Fact-checked by Grok 2 weeks ago

APOBEC

The APOBEC (Apolipoprotein B mRNA Editing Catalytic Polypeptide-like) family comprises a group of zinc-dependent cytidine deaminases that catalyze the deamination of cytosine to uracil in both RNA and single-stranded DNA substrates, thereby introducing targeted mutations essential for physiological processes such as lipid metabolism, immune diversification, and antiviral defense. In humans, the family consists of 11 primary members—AID (activation-induced deaminase), APOBEC1, APOBEC2, APOBEC3A through APOBEC3H, and APOBEC4—each sharing a conserved catalytic domain but exhibiting divergent substrate specificities and functions due to variations in flanking loops and oligomeric states. Originally identified through APOBEC1's role in editing apolipoprotein B mRNA to produce a truncated isoform in the intestine, the family's broader significance emerged with discoveries of AID's involvement in somatic hypermutation and class-switch recombination in B cells, enabling antibody diversity during adaptive immunity. The APOBEC3 subfamily, expanded through gene duplications in primates approximately 200 million years ago, functions primarily as restriction factors against retroviruses like HIV-1 and endogenous retroelements, hypermutating viral genomes to inhibit replication—though viral proteins such as HIV Vif counteract this by promoting APOBEC3 degradation. Beyond immunity, dysregulated APOBEC activity contributes to ; for instance, APOBEC3A and APOBEC3B generate C-to-T at TC dinucleotides, driving genomic instability in cancers such as , , and tumors, where they are overexpressed and linked to poor . APOBEC2 and APOBEC4 have more enigmatic roles, potentially in muscle differentiation and embryogenesis, respectively, with limited deaminase activity. Evolutionarily, the family traces back to jawless around 500 million years ago, underscoring its ancient role in innate immunity, while regulatory mechanisms—including , binding, and subcellular localization—fine-tune activity to prevent off-target .

Introduction

Definition and Discovery

The APOBEC (apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like) family comprises a group of zinc-dependent cytidine deaminases that catalyze the hydrolytic deamination of cytosine to uracil in single-stranded DNA (ssDNA) or RNA, thereby converting C to U in nucleic acids. These enzymes are primarily known for their roles in RNA editing and DNA mutagenesis, with the family encompassing 11 members in humans, including APOBEC1, activation-induced cytidine deaminase (AID), and the APOBEC3 cluster (A3A–H). The nomenclature originates from the founding member, APOBEC1, which was identified for its specific function in editing apolipoprotein B (ApoB) mRNA, and has since evolved to include structurally related proteins like AID despite functional diversification. The discovery of the APOBEC family began with observations of post-transcriptional mRNA editing in the late 1980s. In 1987, two independent studies identified that the expression of two ApoB isoforms—ApoB100 in the liver and ApoB48 in the intestine—arose from the same gene through tissue-specific rather than or genomic differences. This editing event was later pinpointed as a precise C-to-U change at 6666 in the ApoB mRNA, converting a codon to a . In the early 1990s, the catalytic enzyme responsible was isolated and characterized. In 1993, Teng et al. cloned the rat intestinal enzyme, initially termed REPR (RNA editing protein), which was shown to mediate the ApoB mRNA deamination as part of a multiprotein complex. By 1995, this enzyme was formally named APOBEC1, establishing it as the first member of what would become a broader family of cytidine deaminases. The family expanded significantly in the early 2000s with the identification of additional members and their diverse functions. In 1999–2000, AID was discovered by Muramatsu et al. as essential for antibody diversification through somatic hypermutation and class-switch recombination in B cells. Concurrently, the APOBEC3 cluster was recognized in 2002 by Jarmuz et al. as a tandem array of seven genes (A3A–H) on human chromosome 22, evolving from gene duplication events. Key contributions from researchers like Reuben Harris and Michael Neuberger further illuminated the antiviral roles of APOBEC3 proteins; for instance, in 2002, Sheehy et al. demonstrated that APOBEC3G inhibits HIV-1 replication by deaminating viral cDNA, marking a pivotal shift toward understanding the family's innate immune functions.

