Fact-checked by Grok 2 weeks ago

CD36

CD36, also known as scavenger receptor class B member 2 (SR-B2), is an kDa transmembrane that functions as a class B receptor, primarily facilitating the high-affinity uptake of long-chain s (LCFAs) and oxidized across the in various cell types. First identified in 1977 as glycoprotein IV on platelets, CD36 plays a critical role in by enabling efficient transport and utilization, particularly in energy-demanding tissues like the heart and . Structurally, CD36 consists of 472 encoded by a on 7q21.11, featuring a hairpin-like with two hydrophobic transmembrane domains flanking a large extracellular loop that contains multiple N-linked sites for binding and a hydrophobic cavity capable of accommodating up to two molecules. The protein's cytoplasmic tails include palmitoylation sites that regulate its trafficking and vesicular recycling between intracellular endosomes and the plasma membrane, a process modulated by factors such as insulin, , and activity. CD36 is widely expressed on the surface of diverse cells, including platelets, macrophages, monocytes, endothelial cells, adipocytes, cardiomyocytes, skeletal myocytes, hepatocytes, and specialized cells like cells in the oral . In terms of function, CD36 binds a broad array of ligands beyond LCFAs, such as oxidized (oxLDL), thrombospondin-1 (TSP-1), (AGEs), and anionic phospholipids, mediating processes like of apoptotic cells, platelet activation, and inflammatory signaling through downstream pathways involving Src kinases, MAPKs, and . It operates primarily as a and promotes LCFA uptake via a flip-flop mechanism that translocates fatty acids from the outer to the inner leaflet of the plasma membrane, contributing to accumulation under conditions of excess fat supply. Additionally, CD36 negatively regulates by interacting with TSP-1 on endothelial cells and participates in innate immunity by facilitating the recognition of microbial components and modified self-ligands. Physiologically, CD36 is essential for maintaining cellular fatty acid and energy substrate balance, particularly in the heart where fatty acids provide 60-90% of ATP via oxidation and CD36 facilitates a major portion of fatty acid uptake, and in where it aids in lipid storage and mobilization during exercise-induced . Its expression and activity are dynamically regulated by nutritional status, hormones like insulin, and mechanical stimuli such as , enabling adaptive responses to dietary fat intake and exercise. In sensory , CD36 contributes to the oral of dietary fats by detecting LCFAs in cells, influencing feeding behavior and energy intake. Dysregulation of CD36 is implicated in numerous metabolic and cardiovascular disorders, including where it promotes oxLDL uptake and formation in macrophages, and through excessive fatty acid influx leading to in adipocytes and β-cells, and diabetic via impaired cardiac and contractile dysfunction. Elevated levels of soluble CD36 (sCD36), a circulating form shed from cell surfaces, serve as a for , , and non-alcoholic (NAFLD), correlating with disease severity in cohort studies. Furthermore, CD36 contributes to neurodegenerative conditions like by facilitating amyloid-β uptake in and to malarial complications through cytoadherence of infected erythrocytes to endothelial cells expressing CD36. Therapeutic strategies targeting CD36, such as antibodies or small-molecule inhibitors, have shown promise in preclinical models for reducing lipid overload and improving metabolic outcomes.

Structure

Primary Structure

CD36 is a transmembrane encoded by the CD36 located on 7q11.2 in humans. The primary translation product consists of 472 , with an unglycosylated molecular weight of approximately 53 . The polypeptide chain exhibits a hairpin-like topology, characterized by short cytoplasmic tails at the N- and C-termini (approximately 28 and 15 , respectively), two transmembrane domains (spanning residues 2-22 and 449-472), and a large extracellular domain comprising the majority of the protein (residues 30-439). The extracellular domain features specific sequence motifs, including six conserved cysteine residues that form three intramolecular disulfide bridges: Cys243-Cys311, Cys272-Cys333, and Cys313-Cys322. These disulfide bonds contribute to the structural stability of the extracellular region. Posttranslational at multiple sites increases the apparent molecular weight of the mature protein to around 88 . The primary structure of CD36 is highly conserved across species, with identity ranging from 53% to 100% among vertebrates and particularly strong among mammals (e.g., 83% identity between and CD36, and 82% between and bovine CD36). This conservation underscores the evolutionary importance of key motifs, such as the transmembrane glycines and extracellular cysteines, which are preserved in mammalian orthologs.

Tertiary Structure

CD36 is an integral membrane glycoprotein that folds into a predominantly monomeric tertiary structure, characterized by two α-helical transmembrane at the N- and C-termini that span the , connected by a large extracellular spanning approximately residues 30 to 440. This extracellular features a central antiparallel β-barrel core, approximately 30 Å in height and 25 Å in width, adorned with multiple short α-helices that stabilize the overall oval-shaped architecture. The β-barrel and flanking helices collectively form a hydrophobic tunnel that extends laterally through the , approximately 30 Å long and lined with nonpolar residues, enabling the translocation of nonpolar substrates like long-chain fatty acids from the toward the . The high-resolution of the human CD36 extracellular domain, determined by at 2.07 resolution (PDB ID: 5LGD), captures it in a monomeric state bound to long-chain s and a malarial PfEMP1 domain, highlighting a conserved hydrophobic groove on the membrane-distal surface where the fatty acid acyl chain inserts while the polar head group remains exposed. Although the resolved structure depicts a , biochemical and modeling studies indicate potential for dimerization or oligomerization via interfaces involving the transmembrane helices and extracellular loops, which may modulate ligand affinity under physiological conditions. The β-barrel core provides a rigid scaffold for the extracellular domain, while variable loops, particularly those near the hydrophobic groove (e.g., residues 140-155 and 310-320), confer specificity for diverse by altering access to the through dynamic flexing. Recent simulations (2023) have elucidated the binding pockets, revealing that ligand engagement induces subtle conformational shifts in the loops and tunnel entrance, facilitating acyl chain partitioning into the without major domain rearrangements. These insights, complemented by functional-structural analyses (2022), underscore how such changes optimize uptake while maintaining the protein's monomeric integrity in lipid bilayers.

Posttranslational Modifications

CD36 undergoes several posttranslational modifications that are critical for its maturation, stability, trafficking to the plasma membrane, and functional regulation. These include N-linked glycosylation, palmitoylation, , ubiquitination, and , each contributing to the protein's ~88,000 Da molecular weight and its role as a transmembrane . N-linked glycosylation occurs at multiple asparagine residues in the large extracellular domain, with 10 putative sites identified and nine confirmed as glycosylated, such as , , and . This modification is essential for proper , resistance to , and efficient trafficking to the cell surface, thereby enhancing overall stability and membrane localization. In the absence of glycosylation, CD36 exhibits reduced stability and impaired uptake, as demonstrated in mutagenesis studies targeting these sites. Additionally, O-linked (O-GlcNAc) modification further promotes translocation from intracellular stores to the plasma membrane, particularly in response to stimuli like insulin. Palmitoylation involves the attachment of four palmitoyl chains to cytoplasmic residues, specifically Cys3 and Cys7 in the N-terminal tail and Cys464 and Cys466 in the C-terminal tail. This reversible is indispensable for CD36's targeting to rafts, maturation through the secretory pathway, and dynamic trafficking between intracellular compartments and the plasma membrane. Mutants lacking these sites display shortened protein half-lives and defective membrane insertion, underscoring palmitoylation's role in maintaining CD36 levels and function. Phosphorylation occurs at serine and threonine residues within the extracellular domain, notably Thr92 by (PKC) and Ser237 by (PKA). These modifications modulate CD36's ligand-binding affinity and signaling capacity; for instance, Thr92 inhibits binding to thrombospondin-1, while Ser237 reduces fatty acid translocation. Such regulation provides a mechanism for fine-tuning CD36 activity in response to cellular signals, though evidence remains limited. Ubiquitination targets residues in the C-terminal cytoplasmic , primarily Lys469 and Lys472, leading to proteasomal via polyubiquitination (e.g., K48- or K63-linked chains). This process decreases CD36 stability and surface expression, thereby limiting uptake; conversely, monoubiquitination by enzymes like Parkin enhances protein stability and localization. promote ubiquitination to downregulate CD36, while insulin opposes it to sustain levels, illustrating a regulatory balance in metabolic contexts. Acetylation modifies lysine residues such as Lys52, Lys166, Lys231, and Lys403, potentially altering ligand interactions like those with oxidized (oxLDL), which could influence pro-inflammatory signaling pathways involving CD36. However, the functional consequences of remain poorly characterized, with limited evidence on its direct impact. These modifications collectively influence CD36's and contribute to states; for example, reduced N-linked due to variants like the Asn102 in spontaneously hypertensive rats (SHR) impairs , decreases targeting, and lowers uptake, linking to cardiovascular risks. Similarly, dysregulated ubiquitination and palmitoylation in metabolic disorders can accelerate degradation or alter trafficking, exacerbating conditions such as and .

