Alzheimer's disease occurs through specific pathways:
β-amyloid interaction: CD36-dependent interaction with β-amyloid (Aβ) peptide leads to microglial internalization of peptides and triggers proinflammatory responses .
Inflammatory cascade: Upon Aβ binding, CD36 activates Src family kinases and MAPK pathways in microglia, contributing to chronic neuroinflammation characteristic of Alzheimer's disease .
Most appropriate research models include:
Primary microglial cultures: Allow direct assessment of CD36-Aβ interactions and downstream signaling.
Organotypic brain slice cultures: Preserve the cellular architecture while allowing manipulation of CD36 function.
Transgenic mouse models: Combining CD36 deficiency with amyloid precursor protein overexpression provides in vivo insights into CD36's role in Alzheimer's pathology.
CD36 deficiency classification requires comprehensive analysis:
Type I vs. Type II deficiency:
Characterization protocol:
Flow cytometric analysis of platelet and monocyte CD36 expression
Genetic testing for known mutations, particularly the C478T substitution (Pro90Ser) and other documented variants
Family studies to establish inheritance patterns
Functional testing such as fatty acid uptake studies using techniques like [11C]palmitate PET/CT imaging
Clinical correlation: Researchers should assess for immune-mediated thrombocytopenia, including fetal and neonatal alloimmune thrombocytopenia (FNAIT), platelet transfusion refractoriness (PTR), and posttransfusion purpura (PTP) .
CD36 genetic variants impact lipid metabolism through several mechanisms:
Fatty acid utilization impairment: CD36-null mice show abnormal plasma lipid and lipoprotein profiles and resting hypoglycemia attributable to impaired fatty acid utilization in cardiac and striated muscle and reduced fatty acid uptake by adipose tissues .
Contextual effects: The role of CD36 in metabolic disease appears complex and context-dependent:
In some models, ablation of CD36-mediated lipid uptake prevented lipotoxicity and insulin resistance
In other contexts, liver-specific CD36 induction contributed to steatosis
The cd36-null mutation in spontaneously hypertensive rats has been linked to insulin resistance, though results are controversial
Human studies: Studies of individuals lacking CD36 show conflicting results regarding insulin resistance, potentially due to differential effects across organs and variations in mutation types .
Ethnic Group | Platelet CD36 Deficiency Rate (%) | Reference |
---|---|---|
Arabians | 2.6% | |
Asians | 3-11% | |
Sub-Saharan Africans | 8% | |
Caucasians | <0.4% |
Researchers employ multiple complementary approaches to assess CD36 variant functionality:
Massively Parallel Reporter Assay (MPRA): This high-throughput approach effectively identifies expression quantitative trait loci (eQTLs) affecting CD36 transcription. Studies have identified significant eQTLs located 13kb to 55kb upstream of the CD36 transcriptional start site of transcript ENST00000309881 and 49kb to 92kb upstream of transcript ENST00000447544 .
Bayesian statistical methods: Novel Bayesian approaches have proven effective for analyzing MPRA data to identify significant transcription shifts between minor and major alleles .
In vivo metabolic phenotyping: Functional assessment through techniques like [11C]palmitate PET/CT imaging provides direct measurement of fatty acid uptake in multiple tissues simultaneously, as demonstrated in studies of individuals with the Pro90Ser CD36 mutation .
Cell culture models: Expression of variant CD36 in cellular systems allows assessment of effects on fatty acid uptake, signaling pathway activation, and protein localization.
NAFLD research involving CD36 requires multi-faceted approaches:
Human tissue analysis:
Animal models:
Age-comparative design: Young (e.g., 12-week) and middle-aged (e.g., 52-week) C57BL/6J mice allow assessment of age-dependent effects
Dietary intervention: Comparing chow diet, low-fat diet (10% kcal fat), and high-fat diet (60% kcal fat) over 12-week periods
Tissue collection: Liver, adipose, and muscle samples should be analyzed for CD36 expression, localization, and associated metabolic parameters
Mechanistic studies: In vitro models using primary hepatocytes with CD36 modulation (overexpression, knockdown, or chemical inhibition) help establish direct causality.