Evolutionary Origins

The APOBEC family of cytidine deaminases traces its origins to the zinc-dependent deaminase superfamily, with ancestral forms resembling bacterial cytidine deaminases that likely emerged at the dawn of evolution around 500 million years ago during the period. Phylogenetic analyses indicate that and APOBEC2 represent the most ancient members, conserved across jawless vertebrates such as the and jawed vertebrates including cartilaginous like the . These primitive enzymes share a conserved catalytic domain responsible for deaminating to in s, a function adapted from earlier tRNA adenosine deaminases in the deaminase superfamily. Homologs of AID have been identified in bony , such as the pufferfish and , demonstrating broad conservation in early vertebrates where the family likely played roles in basic nucleic acid editing before specializing in immunity. Gene duplication events drove the expansion of the family in higher vertebrates, with arising through duplication of an -like ancestor in early mammals, followed by the emergence of the cluster in placental mammals approximately 100 million years ago. In , this cluster underwent rapid tandem duplications, resulting in seven paralogs (–H) on , an adaptation reflecting strong positive selection for antiviral defense. reveals varying repertoire sizes across species: while retain only and -like genes, mammals show diverse expansions, such as the merger of genes into fewer functional copies in , pigs, and . Pseudogenization events, including variants of in some populations, highlight ongoing evolutionary dynamics, potentially linked to relaxed selective pressures in certain lineages. The evolution of APOBEC genes has been profoundly shaped by host-pathogen co-evolution, particularly with retroviruses, where duplications and sequence diversification enhanced the family's ability to induce hypermutation in viral genomes. Evidence from genomes shows signatures of adaptive evolution in APOBEC3G, with accelerated rates of nonsynonymous substitutions indicating arms-race dynamics against ancient retroviral integrations like endogenous retroviruses (ERVs) and LINE-1 elements. This co-evolutionary pressure explains the expanded APOBEC3 repertoire in compared to more primitive forms in , underscoring the family's transition from general editors to specialized innate immune effectors.

Family Members

Classification

The APOBEC family of cytidine deaminases is systematically classified into four subfamilies—AID/APOBEC1, APOBEC2, APOBEC3, and APOBEC4—primarily based on within their conserved zinc-dependent deaminase (ZDD) domains, which contain the catalytic zinc-binding . This classification also considers structural features such as the number of ZDD domains (single versus double) and chromosomal locations. The AID/APOBEC1 subfamily includes (AID) and APOBEC1, both featuring a single ZDD domain and located on 12p13. APOBEC2 possesses a single ZDD domain and is situated on 6p21, while APOBEC4 similarly has a single ZDD domain on 1q25.3. In contrast, the APOBEC3 subfamily, which encompasses seven isoforms (APOBEC3A through APOBEC3H), exhibits variability in domain architecture: single ZDD domains in APOBEC3A, APOBEC3C, and APOBEC3H, and double ZDD domains in APOBEC3B, APOBEC3D, APOBEC3F, and APOBEC3G; these genes form a cluster on 22q13.1. The of the APOBEC family reflects evolutionary processes like duplications, with the APOBEC3 arising from tandem duplications that expanded the subfamily in . This locus also includes pseudogenes, contributing to the family's complexity, and displays copy number variations across individuals and populations. A notable example is the APOBEC3B deletion polymorphism, a 29.5-kb deletion that removes most of the APOBEC3B and fuses it with the 3' end of APOBEC3A; its frequency varies significantly by ancestry, being rare in (0.9%) and (6%) populations but more prevalent in East Asian (36.9%) and Amerindian (57.7%) groups. Expression patterns of APOBEC family members are predominantly tissue-specific, aligning with their genomic clustering and evolutionary divergence. APOBEC1 is primarily expressed in the and, in some , the liver. APOBEC2 shows broad but low-level expression across various tissues. The APOBEC3 subfamily is mainly active in immune cells, including monocytes, macrophages, and T lymphocytes, with inducible expression in hepatocytes upon interferon-alpha stimulation. APOBEC4 expression is restricted to testis and .