Protein-Protein Interactions

CD36 binds thrombospondin-1 (TSP-1) primarily through its extracellular , with the key located in the spanning 93 to 120, often referred to as the CLESH domain. This binding is mediated by acidic residues within the domain, including Glu101, Asp106, Glu108, and Asp109, which form electrostatic interactions with the TSP-1 type-1 repeat containing the CSVTCG motif. of Thr92 in the adjacent extracellular region can sterically hinder this interface, reducing TSP-1 affinity. At the membrane level, CD36 undergoes homodimerization and forms heterocomplexes with other proteins, such as . Dimerization is facilitated by the first , where conserved small residues like Gly12, Gly16, Ala20, and Gly23 enable close packing and stabilize the oligomeric state, as revealed by and simulations. Förster resonance energy transfer () studies in live cells have quantified this proximity, showing energy transfer efficiencies indicative of dimers and higher-order oligomers with sub-10 nm distances. Additionally, CD36 associates with αvβ3 in heterocomplexes on platelet membranes, confirmed by co-immunoprecipitation experiments that pull down both proteins under non-denaturing conditions, suggesting a 1:1 in these assemblies. These membrane-level interactions often involve tetraspanins like CD9 as scaffolds. The extracellular domain of CD36 also features docking sites for oxidized (oxLDL) and anionic s, centered on 155 to 183. This hydrophobic groove, enriched with positively charged residues, accommodates the polar heads of oxidized via ionic bonds, with Lys164 and Lys166 serving as critical anchors for electrostatic interactions; at these sites abolish affinity by over 90%. Co-immunoprecipitation assays with biotinylated oxLDL have demonstrated saturable stoichiometries of approximately 1-2 ligands per CD36 , while FRET between fluorescently tagged CD36 and analogs confirms nanoscale clustering upon ligand engagement. Similar interfaces overlap for anionic like , highlighting the domain's role in multivalent recognition. Biophysical evidence from co-immunoprecipitation and further elucidates these stoichiometries across interactors. For instance, co-IP from platelet lysates consistently recovers CD36 in equimolar complexes with Src family kinases (, Lyn, ), indicating stable 1:1 associations via the cytoplasmic tail. efficiencies in transfected cells range from 15-25% for CD36-integrin pairs, supporting dynamic heterocomplex formation without fixed oligomerization. These techniques underscore the transient yet specific nature of CD36's molecular interfaces.

Genetics

Gene Organization

The CD36 gene is situated on the long arm of human chromosome 7 at locus 7q11.2 and extends over more than 32 kilobases (kb) of genomic DNA. It comprises 15 exons separated by 14 introns, with the exons encoding both the coding sequence and untranslated regions of the mRNA. The first three exons primarily encompass the 5'-untranslated region and the initial portions of the N-terminal cytoplasmic and transmembrane domains, while exons IV through XIII encode the large extracellular domain; the C-terminal cytoplasmic and transmembrane regions are captured in exon XIV, and exon XV serves as an alternative for the 3'-untranslated region. This organization supports the production of a full-length transmembrane protein while allowing for regulatory flexibility through intron-mediated processes. The proximal promoter region of the CD36 gene, located approximately 289 nucleotides upstream of the translation start site, features a at position -28 and multiple cis-regulatory elements that facilitate basal and inducible transcription. Notably, it includes binding sites for key transcription factors such as (PPARγ), which forms a heterodimer with retinoid X receptor alpha (RXRα) to bind a PPAR (PPRE) in the promoter, enabling ligand-dependent upregulation of . Additionally, a specificity protein 1 (Sp1) binding site near the contributes to constitutive transcriptional activity in various cell types, particularly monocytic lineages. These elements underscore the gene's responsiveness to metabolic and inflammatory signals without relying on distant enhancers for core regulation.81575-5) Alternative splicing of CD36 pre-mRNA generates distinct isoforms, enhancing functional diversity. One prominent variant arises from the skipping of exons 4 and 5, which deletes 103 spanning the first and results in a secreted, soluble form known as sCD36; this isoform retains ligand-binding capability but lacks membrane anchoring, allowing it to circulate and modulate extracellular processes. Other splicing events involve or variations, but the exon-skipping mechanism for sCD36 is particularly conserved and linked to pathological conditions involving dysregulation.43561-0/fulltext) The exon-intron architecture of the CD36 gene exhibits strong across vertebrate species, with most orthologs featuring a similar number of exons—typically 12 to 15, including 12 coding exons—and preserved boundaries that align structural domains like transmembrane regions to specific exons. This evolutionary stability highlights the gene's ancient role in handling and cellular , as evidenced by genomic analyses in mammals, birds, and , where intron positions and splicing signals remain invariant despite sequence divergence in non-coding regions. Such facilitates cross-species functional studies and underscores the gene's essentiality in metazoan physiology.

Genetic Variants

CD36, a member of the scavenger receptor class B family, exhibits significant , with numerous polymorphisms and mutations influencing its expression, protein stability, and function. Common single nucleotide polymorphisms (SNPs) in the CD36 gene, such as rs1761667 (G>A) located in the promoter region, have been identified as regulators of transcriptional activity. This reduces CD36 mRNA and protein expression levels, potentially by altering promoter for transcription factors, and has been linked to modified of fatty acids in . Null mutations in the CD36 gene frequently underlie type I CD36 deficiency, characterized by absence of the protein on both platelets and monocytes, leading to impaired integration and recognition. Exemplary null alleles include the dinucleotide deletion (delAC; c.329_330del) in 5, which causes a frameshift resulting in a truncated, non-functional protein that fails to traffic to the surface. The of such CD36-null genotypes varies by , occurring in approximately 0.3% of Caucasians but reaching 3-11% in Asian populations and 2-8% in African cohorts, reflecting founder effects and selective pressures in these groups. Missense variants further diversify CD36 functionality by altering critical residues, often impacting domains. The p.Pro90Ser (c.268C>T) substitution, for instance, disrupts the extracellular region's conformation, impairing to ligands such as thrombospondin and oxidized low-density lipoproteins, and is a key cause of platelet IV deficiency (type II), where CD36 is absent on platelets but present on monocytes. This variant compromises the protein's role in adhesion and uptake processes at the molecular level. Recent investigations from 2024 have highlighted associations between specific CD36 variants and susceptibility to metabolic perturbations affecting the liver, such as , which may exacerbate hepatic lipid accumulation. For example, certain SNPs in CD36 correlate with altered lipid profiles and increased risk of early-onset , indirectly influencing liver through dysregulated transport. Additionally, a 2025 study elucidated CD36's involvement in pathways, suggesting that functional variants could modulate the efficiency of proteolysis-targeting (PROTAC) uptake, as CD36 facilitates receptor-mediated internalization of such molecules, with implications for therapeutic delivery in variant carriers.00386-1)

Expression and Tissue Distribution

Regulation of Expression

The expression of CD36 is primarily regulated at the transcriptional level by (PPAR) family members, particularly PPARγ, which forms heterodimers with (RXR). These heterodimers bind to peroxisome proliferator response elements (PPREs) in the CD36 promoter and enhancer regions, thereby upregulating transcription in response to ligands such as long-chain s or synthetic agonists like thiazolidinediones (TZDs). For instance, oxidized (oxLDL)-derived s activate PPARγ/RXR, leading to increased CD36 mRNA and protein levels in macrophages and adipocytes, which facilitates uptake and contributes to . TZDs, such as pioglitazone, similarly enhance CD36 expression by stabilizing the PPARγ/RXR complex, promoting insulin sensitivity in metabolic tissues, though this can also drive . Post-transcriptional regulation of CD36 occurs through microRNAs (miRNAs), with miR-33 acting as a key by directly binding to the 3' (3' UTR) of CD36 mRNA, thereby inhibiting translation and promoting mRNA degradation. This downregulation limits uptake and accumulation in hepatocytes and macrophages, playing a protective role against and non-alcoholic . Antagonism of miR-33, for example via inhibitors, elevates CD36 levels and enhances reverse cholesterol transport in mouse models. In hypoxic or ischemic conditions, -inducible factor-1α (HIF-1α) induces CD36 expression by stabilizing under low oxygen levels and translocating to the , where it heterodimerizes with HIF-1β (ARNT) to bind a (HRE) in the CD36 promoter at position -558 to -555 bp. This transcriptional activation is amplified by the phosphatidylinositol 3-kinase (PI3K) pathway and , increasing CD36 mRNA up to 6-fold in retinal and corneal tissues during . Experimental evidence from ARPE-19 cell lines and models of ischemia confirms that HIF-1α knockdown abolishes this induction, highlighting its role in adaptive handling under oxygen deprivation. Epigenetic modifications, particularly histone , further modulate CD36 expression by altering accessibility at its promoter and enhancers. inhibitors like (TSA) increase H4 acetylation at the CD36 promoter in macrophages, elevating mRNA levels and oxidized LDL uptake by 2- to 3-fold. Similarly, chronic intermittent promotes H3K9 acetylation in aortic macrophages via reduced 2 (HDAC2) activity, sustaining elevated CD36 expression. These changes often interact with transcription factors like PPARγ, where acetylated histones facilitate binding to distal enhancers. Circadian rhythms influence CD36 expression in metabolic tissues such as adipose and skeletal muscle, where its mRNA levels oscillate with peaks during the active phase, driven by clock genes like Per1 and Bmal1. In adipose tissue, PPARγ acetylation coordinates these rhythms, linking CD36 to diurnal lipid storage and release, while in muscle, palmitate-induced disruptions alter CD36 transcriptomics, impairing fatty acid oxidation. CD36 deficiency in turn desynchronizes hepatic clocks via the AKT/FoxO1/Per1 pathway, indirectly affecting adipose and muscle rhythms through systemic glucose dysregulation in mouse models. Pharmacological modulators, including retinoids like 9-cis- (9-cis RA), upregulate CD36 by activating RXR, which heterodimerizes with receptors () to bind retinoic acid response elements (RAREs) near the CD36 , increasing mRNA 2.5-fold in after 5 days of exposure. Recent 2025 studies show that agonists (GLP-1RAs) reduce circulating soluble CD36 (sCD36) levels—a secreted form derived from ectodomain shedding—in patients with and diabetic , potentially by suppressing activation and , with median decreases to 195 ng/mL after 12 weeks of treatment.