Several complementary techniques offer valuable insights:
CD36 serves as a critical nexus between metabolism and inflammation:
Mechanism of integration: CD36 functions through dual roles as both a signaling receptor responding to damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), as well as a long chain free fatty acid transporter .
Cellular metabolism impact: CD36-mediated pathways influence immune cell differentiation and activation states, ultimately affecting cell fate decisions. This occurs because many metabolic intermediates themselves function as critical signaling molecules that can lead to either pro- or anti-inflammatory responses .
Disease relevance: The integrated function of CD36 in both innate and adaptive immune cells contributes to the pathogenesis of common diseases including atherosclerosis and tumor progression .
Research approach: Studies should simultaneously assess:
Lipid profiles and CD36 expression
Inflammatory marker production
Metabolic pathway intermediates
Immune cell activation states
CD36 pathway intervention shows significant therapeutic potential:
Targeting approaches:
Direct CD36 modulation via small molecule inhibitors or antibodies
Downstream kinase inhibition: Src family kinases (especially Lyn) and MAPK family members (particularly JNK2 and p38) represent promising targets given their cell-specific roles in CD36 signaling cascades
Cell-specific targeting: Given that Lyn and JNK2 are critical in macrophages and platelets, while Fyn and p38 predominate in endothelial cells, cell-targeted approaches could minimize side effects
Disease applications:
Cardiovascular disease: Targeting CD36-mediated platelet activation and foam cell formation
NAFLD: Inhibiting hepatic CD36-mediated lipid uptake
Alzheimer's disease: Blocking CD36-Aβ interactions to reduce neuroinflammation
Cancer: Modulating CD36's effects on tumor progression through immune cell modulation
Single-cell approaches offer unprecedented resolution for CD36 research:
Single-cell RNA sequencing (scRNA-seq): Enables identification of cell populations with distinct CD36 expression patterns within heterogeneous tissues, revealing previously unrecognized functional subpopulations.
Single-cell proteomics: Provides insights into CD36 protein levels and post-translational modifications at individual cell resolution.
Spatial transcriptomics: Preserves tissue architecture information while providing transcriptional data, offering critical insights into CD36 function in the tissue microenvironment.
Cell-specific CRISPR editing: Allows precise modulation of CD36 or its signaling partners in specific cell populations to assess functional consequences.
Implementation strategy: Researchers should consider integrated multi-omic approaches that combine:
scRNA-seq for broad cell typing and pathway analysis
Targeted protein analysis for CD36 and signaling partners
Spatial information to maintain tissue context
Functional validation in identified cell populations
Contradictory findings regarding CD36 and insulin resistance require careful experimental approaches:
Tissue-specific analysis: Studies should separately assess insulin sensitivity in liver, muscle, and adipose tissues rather than relying on whole-body measures, as CD36-null mice show tissue-specific insulin resistance patterns (liver insulin resistance despite whole-body insulin sensitivity) .
Compensatory mechanism assessment: Researchers should evaluate how CD36 deficiency affects glucose utilization in different tissues, as muscle may mask insulin resistance by essential utilization of glucose when fatty acid uptake is impaired .
Genetic background consideration: The specific mutations and genetic background significantly impact phenotypes. For example, human CD36 deficiency studies show variable insulin resistance results, likely due to different compound mutations affecting not just CD36 but potentially other genes .
Experimental design recommendations:
Tissue-specific CD36 knockout or overexpression models
Hyperinsulinemic-euglycemic clamp studies with tissue-specific glucose uptake measurement
Evaluation of fatty acid and glucose flux in multiple tissues simultaneously
Careful genetic characterization beyond simple CD36 status
CD36 represents a critical node in immunometabolism through several mechanisms:
Dual functionality: CD36 uniquely functions as both a pattern recognition receptor and a fatty acid transporter, allowing it to integrate immune signaling and metabolic regulation .