Specific Isoforms

APOBEC1, the founding member of the APOBEC family, is a single-domain deaminase primarily expressed in the of the , where it edits (apoB) mRNA to produce a truncated apoB48 isoform involved in regulation. This enzyme requires auxiliary factors such as APOBEC1 complementation factor (ACF) for efficient and localizes to both the and . Activation-induced cytidine deaminase (AID), encoded by the AICDA gene on 12p13, is a single-domain selectively expressed in activated B lymphocytes within germinal centers. AID shuttles between the and and is crucial for processes enabling diversification, distinguishing it from other family members by its specialized role in B-cell . APOBEC2, located on 6p21, is predominantly expressed in skeletal and tissues and features a single zinc-dependent deaminase-like domain but exhibits no detectable cytidine deaminase activity in mammals. Its expression is upregulated during muscle , suggesting non-catalytic roles in and tissue maintenance, though its precise functions remain poorly understood. The APOBEC3 subfamily comprises seven closely related genes (APOBEC3A through APOBEC3H) tandemly arrayed on human chromosome 22q13.1, with expression primarily in hematopoietic cells including monocytes, macrophages, dendritic cells, and lymphocytes. These isoforms vary in domain architecture—single-domain for A3A, A3C, and A3H; double-domain for A3B, A3D, A3F, and A3G—and display tissue-specific nuances, such as A3A in peripheral blood mononuclear cells and A3G in + T cells. Among them, APOBEC3G is notable for its ability to form dimers via its N-terminal domain, a property linked to its packaging into virions for restriction. APOBEC3B features a prevalent deletion polymorphism that fuses its 3' end to APOBEC3A, resulting in loss of the full-length protein and altered expression levels associated with cancer susceptibility. APOBEC3H exists in seven major s in human populations, with variants differing in protein stability and subcellular localization, such as the stable haplotype II form that localizes to the . APOBEC4, mapped to chromosome 1q25.3, is selectively expressed in the testis and shows a divergent catalytic motif with no verified deaminase activity in functional assays. Its biological role remains largely enigmatic, with limited studies indicating potential involvement in spermatogenesis but no clear mechanistic insights.

Structure

Domain Architecture

The APOBEC family of enzymes is characterized by a conserved zinc-dependent deaminase domain (ZDD) that forms the core of their catalytic machinery. This domain features a hallmark HAE motif, where the histidine (H) and cysteine (C) residues coordinate a zinc ion essential for substrate binding, while the glutamate (E) acts as a proton shuttle during catalysis. The ZDD adopts a compact α-β-α sandwich fold, stabilized by a five-stranded β-sheet flanked by helices, which is ubiquitous across all 11 human APOBEC members. APOBEC proteins exhibit variation in domain organization, with some isoforms containing a single ZDD and others featuring tandem domains. Single-domain APOBECs, such as APOBEC3A, consist of one ZDD that confers both catalytic activity and binding. In contrast, double-domain members like APOBEC3G possess two ZDDs: an N-terminal domain that is typically non-catalytic but aids in recruitment, and a C-terminal domain that harbors the . These N- and C-terminal lobes in APOBEC3G are connected by a flexible linker, enabling independent folding while facilitating cooperative interactions. Accessory domains and motifs in APOBEC enzymes modulate their subcellular localization and substrate specificity. Nuclear localization signals (NLS), often bipartite sequences rich in basic residues, are present in isoforms such as APOBEC1 and (AID), directing them to the for targeted editing. RNA-binding motifs occur in select members like the N-terminal region of APOBEC3G and APOBEC1, enhancing interactions with substrates. Oligomerization is a key structural feature in several APOBEC3 proteins, promoting enzymatic efficiency through multimeric assemblies. APOBEC3G, for instance, forms dimers and higher-order oligomers via interfaces in its C-terminal domain, which stabilize the enzyme on substrates and are crucial for its antiviral potency. While single-domain APOBEC3A primarily functions as a , it can undergo cooperative dimerization under certain conditions to enhance activity.