Tissue and Cellular Distribution

CD36 exhibits high levels of expression in several key tissues involved in lipid metabolism and hemostasis, including the heart, skeletal muscle, adipose tissue, and platelets. Moderate expression is observed in the liver, kidney, and endothelial cells, where it contributes to local cellular functions without dominating overall tissue profiles. This distribution pattern underscores CD36's role as a versatile transmembrane protein adapted to diverse physiological contexts, with quantitative assessments showing mRNA and protein levels in heart and skeletal muscle often exceeding those in other organs under basal conditions. At the cellular level, CD36 localizes primarily to the plasma membrane in myocytes and adipocytes, facilitating direct interaction with extracellular ligands such as long-chain fatty acids. In contrast, within macrophages, CD36 is often associated with intracellular vesicles and lipid rafts, enabling rapid translocation to the cell surface upon activation. Endothelial cells display CD36 on their luminal surfaces, particularly in microvascular beds of metabolically active tissues. A soluble form of CD36, known as sCD36, circulates in and is generated primarily through proteolytic shedding by ADAM17 from the surface of cells like platelets, monocytes, endothelial cells, and adipocytes, though has also been implicated in some contexts. sCD36 levels are detectable at concentrations typically 20-100 ng/mL (depending on ) in healthy individuals and serve as a for metabolic perturbations. Developmentally, CD36 expression undergoes upregulation during , with preadipocytes showing a 10- to 20-fold increase in CD36 mRNA and protein as they differentiate into mature adipocytes, driven by factors like PPARγ. Recent studies from 2025 highlight elevated CD36 in hepatic Kupffer cells during liver diseases such as nonalcoholic steatohepatitis (NASH), where it correlates with inflammatory infiltration and lipid accumulation in the liver microenvironment.

Functions

Fatty Acid Translocation

CD36 serves as a key facilitator of long-chain fatty acid (LCFA) uptake across the plasma membrane in various cell types, particularly in metabolically active tissues such as the heart and adipose tissue. The protein's ectodomain features a hydrophobic tunnel that binds LCFAs with high affinity, guiding them from the extracellular space into the outer leaflet of the lipid bilayer. This binding promotes the flip-flop translocation of LCFAs, where the hydrophobic acyl chain diffuses across the membrane while the polar head group orients appropriately, enabling efficient transfer to the inner leaflet without the need for ATP hydrolysis in the translocation step itself. The kinetics of LCFA uptake mediated by CD36 involve high-affinity binding with a (Kd) of approximately 50 nM, followed by rapid dissociation and internalization of the CD36-LCFA complex via caveolae-dependent . This process is energy-independent for the core transmembrane flip-flop but is modulated by ATP-dependent mechanisms, such as and vesicular trafficking, which regulate CD36's sarcolemmal localization. In response to stimuli like insulin or , CD36 translocates from intracellular endosomes to the plasma membrane, increasing LCFA uptake rates by 1.3- to 1.6-fold within minutes. CD36 is essential for maintaining in myocardial and adipose tissues, where it accounts for a major fraction of LCFA influx required for energy production and storage. In cardiomyocytes, inhibition of CD36 via genetic or pharmacological agents like sulfo-N-succinimidyl oleate reduces uptake by 50% to 80% under contracting conditions, thereby limiting lipid accumulation and preserving contractile function during lipid overload. This role underscores CD36's position as a rate-limiting of cardiac utilization. Recent reviews highlight CD36 as a in cardiac , emphasizing its dual function in facilitating LCFA uptake while integrating with broader metabolic signaling pathways to prevent dysregulation in high-fat environments.

Scavenger Receptor Activity

CD36 functions as a class B receptor, facilitating the recognition and endocytic uptake of modified lipoproteins and other altered self-molecules by macrophages. It binds oxidized (oxLDL), promoting its internalization through a lipid raft-dependent pathway that does not require caveolin-1. CD36 also interacts with anionic phospholipids exposed on cell-derived microparticles and (AGEs), such as those modified by β-amyloid fibrils, leading to enhanced in phagocytic cells. These interactions underscore CD36's role in clearing potentially harmful modified and glycated proteins from the extracellular environment, preventing their accumulation in tissues. In addition to lipid scavenging, CD36 mediates the phagocytosis of apoptotic cells and certain through of (PS). For apoptotic cells, CD36 binds oxidized PS species on the outer membrane leaflet, which is essential for efficient engulfment and clearance ; this process is independent of non-oxidized PS but relies on oxidative modifications for specificity. Similarly, CD36 facilitates bacterial , as demonstrated with , where it triggers COOH-terminal domain-mediated uptake and subsequent Toll-like receptor 2/6 signaling in s. This phagocytic function extends to gram-negative bacteria like via , enhancing production and pathogen elimination without direct PS involvement in all cases. CD36's scavenger activity contributes to formation in macrophages, a hallmark of , by internalizing oxLDL and oxidized phospholipids, leading to accumulation and cellular transformation. The structural basis for its multiligand specificity lies in the extracellular domain, an antiparallel β-barrel with a hydrophobic and distinct binding sites: oxLDL and anionic phospholipids interact at residues 155–183 (involving Lys164 and Lys166), while other ligands like thrombospondin engage acidic residues in the CLESH domain (93–120). This modular architecture, stabilized by bonds, enables promiscuous binding and transport through channel-like entrances. Recent studies from 2022 to 2025 have highlighted CD36's evolving role in innate immunity, positioning it as a that senses damage-associated molecular patterns (DAMPs) like , heat shock proteins, and oxidized phospholipids, as well as pathogen-associated molecular patterns (PAMPs) such as . In CD36-overexpressing cells, DAMP exposure induces 7- to 10-fold higher IL-8 secretion, while CD36-deficient macrophages show 2- to 3-fold reduced IL-6 responses, emphasizing its pro-inflammatory signaling in sterile and infectious contexts. These findings suggest CD36 amplifies innate responses to both endogenous danger signals and microbial threats, with potential therapeutic implications for blocking DAMP/PAMP-driven using synthetic peptide inhibitors.

Adhesion and Signaling Roles

CD36 serves as a key mediator of to components, particularly and thrombospondin-1 (TSP-1). It binds directly to types I and IV, facilitating platelet and to subendothelial matrices during vascular . Similarly, CD36 interacts with the type I repeats of TSP-1, promoting in various cell types including platelets and endothelial cells. This binding is critical for stabilization, as platelet-derived TSP-1 anchors via CD36 to , enhancing platelet aggregation. A prominent function of the CD36-TSP-1 interaction is the inhibition of . The second type 1 repeat domain (TSR2) of TSP-1 binds the CLESH domain of CD36 on microvascular , triggering anti-migratory signals that suppress , tube formation, and vessel sprouting. This complex formation disrupts VEGF signaling by recruiting the SHP-1 to the CD36-VEGFR2 complex, thereby inhibiting nitric oxide-dependent responses and promoting in angiogenic . Blocking CD36 abolishes TSP-1's anti-angiogenic effects , confirming its essential role. Ligand binding to CD36 initiates intracellular signaling cascades that regulate cytoskeletal dynamics and cellular responses. Upon engagement by TSP-1 or oxidized (oxLDL), CD36 recruits Src family kinases such as Lyn and , leading to activation of MAPK pathways including JNK and p38. This signaling promotes cytoskeletal rearrangement, enhancing cell spreading while inhibiting migration in macrophages and endothelial cells. In platelets, oxLDL-induced CD36 ligation specifically activates and Lyn, phosphorylating JNK2 via MKK4, which drives shape change, aggregation, and prolonged . These pathways also contribute to vascular remodeling by stimulating vascular cell proliferation and neointima formation through JNK-dependent mechanisms. Recent studies highlight CD36's involvement in establishing through pathways intersecting with regulation. In senescent cardiomyocytes, CD36 is upregulated alongside elevated levels, contributing to hallmarks like activity and proliferative arrest. Furthermore, degradation via organizes droplets that upregulate CD36, forming a feed-forward loop that sustains (SASP) and metabolic reprogramming. CD36 signaling via Src-p38-NF-κB further drives SASP production (e.g., IL-6, IL-8) in response to inducers like amyloid-β, independent of direct modulation but amplifying the senescent state.

Clinical Significance

Infectious Diseases

CD36 serves as a key receptor for Plasmodium falciparum-infected erythrocytes in , facilitating cytoadherence to and in microvasculature, which contributes to severe complications such as cerebral . This interaction is mediated by the parasite's variant surface antigen PfEMP1, which binds specifically to CD36 on microvascular , promoting parasite survival by avoiding splenic clearance. Studies have shown that CD36 expression levels influence the extent of , with polymorphisms in the CD36 gene associated with altered susceptibility to severe outcomes. In tuberculosis, CD36 facilitates the uptake of Mycobacterium tuberculosis by macrophages through its scavenger receptor activity, particularly by mediating the internalization of surfactant lipids and oxidized lipoproteins that the pathogen exploits for intracellular growth. This uptake promotes the formation of lipid-laden foam cells within granulomas, enabling bacterial persistence and chronic infection by providing a nutrient-rich environment. CD36-mediated lipid accumulation in these structures exacerbates granuloma formation and inflammation, contributing to tissue pathology in the lungs. CD36 also binds to , aiding parasite invasion into phagocytic cells such as macrophages, where it promotes for these host cells in avirulent strains. This binding is regulated by the parasite's ROP18 , which suppresses CD36 interaction in virulent strains to evade immune detection. Regarding HIV-1, CD36 on dendritic cells contributes to viral attachment and uptake, potentially facilitating trans-infection to T cells, though its role is modulated by HIV-1 Nef protein, which downregulates CD36 expression to impair phagocytic functions. Studies from 2021 to 2025 have demonstrated that CD36 neutralization or deficiency reduces infection burden in animal models across these pathogens. For instance, CD36-deficient mice exhibit attenuated M. tuberculosis growth, decreased density, and lower bacterial loads in lungs compared to wild-type controls. Similarly, blocking CD36 engagement with T. gondii limits parasite dissemination and improves host survival by disrupting . These findings highlight CD36 as a therapeutic target, with antibody-mediated neutralization showing promise in reducing pathogen persistence without compromising essential immune responses.