Metabolic intermediates as signals: CD36-mediated pathways generate metabolic intermediates that themselves function as signaling molecules, influencing inflammatory responses. This creates a feedback loop between metabolism and immunity .
Novel research approaches:
Stable isotope tracing combined with metabolomics to track metabolic flux through CD36-dependent pathways
Multi-parametric flow cytometry simultaneously assessing metabolic state and immune activation
CRISPR-based screens to identify metabolic dependencies in CD36-expressing immune cells
Systems biology approaches integrating transcriptomics, proteomics, and metabolomics data
Relevance: This intersection is particularly important for diseases including atherosclerosis, cancer, and age-related conditions where both metabolic dysfunction and inflammation play central roles .
CD36's age-related disease involvement extends beyond fatty liver:
Alzheimer's disease: CD36-dependent interaction with β-amyloid peptides triggers microglial inflammatory responses that may contribute to disease progression .
Cardiovascular disease: Age-related increases in CD36 expression may enhance platelet reactivity and foam cell formation, contributing to atherosclerosis progression .
Cancer: CD36 influences tumor microenvironment through effects on both metabolism and immune cell function, with potential impacts on tumor progression .
Research approaches:
Age-stratified study designs comparing young, middle-aged, and elderly populations
Longitudinal studies tracking CD36 expression and disease progression
Multi-tissue analysis to capture CD36's diverse effects across organs
Correlation of CD36 expression/function with established biomarkers of aging
Environmental-genetic interaction studies for CD36 require specialized approaches:
Study design considerations:
Case-control designs comparing CD36 variant carriers exposed to different environmental conditions
Family-based designs studying genotype-matched relatives under different environmental exposures
Longitudinal cohorts with repeated measures of both genetic and environmental factors
Key environmental variables:
Dietary factors: Fat content, fatty acid composition, carbohydrate type/amount
Physical activity patterns
Metabolic stressors (fasting, overfeeding)
Inflammatory exposures
Analysis approaches:
Stratified analysis by genotype and environmental exposure
Formal tests of interaction with sufficient statistical power
Mendelian randomization where appropriate
Systems genetics approaches incorporating transcriptomic responses to environmental challenges
CD36 is a single-chain protein consisting of 472 amino acid residues with both N- and C-terminal cytoplasmic tails and an extracellular loop . It is expressed on a variety of cell types, including platelets, erythrocytes, monocytes, differentiated adipocytes, mammary epithelial cells, spleen cells, and some skin microdermal endothelial cells . The protein is localized to the Golgi apparatus and is found in the cytoplasm of endothelial cells, liver sinusoids, adipocytes, thrombocytes, megakaryocytes, skeletal and cardiac muscle cells .
CD36 serves as a multiligand pattern recognition receptor that interacts with a wide range of structurally dissimilar ligands. These include long-chain fatty acids (LCFA), advanced glycation end products (AGE), thrombospondin-1, oxidized low-density lipoproteins (oxLDLs), high-density lipoprotein (HDL), phosphatidylserine, apoptotic cells, beta-amyloid fibrils, collagens I and IV, and Plasmodium falciparum-infected erythrocytes . Its primary functions include:
Recombinant human CD36 is produced using various expression systems, including insect cells (Spodoptera frugiperda, Sf21) and human embryonic kidney (HEK) 293 cells . The recombinant protein is often used in research to study its binding properties and interactions with other molecules. For example, recombinant human CD36/Fc chimera has been shown to bind to thrombospondin-2 in a functional ELISA .
CD36 is associated with several diseases, including cardiovascular diseases, metabolic disorders, and cancer . Its role in fatty acid metabolism and inflammation makes it a potential target for therapeutic interventions in these conditions. Additionally, CD36 is being investigated as a potential biomarker for various diseases due to its involvement in multiple physiological processes .