Active Site and Catalysis

The of APOBEC enzymes centers on a ion (Zn²⁺) that is tetrahedrally coordinated by the side chains of one (His) and two cysteines (Cys), with a molecule completing the coordination. This coordination is essential for stabilizing the ion and positioning it to activate substrates during , as revealed in crystal structures of APOBEC3 catalytic domains. The residue plays a key role in proton abstraction, polarizing the substrate and facilitating the reaction by shuttling protons within the pocket. The core is HxEx₂₅₋₃₀PCx₂₋₄C. The of APOBEC-mediated follows a hydrolytic conserved across the family, converting to uracil through nucleophilic attack and elimination. A molecule bound to the is deprotonated to generate a , which adds to the C4 carbonyl of the base, forming a tetrahedral . The then abstracts a proton from the N3 position of , promoting the departure of the amino group as (NH₃) and restoring the of the uracil product. This zinc-stabilized process ensures efficient , with the metal lowering the pKₐ of the bound and stabilizing negative charge development in the . The overall deamination reaction can be represented as: \text{Cytosine-N3} + \text{H}_2\text{O} \rightarrow \text{Uracil-N3} + \text{NH}_3 where the zinc ion coordinates the to enhance reactivity. APOBEC enzymes exhibit a strong preference for single-stranded nucleic acids as substrates, as double-stranded structures sterically hinder access to the . Within these substrates, they display specificity, with many APOBEC3 family members favoring cytosines embedded in or TCW (where W is A or T) contexts to optimize binding and positioning in the catalytic pocket. This selectivity arises from interactions between flanking and residues near the , influencing the enzyme's efficiency without altering the core chemistry.

Mechanisms of Action

RNA Editing

APOBEC1, the founding member of the APOBEC family, mediates site-specific C-to-U RNA editing primarily in apolipoprotein B (APOB) mRNA, a process essential for generating protein isoforms involved in lipid metabolism. In mammals, APOB mRNA is transcribed as a full-length transcript encoding the 4536-amino-acid ApoB100 protein, which is secreted by the liver as a component of low-density lipoproteins (LDL). In the small intestine, APOBEC1 catalyzes deamination of cytidine 6666 (C6666) to uridine (U6666) within codon 2153, introducing a premature stop codon (CAA to UAA) that truncates the protein to the 2152-amino-acid ApoB48 isoform. This shorter form is crucial for assembling chylomicrons, which transport dietary lipids from the intestine to peripheral tissues, thereby regulating lipid homeostasis. The editing mechanism relies on a multiprotein complex where acts as the catalytic subunit, performing hydrolytic of the target . APOBEC1 lacks intrinsic -binding specificity and is recruited to the editing site through interaction with auxiliary factors, notably APOBEC1 complementation factor (ACF), a 70-kDa protein containing three RNA recognition motifs (RRMs). ACF binds an 11-nucleotide "mooring sequence" (5'-UUUUUAUUUAU-3') approximately 14 nucleotides downstream of the editing site in APOB mRNA, forming a stem-loop structure that positions APOBEC1 precisely for site-specific . This mooring sequence model ensures high-fidelity , with the efficiency regulated by nuclear factors and post-transcriptional modifications; for instance, ACF phosphorylation by enhances complex assembly in intestinal cells. The process occurs in the , after which the edited mRNA is exported to the for translation into ApoB48.52804-0/fulltext) Beyond APOB, APOBEC1 editing is limited to a small number of other RNA targets, primarily in the 3' untranslated regions (UTRs) of mRNAs, where it introduces C-to-U changes at low frequencies (typically <1% compared to ~90% for APOB). Transcriptome-wide analyses have identified around 30 such sites, often in AU-rich contexts with downstream mooring-like motifs, but these edits rarely alter protein coding and may influence mRNA stability or localization instead. Editing in non-coding RNAs is even more restricted, with sporadic reports of modifications in transcripts like NEAT1, though without clear functional consequences. This conservation of the editing machinery across mammals underscores its role in intestinal lipid transport, as evidenced by the presence of similar APOB editing sites and APOBEC1 orthologs in diverse species from rodents to marsupials, but its absence in non-mammalian vertebrates suggests an evolutionary adaptation tied to endothermic lipid homeostasis.