Metabolic and Cardiovascular Disorders

CD36 plays a pivotal role in metabolic disorders by facilitating excessive uptake in , which promotes and contributes to during . In obese individuals and high- (HFD)-fed mice, CD36 expression is upregulated in preadipocytes and , enhancing lipid accumulation and triggering stress, , and . This process drives remodeling, with increased infiltration and pro-inflammatory cytokine secretion, such as IL-6 and TNF-α, exacerbating systemic . Studies in CD36 knockout (CD36^{-/-}) mice demonstrate protection against HFD-induced , showing reduced body weight gain, lower mass, improved glucose tolerance, and decreased fasting insulin levels compared to wild-type controls after 16 weeks on a 60% . In non-alcoholic fatty liver disease (NAFLD), CD36 overexpression in hepatocytes significantly elevates uptake, leading to , , and progression to non-alcoholic steatohepatitis (). Recent analyses indicate that CD36 mRNA and protein levels increase up to 20-fold in NAFLD patients and livers, promoting synthesis and through enhanced long-chain translocation to the plasma membrane under hyperinsulinemic conditions. Hepatocyte-specific CD36 disruption reduces hepatic content, , and in HFD models, while global CD36^{-/-} mice exhibit 60-70% lower uptake in liver , though outcomes vary by model with potential compensatory increases in . A 2025 review underscores CD36's dual role in NAFLD/, where its inhibition via palmitoylation blockade enhances β-oxidation and mitigates Kupffer cell-mediated . CD36 contributes to cardiovascular disorders, particularly , through mechanisms in renal tubular cells and vascular . In the , reduced renal CD36 expression correlates with elevated in spontaneously hypertensive rat strains, as deficient Cd36 acts as a genetic for by impairing handling and promoting sodium reabsorption via interactions with Na/K-ATPase, leading to glomerular hyperfiltration and tubular . In vascular cells (VSMCs), CD36 signaling, activated by ligands like thrombospondin-1, upregulates A to enhance and neointimal , contributing to vascular remodeling and hypertension-associated stiffness; CD36 deficiency reduces VSMC by over 75% in ApoE^{-/-} models of arterial , improving vessel distensibility. In , CD36 serves as a key scavenger receptor on macrophages, mediating oxidized (oxLDL) uptake and driving formation, a hallmark of plaque development. Binding of oxLDL to CD36 activates JNK and Vav signaling pathways, promoting lipid internalization, activation, and pro-inflammatory release, which amplify progression. Experimental evidence from ApoE^{-/-}/CD36^{-/-} double-knockout mice shows a 51-60% reduction in aortic area compared to ApoE^{-/-} controls, with transplantation confirming that CD36-null macrophages halve plaque size by limiting oxLDL scavenging. CD36 also underlies myocardial lipotoxicity in cardiovascular disease, where dysregulated translocation increases overload in cardiomyocytes, leading to triacylglycerol accumulation, , and contractile dysfunction. In diabetic and pressure-overload models, such as obese Zucker rats or transverse aortic constriction, elevated sarcolemmal CD36 (>1.5-fold) shifts metabolism toward storage, inducing and ; partial CD36 knockdown attenuates these effects, preserving energy metabolism and reducing infarct size. A 2023 review positions CD36 as a gatekeeper of cardiac , with therapeutic targeting—such as sulfo-N-succinimidyl oleate inhibition—offering potential to rebalance utilization and mitigate lipotoxic . Recent 2025 studies further demonstrate that CD36 knockdown in pressure-overload models prevents functional impairment by curbing accumulation and enhancing mitochondrial efficiency.

Cancer

CD36 plays a multifaceted role in cancer progression, often acting as a promoter of through its facilitation of and interactions within the . In various solid tumors, CD36 enhances metastatic potential by mediating the uptake of s, which provides energy for and . For instance, in , CD36 upregulation following anti-HER2 therapy increases uptake, compensating for reduced and correlating with aggressive and poor outcomes. Similarly, in models, CD36 drives the accumulation of in lipid droplets, fueling particularly under high-fat dietary conditions where saturated s are preferentially transported to support tumor cell motility. Paradoxically, CD36 can also inhibit in certain contexts by binding thrombospondin-1 (TSP-1), a process that suppresses endothelial responses to pro-angiogenic signals. This anti-angiogenic function, mediated through CD36 on vascular cells, limits tumor vascularization and in preclinical models, highlighting CD36's context-dependent effects in the tumor vasculature. Within the , CD36 facilitates metabolic crosstalk between cancer cells and tumor-associated macrophages (TAMs) via lipid transfer. Cancer cells release lipid-enriched extracellular vesicles that are selectively internalized by TAMs through CD36, promoting a pro-tumorigenic in macrophages by enhancing their and immunosuppressive functions. This interaction sustains tumor growth and immune evasion, as demonstrated in models where CD36 blockade disrupts vesicle uptake and reduces TAM toward tumor-promoting states. Recent therapeutic strategies leverage CD36 for in cancer. A 2025 study revealed that CD36 mediates the of proteolysis-targeting chimeras (PROTACs), large-molecule degraders that induce targeted protein degradation in cancer cells; modifying PROTACs to enhance CD36 binding via strategies increased cellular uptake by 7.7- to 22.3-fold, improving efficacy against diverse tumors. In (HCC), CD36 overexpression in tumor cells and associated fibroblasts correlates with advanced disease stages and poor patient prognosis, driven by enhanced lipid uptake and immunosuppressive signaling. Emerging 2025 data support CD36 blockade as an , with selective inhibitors like those targeting CD36 in HER2-positive cancers or humanized antibodies such as PLT012 showing promise in reprogramming the HCC immune landscape, reducing , and enhancing antitumor immunity when combined with standard treatments.

Immunohematological Disorders

CD36 deficiency results in the absence of platelet (GPIV), a surface protein essential for platelet function, which can lead to fetal/ (FNAIT). This condition arises when maternal anti-CD36 antibodies cross the and target fetal platelets expressing CD36, particularly in cases where the is CD36-deficient (type I deficiency) and the fetus inherits a normal CD36 from the father. The incidence of FNAIT overall is approximately 1 in 1,000 to 2,000 live births, with anti-CD36 antibodies accounting for a significant proportion—up to 25-30%—in Asian and populations where CD36 deficiency is more prevalent, though specific CD36-related cases are estimated around 1 in 5,000 in high-risk ethnic groups. Individuals with CD36 deficiency exhibit associations with coronary heart disease due to altered lipid metabolism. Studies have shown that CD36-deficient subjects have normal insulin sensitivity, though dyslipidemia may contribute to cardiovascular risk. Furthermore, CD36 deficiency is linked to an increased risk of coronary artery disease, as evidenced by a threefold higher frequency of deficiency in patients with severe coronary atherosclerosis compared to healthy controls, likely stemming from disrupted myocardial fatty acid transport and heightened susceptibility to thrombotic events. CD36 serves as the Naka antigen, and its deficiency can provoke alloimmunization leading to transfusion reactions, including platelet refractoriness and neonatal isoimmune . Anti-Naka (anti-CD36) antibodies in CD36-deficient recipients cause poor responses to platelet transfusions, with clinical manifestations ranging from mild to severe hemorrhagic complications. The prevalence of CD36 deficiency varies ethnically, occurring in less than 0.5% of Caucasians, 3-11% of Asians, and up to 8% of sub-Saharan Africans, influencing the risk of such reactions in transfusion-dependent patients from these groups. Recent 2024 research emphasizes the importance of screening CD36 variants in (HSCT) contexts, particularly regarding responses to infection. Haplotype analyses have identified specific genetic variants underlying type I CD36 deficiency, informing donor-recipient matching to mitigate alloimmunization risks during HSCT. Additionally, studies highlight how CD36 variants impair oxidation in hematopoietic stem cells during bacterial infections, underscoring the need for variant screening to predict emergency hematopoiesis efficiency and improve outcomes in immunocompromised patients post-transplant.