DNA Deamination

APOBEC enzymes catalyze the deamination of cytosine to uracil specifically in single-stranded DNA (ssDNA), a process that occurs preferentially during cellular events such as DNA replication and transcription, where ssDNA regions are transiently exposed. This substrate specificity ensures that APOBEC activity is confined to accessible ssDNA stretches, avoiding interference from the more stable double-stranded DNA (dsDNA) structure, which inhibits deamination due to its inability to accommodate the enzyme's binding requirements. The catalytic mechanism, involving zinc coordination and proton abstraction at the active site, facilitates this targeted editing, as detailed in structural studies. A hallmark of APOBEC-induced DNA deamination is the generation of hypermutation signatures, characterized by C→T transitions, particularly in the TC dinucleotide context, resulting in TC→TT mutations upon replication. Enzymes such as and exhibit a strong preference for deaminating cytosines flanked by thymine on the 5' side, contributing to clustered mutations in ssDNA substrates.00042-5) In APOBEC3 family members, this deamination is often processive, involving enzymatic scanning along ssDNA to access multiple target sites without dissociation, as demonstrated in biochemical assays with and , which can deaminate at least two cytosines per binding event. This processivity enhances the efficiency of hypermutation in extended ssDNA regions, such as those formed during lagging-strand synthesis. Following deamination, the resulting uracil in DNA is subject to cellular repair mechanisms, primarily through the base excision repair (BER) pathway initiated by uracil-DNA glycosylase (UNG). UNG excises the uracil, creating an abasic site that is processed by downstream BER enzymes, including AP endonuclease and DNA polymerase, to restore the original cytosine if repair is faithful. However, if the uracil persists unrepaired until the next round of replication, DNA polymerase incorporates adenine opposite the uracil, leading to a permanent C→T transition mutation upon completion of synthesis. This interplay between deamination and repair determines the mutagenic outcome, with UNG activity counteracting APOBEC-induced damage in various contexts.

Biological Roles

Innate Immunity and Antiviral Defense

APOBEC3 enzymes, particularly (A3G), play a crucial role in innate immunity by restricting viral replication through cytidine deamination of viral nucleic acids. In the case of human immunodeficiency virus type 1 (), A3G is packaged into virions during virus assembly in producer cells, where it colocalizes with viral RNA and Gag proteins. Upon infection of target cells, A3G deaminates deoxycytidines to deoxyuridines in the single-stranded retroviral cDNA during reverse transcription, leading to G-to-A hypermutations that introduce premature stop codons and disrupt essential viral genes such as gag and pol. This deamination-dependent mechanism severely impairs viral infectivity, as demonstrated in vif-deficient , where hypermutated proviruses predominate. However, counters this defense via its Vif protein, which binds A3G and recruits a Cullin5 E3 ubiquitin ligase complex to induce its polyubiquitination and proteasomal degradation, thereby preventing packaging into nascent virions. Beyond HIV-1, APOBEC3 proteins target other retroviruses, including human T-cell leukemia virus type 1 (HTLV-1), where A3G and other family members like A3F induce similar G-to-A mutations in proviral DNA, reducing viral propagation despite partial resistance conferred by HTLV-1's nucleocapsid protein. APOBEC3 enzymes also restrict endogenous retroelements, such as (L1) retrotransposons, by deaminating their cDNA intermediates during retrotransposition, which inhibits mobilization and genomic integration; for instance, A3A and A3C effectively suppress L1 activity in a deaminase-dependent manner. APOBEC3A has also been shown to deaminate cytidines in RNA, contributing to antiviral defense against coronaviruses. Species-specific variations further modulate these restrictions: human APOBEC3B potently inhibits vif-deficient HIV-1, whereas its rhesus macaque ortholog shows minimal activity, highlighting evolutionary adaptations that influence cross-species transmission barriers. Expression of APOBEC3 genes is tightly regulated to enhance antiviral responses, with interferon-alpha (IFN-α) and IFN-β potently upregulating A3G, A3F, and A3H transcripts in various cell types, including hepatocytes and peripheral blood mononuclear cells, via JAK-STAT signaling pathways. This induction amplifies the innate immune barrier against retroviral infections, as seen in IFN-treated cells where elevated APOBEC3 levels correlate with reduced HIV-1 replication. Genetic polymorphisms in APOBEC3H, particularly its seven haplotypes (I-VII), significantly affect susceptibility; stable haplotypes II, V, and VII produce antiviral proteins resistant to Vif degradation, conferring better HIV-1 control in vivo, while unstable variants (e.g., haplotype I) degrade rapidly and offer limited protection. These polymorphisms vary across populations and influence disease progression rates in high-risk cohorts.

Adaptive Immunity

Activation-induced cytidine deaminase (AID), a member of the family, plays a central role in adaptive immunity by facilitating antibody diversification in B cells through two key processes: (SHM) and (CSR). SHM introduces point mutations into the variable regions of immunoglobulin genes, enabling the affinity maturation of antibodies to enhance antigen recognition, while CSR allows B cells to switch from producing IgM to other isotypes like IgG, IgA, or IgE, thereby diversifying effector functions without altering antigen specificity. These processes occur primarily in germinal centers of secondary lymphoid organs during T cell-dependent immune responses, where AID expression is tightly regulated to balance diversity generation with genomic stability. The mechanism of AID action involves cytidine deamination within single-stranded DNA, preferentially targeting WRC hotspots (where W is A or T, and R is A or G) in transcriptionally active immunoglobulin loci. In SHM, AID deaminates cytidines in the V(D)J segments of rearranged immunoglobulin genes, leading to U:G mismatches that are processed by error-prone repair pathways such as base excision repair (via UNG and APE1) or mismatch repair (via MSH2-MSH6), resulting in mutations that drive antibody evolution. For CSR, AID targets repetitive switch (S) regions upstream of constant region exons and G-quadruplex structures, generating staggered double-strand breaks through closely spaced deaminations on both strands; these breaks are resolved by non-homologous end joining to recombine and delete intervening DNA, enabling isotype switching. This targeted deamination is facilitated by transcription-induced DNA structures like R-loops, which expose single-stranded DNA substrates for AID. Beyond its physiological roles, AID can cause off-target deamination at non-immunoglobulin loci, contributing to genomic instability in B-cell lymphomas. Aberrant AID activity leads to clustered mutations (kataegis) and chromosomal translocations in genes like PIM1, MYC, and BCL6, which are recurrent in germinal center-derived lymphomas such as diffuse large B-cell lymphoma, promoting oncogenesis when combined with defective DNA repair. AID expression is stringently controlled at the transcriptional level, primarily in germinal center B cells, through signaling pathways involving CD40 ligation, cytokines (e.g., IL-4, TGF-β), and transcription factors like Pax5, Bcl6, and STAT6. This regulation ensures AID activity is confined to specific immune contexts, preventing widespread mutagenesis. Dysregulation of AID, such as ectopic or prolonged expression, has been linked to autoimmunity; for instance, in systemic lupus erythematosus (SLE), elevated AID levels in B cells correlate with hypermutation of autoantigen-specific antibodies and increased CSR to pathogenic isotypes, exacerbating disease in both mouse models and human patients.

Pathological Implications

Role in Cancer

APOBEC enzymes, particularly APOBEC3A and APOBEC3B, contribute to cancer development by catalyzing cytosine deamination in DNA, leading to characteristic mutational signatures dominated by C>T transitions at TC dinucleotide motifs. These signatures are prevalent across multiple tumor types, accounting for a significant proportion of mutations—often 10-50% in and cancers—in (TCGA) datasets for cancers such as , lung, and carcinomas. In these malignancies, APOBEC-driven mutations often manifest as kataegis, focal hypermutation events producing clustered C>T transitions, which fuel tumor heterogeneity and evolutionary adaptation. Overexpression of APOBEC3A and APOBEC3B in tumor cells promotes this , frequently triggered by activation of the signaling pathway in response to inflammatory cues or oncogenic stress. This elevated expression evades efficient mechanisms, such as those involving uracil-DNA glycosylase, allowing products to persist and convert to mutations during replication. Consequently, APOBEC activity generates subclonal diversity that can confer resistance to therapies, including targeted inhibitors in and cancers. APOBEC enzymes exhibit a dual role in oncogenesis: initially suppressing tumor growth through immunogenic mutations that enhance antitumor immunity, but later driving progression by increasing genomic instability and . In early stages, APOBEC-induced neoantigens can bolster T-cell responses, potentially limiting tumor expansion, whereas chronic activity accelerates and evasion in advanced disease. Recent studies up to 2025 highlight APOBEC3B germline polymorphisms, such as the 29.5 kb deletion, as risk factors for breast cancer by elevating somatic mutation rates and tumor burden. In thyroid cancer, high APOBEC enrichment correlates with immune evasion, malignant progression, and poor prognosis, underscoring subtype-specific impacts. Additionally, APOBEC hypermutation signatures predict improved responses to immunotherapy in breast and lung cancers, linking mutational load to enhanced checkpoint inhibitor efficacy. Recent 2025 studies indicate that APOBEC3 mutagenesis contributes to resistance against targeted therapies like CDK4/6 inhibitors in breast cancer.