Emerging Pathological Roles

CD36 has been implicated in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) through enhanced lipid uptake in hepatic cells, particularly Kupffer cells. In NAFLD patients, CD36 expression is significantly upregulated in the liver, correlating with increased influx and severity. A 2025 review highlights that CD36 in Kupffer cells facilitates the of modified lipids like oxidized (oxLDL), leading to accumulation, formation, and subsequent that drives progression to non-alcoholic (NASH). Animal models support this, showing that CD36 knockout in hepatocytes reduces hepatic content and attenuates induced by high-fat diets. Recent studies have uncovered CD36's role in promoting within aging s, particularly in muscle stem cells. A 2022 investigation revealed that CD36 is highly expressed in senescent muscle stem cells and fibroadipogenic progenitors, where it amplifies the (SASP) via signaling, fostering a pro-inflammatory niche that impairs regeneration. This upregulation contributes to age-related muscle dysfunction by inducing paracrine senescence in neighboring healthy cells and reducing proliferative capacity. Inhibition of CD36, through neutralizing antibodies or , diminishes SASP factors such as IL-6 and , thereby alleviating the senescent burden, improving muscle force, and enhancing regeneration in both young and aged models. CD36's interaction with damage-associated molecular patterns (DAMPs) extends to emerging roles in autoimmune conditions and neurodegeneration, building on its established scavenging functions. In autoimmune disorders, CD36 on regulates formation and production by interacting with FcγRIIb to modulate apoptotic cell clearance; deficiency in this pathway reduces anti-DNA IgG levels and expansion in murine models of . For neurodegeneration, CD36 in binds DAMPs like amyloid-β and oxLDL, activating and ROS production, which exacerbate and amyloid deposition in models. These DAMP-mediated effects highlight CD36's potential as a bridge between sterile and neuronal damage, though therapeutic targeting remains exploratory. Despite these advances, significant gaps persist in human data on CD36 post-2020, with most evidence derived from models and limited translational studies in patients. The precise roles of CD36's bonds—beyond structural stabilization and known switches like Cys333-Cys272 for sensing—remain unclear, particularly in disease-specific contexts. Similarly, applications of proteolysis-targeting chimeras (PROTACs) against CD36 are predominantly explored in , with nascent potential in metabolic disorders like NAFLD but lacking robust non-cancer clinical validation.

Scavenger Receptor Family

CD36 is classified as a member of the class B scavenger receptor family, also referred to as the SR-B family or the CD36 superfamily, which is defined by the presence of a characteristic CD36 domain in the extracellular region. In mammals, this family comprises three principal members: scavenger receptor class B type I (SR-BI, encoded by ), CD36 (also designated as SR-B2 or SCARB3), and lysosomal integral membrane protein-2 (LIMP-2, encoded by SCARB2). These receptors share structural features, including two transmembrane domains that flank a large extracellular loop, forming a hairpin-like that facilitates anchoring and . The class B scavenger receptors exhibit multi-ligand binding capabilities, recognizing a diverse array of polyanionic and hydrophobic ligands such as modified lipoproteins, oxidized phospholipids, and microbial components, which underpin their roles in both and innate immunity. In homeostasis, they mediate the uptake and transport of s and cholesterol-rich particles, contributing to cellular and utilization; concurrently, in immune contexts, they function as receptors on , promoting clearance of apoptotic cells and pathogens. Structurally, family members display in their extracellular domains, characterized by a conserved asymmetric β-barrel fold with an intramolecular hydrophobic tunnel and a ligand-sensing featuring a three-helix bundle, despite low amino acid identity of approximately 13% across eukaryotic SR-B homologs, while mammalian members such as SR-BI and CD36 share about 30% identity. This conserved tertiary structure supports shared binding mechanisms, while functional divergence arises from variations in the apex region, such as lineage-specific expansions that influence specificity; for instance, SR-BI predominantly facilitates selective cholesteryl ester uptake from (HDL) in hepatic and steroidogenic tissues, distinct from CD36's broader involvement in long-chain translocation. Evolutionarily, the SR-B family traces its origins to the last eukaryotic common ancestor (LECA), with phylogenetic analyses identifying 279 homologs across 165 eukaryotic , indicating an ancient role as sensors that has been retained through metazoan diversification and independently lost in certain clades like streptophytes. This deep evolutionary conservation highlights the family's fundamental contributions to eukaryotic handling and host defense.

Functional Homologs

CD36, also known as fatty acid translocase (FAT), has functional homologs across species that share roles in transport and sensing, particularly in non-mammalian organisms. In insects such as , sensory neuron membrane protein-1 (Snmp-1) serves as a homolog of the CD36 family, facilitating the detection and transport of lipophilic pheromones in olfactory neurons through similar membrane association and ligand-binding mechanisms. Similarly, in the silkworm , a CD36 homolog in silk gland cells selectively mediates the uptake of specific , highlighting divergence in ligand specificity within the family for specialized handling. These insect proteins underscore CD36's conserved role in facilitating the transmembrane movement of hydrophobic molecules, adapting to sensory and nutritional contexts distinct from mammalian functions. In mammals, proteins like plasma membrane-associated fatty acid-binding protein (FABPpm) act as functional analogs to CD36 in binding and , though they differ in sequence. FABPpm enhances long-chain uptake across the plasma membrane in and other tissues, often working in concert with CD36 to increase rates additively without synergistic interaction, suggesting independent yet complementary mechanisms for translocation. This overlap emphasizes how multiple membrane proteins converge on homeostasis, with FABPpm providing an alternative pathway for extracellular-to-intracellular shuttling in energy-demanding cells. For adhesion functions, such as α5β1 exhibit overlapping roles with CD36, particularly in facilitating cell-matrix and pathogen interactions. CD36 recruits α5β1 integrin to endothelial surfaces, promoting the cytoadherence of Plasmodium falciparum-infected erythrocytes, which contributes to pathology by enhancing parasite sequestration. This cooperative adhesion mechanism illustrates how CD36 amplifies integrin-mediated binding to thrombospondin-rich matrices or infected cells, broadening cellular responses to environmental cues. In scavenging modified lipids, low-density lipoprotein receptor-related protein 1 (LRP1) functions analogously to CD36 by internalizing oxidized or nitro-modified lipoproteins in macrophages, contributing to foam cell formation in atherosclerosis. Unlike CD36, which broadly recognizes diverse oxidized lipids, LRP1 preferentially handles aggregated or protease-modified forms, yet both receptors drive inflammatory lipid uptake when dysregulated. Comparatively, CD163, another macrophage scavenger receptor, specializes in hemoglobin-haptoglobin complex clearance to prevent oxidative damage post-hemolysis, contrasting CD36's wider ligand repertoire including apoptotic cells and bacteria. This specificity in CD163 limits its role to heme detoxification, while CD36's versatility supports broader immunometabolic scavenging. Distant structural mimics of CD36's transmembrane include bacterial porins, which form β-barrel channels in outer membranes for , akin to CD36's predicted β-barrel ectodomain that accommodates hydrophobic ligands like fatty acids. Although not sequence-related, this architectural similarity enables passive of solutes across bilayers, paralleling CD36's role in non-energy-dependent flux.