Involvement in Other Diseases

APOBEC3 proteins play a critical role in restricting HIV-1 replication, and deficiencies or genetic variants in these enzymes are associated with accelerated disease progression. For instance, the deletion of APOBEC3B has been associated with increased susceptibility to HIV-1 acquisition in some studies, potentially by impairing the hypermutation of viral genomes, leading to higher viral loads in affected individuals. Similarly, certain haplotypes of APOBEC3G and APOBEC3F, such as those conferring resistance to Vif-mediated degradation, correlate with slower progression to AIDS in long-term non-progressors, while deleterious variants enhance viral infectivity and hasten clinical decline. The frequency of APOBEC3-mediated G-to-A hypermutations in proviral DNA further contributes to reduced viral fitness, underscoring how APOBEC3 deficiencies promote persistent infection and immune evasion. In hepatitis B virus (HBV) infection, APOBEC3 enzymes, particularly APOBEC3G and APOBEC3B, induce extensive mutagenesis of viral DNA during reverse transcription, which can influence viral integration into the host genome. In patients with HBV-associated cirrhosis, up to 35% of integrated HBV DNA sequences exhibit APOBEC3-mediated G-to-A mutations, often driven by upregulated expression of multiple APOBEC3 family members in response to chronic inflammation and interferon-α signaling. This editing disrupts viral replication and promotes immune escape variants, but hyper-edited genomes in co-infections with hepatitis C virus show even higher mutation loads (up to 47%), potentially exacerbating liver damage and oncogenic integration events. Activation-induced cytidine deaminase (AID), a member of the APOBEC family, drives and class-switch recombination in B cells, and its dysregulation contributes to production in systemic lupus erythematosus (SLE) and (RA). In SLE-prone mouse models, elevated AID expression, often upregulated by HoxC4 transcription factors, increases hypermutation rates and interchromosomal translocations (e.g., c-Myc/IgH), resulting in pathogenic IgG autoantibodies like anti-dsDNA and severe . Human SLE patients similarly display heightened AID activity in peripheral B cells, leading to diversified autoreactive clones and exacerbated severity. In RA, AID-mediated hypermutation in tertiary lymphoid structures generates high-affinity autoantibodies, promoting synovial inflammation and joint destruction, with AID inhibition shown to reduce autoantibody levels and pathology in experimental models. APOBEC1, through its RNA editing of apolipoprotein B (apoB) mRNA, regulates homeostasis, and its dysregulation links to metabolic disorders such as . In mouse models lacking both low-density lipoprotein receptor (LDLR) and APOBEC1, exclusive production of unedited apoB100 leads to severe LDL accumulation, , and accelerated atherosclerotic plaque formation compared to LDLR-deficient controls. This occurs because APOBEC1 normally generates apoB48 for assembly in the intestine, preventing excessive circulating LDL; its absence shifts transport toward atherogenic pathways, highlighting APOBEC1's protective role against . APOBEC2, predominantly expressed in cardiac and , influences myocardial remodeling, with emerging evidence suggesting involvement in pathological cardiac . Knockout studies reveal that APOBEC2 deficiency alters muscle fiber types and assembly, potentially predisposing to hypertrophic responses under stress, as seen in altered expression profiles during cardiogenic . Recent analyses of regulators in hypertrophic hearts indicate APOBEC2 downregulation correlates with ventricular dysfunction, implying a role in maintaining cardiac stability. Dysregulation of APOBEC-mediated RNA editing has been implicated in neurodegeneration, particularly through APOBEC1 activity in . Loss of APOBEC1 in murine disrupts editing of transcripts like Lamp2, leading to lysosomal dysfunction, , and progressive demyelination in the by middle age, accompanied by behavioral deficits resembling anxiety and cognitive decline. In 2024-2025 studies on models, reduced APOBEC/ RNA editing events contribute to neuronal protein misfolding and synaptic loss, with epitranscriptomic alterations enriching in pathways for and neurodegeneration during progression.