References

  1. [1]
    CD36 (SR-B2) as master regulator of cellular fatty acid homeostasis
    Recent studies have disclosed CD36 to play a prominent regulatory role in cellular fatty acid metabolism in both health and disease.<|control11|><|separator|>
  2. [2]
    Dynamic role of the transmembrane glycoprotein CD36 (SR-B2) in ...
    Here, we review CD36's role in facilitating cellular long-chain fatty acid uptake across the plasma membrane, particularly in heart and skeletal muscles.
  3. [3]
    CD36 Signal Transduction in Metabolic Diseases - PubMed Central
    Jul 20, 2021 · The cluster of differentiation 36 (CD36) is a scavenger receptor present on various types of cells and has multiple biological functions ...
  4. [4]
    Molecular Basis of Human CD36 Gene Mutations
    May 15, 2007 · The mature polypeptide is 471 AA long, beginning at the AA residue immediately following the initiator methionine (4).Missing: length | Show results with:length
  5. [5]
    CD36 Gene - GeneCards | CD36 Protein | CD36 Antibody
    Protein attributes for CD36 Gene. Size: 472 amino acids; Molecular mass: 53053 Da. Protein existence level: PE1. Quaternary structure: Interacts with THBS1 and ...
  6. [6]
    Structure-Function of CD36 and Importance of Fatty Acid Signal ...
    CD36 is a scavenger receptor that functions in high affinity tissue uptake of long chain fatty acids (FA) and contributes under excessive fat supply to lipid ...
  7. [7]
    CD36, a Scavenger Receptor Involved in Immunity, Metabolism ...
    On microvascular endothelial cells, CD36 is a receptor for thrombospondin-1 and related proteins and functions as a negative regulator of angiogenesis.
  8. [8]
  9. [9]
    Comparative Studies of Vertebrate Platelet Glycoprotein 4 (CD36)
    The six conserved exoplasmic vertebrate CD36 cysteine residues participate in disulfide bridge ... Residues within the CD36 Exoplasmic Domain. Supplementary ...
  10. [10]
    The structural basis for CD36 binding by the malaria parasite - Nature
    Sep 26, 2016 · Here we describe the structure of CD36 in complex with long chain fatty acids and a CD36-binding PfEMP1 protein domain.
  11. [11]
    Now in 3D! Novel insights into CD36 structure and function - May
    The extracellular domain of CD36 contains six cysteine residues that were all reported to reside in disulfide bonds (Cys243−Cys311, Cys272−Cys333, and ...General Cd36 Structural... · Cd36 Ligands And Binding... · Cd36-Interacting Proteins...
  12. [12]
    Critical residues and motifs for homodimerization of the first ...
    May 26, 2017 · We have characterized the homo-oligomeric interaction of CD36 TM domains in membrane environments and with molecular dynamics (MD) simulations.
  13. [13]
    Molecular Drug Simulation and Experimental Validation of the CD36 ...
    Jul 28, 2023 · Structure-function of CD36 and importance of fatty acid signal transduction in fat metabolism. Pepino, Marta Yanina; Kuda, Ondrej; Samovski ...
  14. [14]
    The Human Scavenger Receptor CD36: glycosylation status and its ...
    Jun 12, 2009 · CD36 is heavily modified post-translationally by N-linked glycosylation, and 10 putative glycosylation sites situated in the large extracellular loop of the ...
  15. [15]
    Post-translational modifications of CD36 (SR-B2) - ScienceDirect.com
    CD36 has two membrane spanning regions surrounding a large extracellular loop that contains a large hydrophobic cavity traversing the entire length of the ...Missing: tertiary monomeric
  16. [16]
    CD36 is palmitoylated on both N- and C-terminal cytoplasmic tails
    Sep 13, 1996 · We confirmed that human CD36 is palmitoylated and identified cysteines 3, 7, 464, and 466 as the palmitoylation sites using a mutagenesis ...Missing: Cys3 Cys7
  17. [17]
    CD36 Is Palmitoylated on Both N- and C-terminal Cytoplasmic Tails
    We confirmed that human CD36 is palmitoylated and identified cysteines 3, 7, 464, and 466 as the palmitoylation sites using a mutagenesis approach. This result ...Missing: Cys3 Cys7
  18. [18]
    Opposite Regulation of CD36 Ubiquitination by Fatty Acids and Insulin
    Fatty acids and insulin induce opposite alterations in CD36 ubiquitination, modulating CD36 level and fatty acid uptake.
  19. [19]
    Ubiquitinated CD36 sustains insulin-stimulated Akt activation by ...
    Overexpression of WT CD36, but not a ubiquitination-defective CD36 mutant, delayed IRS1 degradation. We also found that CD36 functioned through ubiquitination ...
  20. [20]
    Post-translational modifications, a key process in CD36 function
    Other results demonstrate alterations in CD36 PTM in SHR hearts, specifically by N-glycosylation, and the importance of O-linked-β-N-acetylglucosamine for its ...
  21. [21]
    The role of CD36 in cardiovascular disease - Oxford Academic
    CD36 is a double transmembrane protein that cannot form a channel by itself to allow for fatty acids to transfer through to the inside.48 Interestingly, the ...The Role Of Cd36 In... · 3. Cd36 Protein · 4. Cd36 And Cardiovascular...
  22. [22]
    Recombinant GST/CD36 fusion proteins define a thrombospondin ...
    These data demonstrate that the region between amino acids 93 and 120 has all of the characteristics required of the TSP binding domain. Publication types.
  23. [23]
    CD36 ectodomain phosphorylation blocks thrombospondin-1 binding
    Objective: CD36 phosphorylation on its extracellular domain inhibits binding of thrombospondin-1. The mechanisms of cellular CD36 ectodomain phosphorylation and ...
  24. [24]
    CD36 associates with CD9 and integrins on human blood platelets
    Mar 15, 2001 · The membrane glycoprotein CD36 is involved in platelet aggregation, inhibition of angiogenesis, atherosclerosis, and sequestration of ...
  25. [25]
    Mapping and characterization of the binding site for specific oxidized ...
    Mar 28, 2008 · Two conserved, positively charged amino acids in the region 157-171 (lysines at positions 164 and 166) were identified as critical for oxPC(CD36) ...Missing: Lys164 Lys166
  26. [26]
    CD36 Binds Oxidized Low Density Lipoprotein (LDL) in a ...
    The site of this oxLDL binding to CD36 has been identified and recently mapped to amino acids 157–171, with critical lysines at positions 164 and 166 (13–15).
  27. [27]
    The association between CD36 and Lyn protein tyrosine kinase is ...
    CD36 in platelets has been shown physically and functionally to associate with members of the Src family of protein tyrosine kinases, Fyn, Lyn, and Yes, but the ...
  28. [28]
  29. [29]
    Entry - *173510 - CD36 ANTIGEN; CD36 - OMIM - (OMIM.ORG)
    ► Mapping. Fernandez-Ruiz et al. (1993) mapped the human CD36 gene to chromosome 7q11. 2 by fluorescence in situ hybridization.
  30. [30]
    Molecular Basis of Human CD36 Gene Mutations - PMC
    The CD36 gene is located on chromosome 7 q11.2 and is encoded by 15 exons. Defective CD36 is a likely candidate gene for impaired fatty acid metabolism, glucose ...
  31. [31]
    Comparative Studies of Vertebrate Platelet Glycoprotein 4 (CD36)
    The human CD36 gene contained transcription factor binding sites (including PPARG and PPARA) contributing to a high gene expression level (6.6 times average).Table 2 · 3.1. Vertebrate Cd36 Gene... · Supplementary Table 1
  32. [32]
    Polymorphism rs1761667 in the CD36 Gene Is Associated to ... - NIH
    Dec 6, 2017 · The polymorphisms of CD36 gene, rs1761667, rs1527483, and rs3840546, have been associated with taste perception to and preference for fat ( ...
  33. [33]
    Associations of CD36 rs1761667 with Fat and Umami Food ... - MDPI
    The CD36 single-nucleotide polymorphism (SNP) rs1761667 (G>A) has been associated with a reduced protein expression [6,7], fat taste hyposensitivity, high ...
  34. [34]
    The A allele of cluster of differentiation 36 (CD36) SNP 1761667 ...
    Mar 30, 2015 · The A allele of cluster of differentiation 36 (CD36) SNP 1761667 associates with decreased lipid taste perception in obese Tunisian women.
  35. [35]
    Preliminary studies on CD36 gene in type 2 diabetic patients - LWW
    Two important mutations in CD36 gene responsible for CD36 deficiency are SNP T/C at nt 478 of CD36 (Exon 4) and a dinucleotide deletion (delAC in Exon 5)27 –29.
  36. [36]
    CD36 gene variants and their clinical relevance: a narrative review
    This article aimed to describe CD36 (SR-B2) gene variants and their clinical relevance by referring to existing related literature and electric database.
  37. [37]
    NM_001001548.3(CD36):c.268C>T (p.Pro90Ser) AND Platelet-type ...
    268C>T (p.Pro90Ser) missense variant has been identified in at least 18 individuals with platelet glycoprotein IV deficiency, including in a homozygous ...Missing: ligand binding
  38. [38]
    Impacts of CD36 Variants on Plasma Lipid Levels and the Risk of ...
    Recent studies have indicated that cluster of differentiation 36 (CD36) is closely linked to dyslipidemia and early-onset coronary artery disease (EOCAD).
  39. [39]
    The CD36-PPARγ Pathway in Metabolic Disorders - PMC
    May 21, 2018 · The CD36-PPARγ pathway appears to be functional in hepatocytes with the activation of PPARγ by hexarelin, which identified Insig-1 and Insig-2 ...Missing: Sp1 | Show results with:Sp1
  40. [40]
  41. [41]
    Antagonism of miR-33 in mice promotes reverse cholesterol ... - JCI
    We demonstrated herein that inhibition of miR-33 expression effectively raises circulating HDL, increases RCT, and decreases atherosclerotic plaque size.
  42. [42]
  43. [43]
    Hypoxia Up-regulates CD36 Expression and Function via Hypoxia ...
    Our findings indicate a novel mechanism by which hypoxia induces CD36 expression via activation of HIF-1 and the phosphatidylinositol 3-kinase pathway.
  44. [44]
  45. [45]
    CD 36: Focus on Epigenetic and Post-Transcriptional Regulation - NIH
    CD36 is a transmembrane protein involved in fatty acid translocation, scavenging for oxidized fatty acids acting as a receptor for adhesion molecules.Missing: Sp1 | Show results with:Sp1
  46. [46]
  47. [47]
  48. [48]
    CD36 regulates diurnal glucose metabolism and hepatic clock to ...
    Mar 29, 2023 · As a key fatty acid transporter, the interaction between the clock component CD36 ... adipose plasticity and metabolic rhythms. Adv. Sci ...
  49. [49]
  50. [50]
    Palmitate impairs circadian transcriptomics in muscle cells through ...
    Oct 27, 2022 · Men and women with obesity present altered mRNA expression of core clock genes in skeletal muscle, blood cells, and visceral adipose tissue ( ...
  51. [51]
  52. [52]
    The association of soluble cluster of differentiation 36 with metabolic ...
    Jun 12, 2023 · A cluster of differentiation 36 (CD36), also known as fatty acid translocase, plays an important role in developing and progressing metabolic diseases.
  53. [53]
    CD36, a signaling receptor and fatty acid transporter that regulates ...
    CD36 functions as both a signaling receptor and fatty acid transporter in various immune and non-immune cells. This review summarizes how its dual functions ...Missing: horseshoe- shaped<|separator|>
  54. [54]
    The role of CD36 in cardiovascular disease - PMC - NIH
    CD36 is expressed in various tissues, including endothelial cells, cardiac muscle cells, renal tubular epithelial cells, liver cells, adipocytes, platelets ...
  55. [55]
    CD36 as a gatekeeper of myocardial lipid metabolism and ...
    CD36 facilitates the cellular uptake of long-chain fatty acids, mediates lipid signaling, and regulates storage and oxidation of lipids in various tissues.
  56. [56]
    CD36, a signaling receptor and fatty acid transporter that regulates ...
    Apr 19, 2022 · CD36 is a type 2 cell surface scavenger receptor widely expressed in many immune and non-immune cells. It functions as both a signaling receptor responding to ...Missing: horseshoe- shaped
  57. [57]
    Endothelial cell CD36 optimizes tissue fatty acid uptake - JCI
    The data reveal that CD36-mediated FA transport across ECs limits tissue FA uptake and that its loss leads to metabolic effects in parenchymal cells.
  58. [58]
  59. [59]
    The association of soluble cluster of differentiation 36 with metabolic ...
    Jun 12, 2023 · Soluble CD36 (sCD36), a circulating form of CD36, is identified in human plasma. Current studies have demonstrated that sCD36 is an early ...Missing: alternative | Show results with:alternative
  60. [60]
    CD36 promotes adipocyte differentiation and adipogenesis - PubMed
    CD36 promotes adipocyte differentiation and adipogenesis. Silencing CD36 impairs differentiation and reduces fat pad formation. CD36 promotes adipogenesis in ...
  61. [61]
    CD36 in liver diseases - Hepatology Communications
    Accumulating evidence suggests that CD36 plays a complex role in the development of nonalcoholic simple fatty liver disease and NASH and contributes to the ...
  62. [62]
    CD36 (SR-B2) as master regulator of cellular fatty acid homeostasis
    Transmembrane translocation ('flip-flop') of single fatty acids. 6 ... The central ectodomain of these receptors forms a hydrophobic tunnel structure ...
  63. [63]
    CD36 facilitates fatty acid uptake by dynamic palmitoylation ... - Nature
    Sep 21, 2020 · Here, we show that FAs trigger caveolae-dependent CD36 internalization, which in turn delivers FAs into adipocytes.
  64. [64]
    Oxidized phosphatidylserine–CD36 interactions play an essential ...
    Nov 13, 2006 · Multiple distinct binding sites on CD36 are reported to facilitate its broad ligand specificity and functions. CD36 interaction with oxLDL is ...
  65. [65]
    Response to Staphylococcus aureus requires CD36-mediated ...
    Response to S. aureus requires CD36-mediated phagocytosis triggered by the COOH-terminal cytoplasmic domain, which initiates TLR2/6 signaling.
  66. [66]
    Role of human CD36 in bacterial recognition, phagocytosis, and ...
    Nov 15, 2008 · These results indicate that CD36 functions as a phagocytic receptor for a variety of bacteria and mediates signaling induced by gram-negative ...
  67. [67]
    CD36 Provides Host Protection Against Klebsiella pneumoniae ...
    We demonstrate that CD36 enhances LPS responsiveness to K. pneumoniae to increase downstream cytokine production and macrophage phagocytosis that is independent ...
  68. [68]
    Class B Scavenger Receptor CD36 as a Potential Therapeutic ...
    Our findings demonstrate that CD36 is a PRR contributing to the innate immune response via mediating DAMP-induced inflammatory signaling.Missing: 2023 | Show results with:2023
  69. [69]
    Hidden features: CD36/SR-B2, a master regulator of macrophage ...
    CD36 plays a key role in innate immunity against infectious diseases by recognizing infected host cells due to PAMPs and through recognition of alterations ...
  70. [70]
  71. [71]
    Analysis of CD36 binding domains: ligand specificity controlled by ...
    The protein CD36 is a membrane receptor for thrombospondin (TSP), malaria-infected erythrocytes, and collagen. Three functional sequences were identified ...
  72. [72]
    Identification of SVTCG in thrombospondin as the ... - PubMed
    Thrombospondin (TSP) binds to its cellular receptor, CD36 (glycoprotein IV or IIIb), and participates in many adhesive cell interactions.
  73. [73]
    Supporting roles of platelet thrombospondin-1 and CD36 ... - PubMed
    Conclusions: These results demonstrate an anchoring role of platelet-released TSP1 via CD36 in platelet adhesion and collagen-dependent thrombus stabilization.
  74. [74]
    Molecular basis of anti-angiogenic thrombospondin-1 type 1 repeat ...
    We show that only the second TSR domain (TSR2) of TSP-1 binds to the CD36 CLESH domain to inhibit MVEC angiogenic functions. We propose and validate key ...
  75. [75]
    Thrombospondin-1 modulates VEGF signaling via CD36 by ...
    Sep 5, 2013 · TSP-1 binding to CD36 recruits SHP-1 to CD36-VEGFR2 complex ... TSP-1 was identified as an angiogenesis inhibitor more than 20 years ago.Abstract · Introduction · Results · Discussion
  76. [76]
    Thrombospondin-1 inhibits endothelial cell responses to nitric oxide ...
    CD36 is one of several endothelial cell TSP1 receptors and is necessary for its activity to inhibit microvascular endothelial cell chemotaxis (29). CD36 is also ...
  77. [77]
    CD36 mediates the In vitro inhibitory effects of thrombospondin-1 on ...
    Aug 11, 1997 · Thrombospondin-1 inhibits endothelial cell migration, and this inhibition is mediated by CD36, a transmembrane glycoprotein. Blocking CD36 ...
  78. [78]
    Mechanisms of Cell Signaling by the Scavenger Receptor CD36 - NIH
    CD36 independently binds three major classes of extracellular ligands and effects different cellular functions in a tissue and cell-specific context. It was ...
  79. [79]
    A Specific CD36-Dependent Signaling Pathway Is Required for ...
    Jun 20, 2008 · These studies suggest a context-dependent mechanism for CD36 signaling involving specific src and MAP kinases. In platelet biology, MAP kinases ...
  80. [80]
    CD36 Enhances Vascular Smooth Muscle Cell Proliferation and ...
    Dec 20, 2018 · Our previous work revealed that engagement of CD36 ligands in platelets and macrophages induced JNK activation via Fyn and Lyn. We thus ...
  81. [81]
    pivotal role of the CD36 receptor in AC16 human cardiomyocytes
    Jul 1, 2025 · Senescent differentiated cardiomyocytes exhibit hallmarks of cellular senescence, including increased β-galactosidase activity and elevated p53 ...
  82. [82]
    Lipid Droplet‐Organized MDM2‐Mediated P53 Degradation: A ...
    Jun 5, 2025 · Degraded p53 releases the RPS3A-C/EBPβ complex, upregulating LD-promoting factors such as CD36 to establish a cell-autonomous feed-forward loop.
  83. [83]
    CD36 initiates the secretory phenotype during the establishment of ...
    Upon Aβ or oxLDL binding, CD36 stimulates MAPK signaling through Src family kinase activation, leading to the activation of NF‐κB and subsequent cytokine ...Cd36 Is Induced In Multiple... · Long‐term Cd36 Expression... · Cd36‐dependent Secretory...
  84. [84]
    A human 88-kD membrane glycoprotein (CD36) functions in vitro as ...
    A human 88-kD membrane glycoprotein (CD36) functions in vitro as a receptor for a cytoadherence ligand on Plasmodium falciparum-infected erythrocytes. J Clin ...
  85. [85]
    CD36 peptides that block cytoadherence define the ... - PubMed - NIH
    Sep 15, 1999 · The scavenger receptor, CD36, is a major host receptor for PE adherence. PE adhesion to CD36 is mediated by the malarial variant antigen, P.
  86. [86]
    Identification of a platelet membrane glycoprotein as a falciparum ...
    A leukocyte differentiation antigen (CD36) was previously suggested to have a role in sequestration of malaria-infected erythrocytes.
  87. [87]
    CD36-mediated uptake of surfactant lipids by human macrophages ...
    Dec 15, 2017 · We provide evidence that CD36 mediates surfactant lipid uptake by human macrophages and that M.tb exploits this function for growth.
  88. [88]
    The function of CD36 in Mycobacterium tuberculosis infection - PMC
    May 31, 2024 · CD36 plays critical roles in the process of lipid metabolism, inflammatory response and immune process caused by Mycobacterium tuberculosis infection.
  89. [89]
    CD36 Mediates Phagocyte Tropism and Avirulence of Toxoplasma ...
    Aug 16, 2021 · In this study, we present evidence that avirulence in Toxoplasma requires parasite engagement of the scavenger receptor CD36.Missing: invasion | Show results with:invasion
  90. [90]
    CD36 mediates phagocyte tropism and avirulence of Toxoplasma ...
    We present evidence that avirulence in Toxoplasma requires parasite engagement of the scavenger receptor CD36. CD36 promotes macrophage tropism.
  91. [91]
    HIV-1 Nef Impairs Key Functional Activities in Human Macrophages ...
    The present work shows that HIV-1 Nef protein may have a role in the strategies elaborated by HIV-1 to alter pathogen disease outcomes, by modulating CD36 ...Missing: attachment | Show results with:attachment
  92. [92]
    Scavenger Receptor CD36 Expression Contributes to Adipose ...
    Compared to WT mice, CD36 KO mice fed a high fat diet exhibited reduced adiposity and adipose tissue inflammation, with decreased adipocyte cell death, pro- ...Missing: acetylation | Show results with:acetylation
  93. [93]
    Obesity induces preadipocyte CD36 expression promoting ...
    Our data support that the abnormal upregulation of CD36 in preadipocytes may contribute to the development of adipose tissue inflammation. CD36/Fyn/IP3R1- ...
  94. [94]
  95. [95]
    Identification of renal Cd36 as a determinant of blood pressure and ...
    Deficient renal expression of Cd36 encoding fatty acid translocase as a genetically determined risk factor for spontaneous hypertension.Missing: sodium reabsorption<|separator|>
  96. [96]
    CD36, a scavenger receptor implicated in atherosclerosis - Nature
    Jun 6, 2014 · CD36 is a transmembrane receptor composed of two transmembrane domains, two very short cytoplasmic domains and a large glycosylated ...Cd36 Structure And Ligands · Macrophage Cd36 Mediates... · Cd36: A Signaling Molecule...Missing: shape | Show results with:shape<|separator|>
  97. [97]
    CD36 knockdown attenuates pressure overload-induced cardiac ...
    Feb 18, 2025 · This study aims to investigate the feasibility of CD36 partially knockdown in the prevention of cardiac lipotoxicity and functional impairment in pressure ...
  98. [98]
    Suppressing fatty acid uptake has therapeutic effects in preclinical ...
    Feb 6, 2019 · Silencing of CD36 reduces fatty acid uptake and attenuates cancer aggressiveness in prostate cancer cells. The increase in fatty acid uptake ...
  99. [99]
    CD36 tango in cancer: signaling pathways and functions
    Macrophages do not express αvβ5 integrin on the surface, and their surface CD36, in coordination with the entire αvβ3 integrin protein, phagocytoses apoptotic ...
  100. [100]
    CD36 Mediates the In Vitro Inhibitory Effects of Thrombospondin-1 ...
    To determine if an interaction between CD36 and TSP-1 could block the antiangiogenic activity of TSP-1, soluble GST–CD36 fusion proteins were tested for their ...
  101. [101]
    CD36-mediated metabolic crosstalk between tumor cells and ...
    Oct 2, 2022 · We find that the scavenger receptor CD36 is upregulated in metastasis-associated macrophages (MAMs) and deletion of CD36 in MAMs attenuates liver metastasis in ...Missing: acetylation | Show results with:acetylation
  102. [102]
    CD36-mediated endocytosis of proteolysis-targeting chimeras
    We discovered that the membrane cluster of differentiation 36 (CD36) binds to and facilitates the uptake and efficacy of diverse PROTACs.Missing: variants 2024
  103. [103]
    Role of CD36 in cancer progression, stemness, and targeting
    CD36 is highly expressed in diverse tumor types and its expression correlates with advanced stages, poor prognosis, and reduced survival.<|control11|><|separator|>
  104. [104]
    Identification of new selective CD36 inhibitors to potentiate HER2 ...
    Aug 6, 2025 · The crystal structure of CD36 was modified by adding hydrogen atoms and Kollman charges (AutoDockTools and Python Molecular Viewer; https://ccsb ...
  105. [105]
    PLT012, a Humanized CD36-Blocking Antibody, Is Effective for ...
    PLT012 rejuvenates antitumor immunity by targeting metabolic pathways to reprogram the immune landscape of liver cancer and liver metastasis, with potential to ...Missing: prognosis | Show results with:prognosis
  106. [106]
    Fetal/neonatal alloimmune thrombocytopenia due to anti-CD36 ...
    Oct 13, 2017 · This case reemphasizes the role of anti-CD36 isoantibodies on the pathomechanism of fetal/neonatal alloimmune thrombocytopenia.
  107. [107]
    Fighting anti-CD36–mediated FNAIT | Blood - ASH Publications
    Nov 4, 2021 · Whereas CD36 deficiency is rare in White populations, it is more frequent in African and Asian populations2,3 (type 1 reported in <2%), ...
  108. [108]
    Successful prenatal therapy for anti-CD36-mediated severe FNAIT ...
    Nov 4, 2021 · Recent studies have shown that maternal anti-CD36 antibodies represent a frequent cause of fetal/neonatal alloimmune thrombocytopenia ...
  109. [109]
    Insulin Sensitivity and Lipid Metabolism in Human CD36 Deficiency
    Feb 1, 2003 · Lipid abnormalities in CD36 deficiency may partly depend on the presence of diabetes, and increased levels of triglycerides and HDL cholesterol ...
  110. [110]
    Patients with CD36 deficiency are associated with ... - PubMed
    Three representative CD36-D cases demonstrated severe CAD and atherosclerosis. The frequency of CD36-D was three times higher in CAD patients than in healthy ...Missing: heart | Show results with:heart
  111. [111]
    [PDF] © SIMTI Servizi Srl - Blood Transfusion
    It has been shown that the Naka antigen is located on GPIV or CD36 and that individuals who do not express CD36 on their platelets can probably be immunized ...
  112. [112]
    High-Throughput CD36 Phenotyping on Human Platelets Based on ...
    Jun 27, 2023 · Background: CD36 deficiency is closely associated with fetal/neonatal alloimmune thrombocytopenia, platelet transfusion refractoriness, and ...Abstract · Introduction · Materials and Methods · Discussion
  113. [113]
    The polymorphism analysis for CD36 among platelet donors - PMC
    Apr 12, 2024 · Africans exhibit a higher incidence at 7.70%, while Asians range from 1.68 to 8.20%, and Caucasians have a lower prevalence at less than 0.40%.
  114. [114]
    Haplotypes analysis reveals the genetic basis of type I CD36 ...
    Oct 14, 2024 · This delineates the human CD36 gene's exon-intron structure, with long-read PCR amplicon locations and the exonic locations. Untranslated ...Missing: organization | Show results with:organization
  115. [115]
    Allogeneic Hematopoietic Stem Cell Transplantation From a CD36+ ...
    Dec 1, 2024 · Allogeneic hematopoietic stem cell transplantation from a CD36+ donor in an immunized patient with type I hereditary CD36 deficiency.Missing: screening responses infection
  116. [116]
    The regulatory role of CD36 in hematopoiesis beyond fatty acid uptake
    Feb 15, 2024 · CD36 also mediates free fatty acid transportation to hematopoietic stem cells in response to infections.
  117. [117]
    Senescence atlas reveals an aged-like inflamed niche that blunts ...
    Dec 21, 2022 · Senescent cells create an aged-like inflamed niche that mirrors inflammation associated with ageing (inflammageing 4 ) and arrests stem cell proliferation and ...
  118. [118]
    Unveiling the hidden role of the interaction between CD36 and FcγRIIb
    May 18, 2024 · Our data indicate that CD36 in B cells is a critical regulator of autoimmunity. The interaction of CD36-FcγRIIb has the potential to serve as a therapeutic ...Missing: FRET | Show results with:FRET
  119. [119]
    Role of CD36 in central nervous system diseases - PMC
    CD36 is involved in mediating multiple signaling pathways that play an essential part in the pro-inflammatory response, including the NF-κB signaling pathway.
  120. [120]
    Fatty-acid receptor CD36 functions as a hydrogen sulfide-targeted ...
    Fatty-acid receptor CD36 functions as a hydrogen sulfide-targeted receptor with its Cys333-Cys272 disulfide bond serving as a specific molecular switch to ...
  121. [121]
    CD36-mediated endocytosis of proteolysis-targeting chimeras
    Jun 12, 2025 · We discovered that the membrane cluster of differentiation 36 (CD36) binds to and facilitates the uptake and efficacy of diverse PROTACs.
  122. [122]
    Phylogenetic and Protein Structure Analyses Provide Insight into the ...
    The cluster of differentiation 36 (CD36) domain defines the characteristic ectodomain associated with class B scavenger receptor (SR-B) proteins.
  123. [123]
    A Consensus Definitive Classification of Scavenger Receptors ... - NIH
    Class B scavenger receptors include receptors currently known as SR-BI, and its alternatively spliced isoform SR-BII, as well as CD36 and LIMP2. Receptors ...
  124. [124]
    Lysosomal integral membrane protein-2 (LIMP-2/SCARB2 ... - Nature
    Aug 6, 2019 · LIMP-2 is the only endomembrane member of the CD36 superfamily of scavenger receptors, which also include the plasmalemmal SR-BI and CD36.
  125. [125]
    CD36: a class B scavenger receptor involved in angiogenesis ... - JCI
    There are two CD36-family homologs in Drosophila, Croquemort, and epithelial membrane protein (emp) (7, 8). Croquemort (“catcher of death”) has 23% homology to ...Missing: mammals | Show results with:mammals
  126. [126]
    SR-BI/CD36 Chimeric Receptors Define Extracellular Subdomains ...
    The C-terminal half of the EC domain of SR-BI contains cysteine (Cys) residues that are involved in intramolecular disulfide bonds (43, 64) that most likely ...Hdl Labeling, Cell... · Design Of Sr-Bi/cd36... · Sr-Bi/cd36 Chimeric...
  127. [127]
    Identification of Scavenger Receptor SR-BI as a High Density ...
    SR-BI binds HDL with high affinity, is expressed primarily in liver and nonplacental steroidogenic tissues, and mediates selective cholesterol uptake.
  128. [128]
    Phylogenetic and Protein Structure Analyses Provide Insight into the ...
    CD36 Disulfide Bridge Conservation in Metazoa. Three pairs of cysteine residues forming intramolecular disulfide bridges have been identified in the CD36 ...Results · Cd36 Disulfide Bridge... · Cd36 Domain Phylogenetic...
  129. [129]
    Snmp-1, a novel membrane protein of olfactory neurons of ... - PubMed
    Sequence analysis suggests Snmp-1 is homologous with the CD36 protein family, a phylogenetically diverse family of receptor-like membrane proteins. CD36 family ...
  130. [130]
    CD36 homolog divergence is responsible for the selectivity of ...
    Nov 16, 2012 · Our findings indicate that divergence of genes in the CD36 family determines the selectivity of carotenoid species uptake by silk gland tissue.
  131. [131]
    Evidence for concerted action of FAT/CD36 and FABPpm ... - PubMed
    It appears based on a number of experiments that FAT/CD36 and FABPpm may collaborate to increase the rates of fatty acid transport, as these proteins co- ...
  132. [132]
    CD36 recruits α₅β₁ integrin to promote cytoadherence of P ...
    In this study, we addressed whether CD36 supports IRBC adhesion as part of an assembly of membrane receptors.Missing: α5β1 | Show results with:α5β1
  133. [133]
    Nitro-fatty acid modulates expression of CD36 and LRP1 scavenger ...
    Inflammatory profile and uncontrolled modified-lipoprotein uptake through scavenger receptors (SRs) such as CD36, LRP1 and SRA-1 in macrophages drive to ...
  134. [134]
    CD163 is the macrophage scavenger receptor for native and ...
    These results identify CD163 as a scavenger receptor for native Hb and small-molecular-weight Hb-based blood substitutes after Hp depletion.