Therapeutic Targeting

Inhibitors and Modulators

Inhibitors of APOBEC enzymes primarily target the zinc-dependent catalytic site or allosteric pockets to block activity, with classes including chelators and binders. chelators, such as N,N,N′,N′-tetrakis(2-pyridylmethyl)ethane-1,2-diamine (TPEN), disrupt the coordination essential for APOBEC function, particularly by inhibiting HIV-1 Vif-mediated of APOBEC3G and restoring its antiviral activity in cell-based assays. Additional chelators like SN-1 and SN-2 have been tested for their ability to prevent Vif-APOBEC3G binding by leaching from Vif, highlighting a strategy to enhance endogenous APOBEC3 restriction of . Active site binders represent a key class of small-molecule inhibitors designed to occupy the catalytic pocket shared across APOBEC family members. Seminal structure-based efforts identified first-generation compounds that inhibit purified activation-induced cytidine deaminase (AID), APOBEC3A, and APOBEC3B with low micromolar IC50 values, demonstrating activity against endogenous AID in lymphoma cell extracts. More recent structure-guided designs from 2023 focus on APOBEC3A, using U-shaped DNA hairpin oligonucleotides containing 2'-deoxyzebularine (dZ) or 5-fluoro-dZ analogs, such as TTFdZ-hairpin, which bind the active site with micromolar Ki (31 μM) and reduce APOBEC3A-mediated editing by over twofold in 293T cell models without significant cytotoxicity. These inhibitors exploit the enzyme's preference for single-stranded DNA substrates, mimicking the transition state to block mutagenesis. From 2023 to 2025, advances in structure-guided inhibitor development have emphasized selectivity, particularly for APOBEC3B, a major driver of cancer mutations. of diverse chemical libraries identified allosteric small molecules targeting a novel pocket in the APOBEC3B C-terminal domain, yielding hits with values in the 100 μM to 1 mM range that selectively inhibit APOBEC3B over the homologous APOBEC3A due to steric differences at residue H56. A 2025 review notes emerging selective APOBEC3B inhibitors in early stages. For instance, the natural product 3,5-diiodotyrosine, identified in 2022, suppresses APOBEC3B activity and correlates with improved prognosis in pan-cancer analyses. Modulators of APOBEC enzymes include biologic agents like and natural protein antagonists. Next-generation APOBEC3 inhibitors utilize phosphorothioate-modified with dZ motifs, such as H4A for APOBEC3A ( 9.2 nM) and linear I12G for APOBEC3G ( 670 nM), offering enhanced stability (>60% intact after three days) and improved potency over prior designs through 2'-fluoro and modifications. These function as competitive modulators by binding the catalytic site, akin to aptamers in their sequence-specific targeting. Vif-like proteins from lentiviruses, such as HIV-1 Vif, serve as natural modulators by promoting ubiquitination and degradation of APOBEC3G, counteracting its antiviral effects. Genetic polymorphisms in APOBEC3 genes, including copy number variations, influence baseline enzymatic activity and may affect inhibitor efficacy by altering substrate binding or expression levels. Preclinical studies demonstrate that APOBEC inhibitors reduce mutagenesis in cellular models, supporting their potential in cancer and viral therapies. For instance, phosphorothioated hairpin inhibitors of APOBEC3A decrease DNA editing signatures in human cell lines, slowing tumor evolution by limiting cytosine-to-thymine mutations. In HIV contexts, 2025 developments include optimized small-molecule inhibitors like NU-611, which stabilize APOBEC3G levels in 293T cells, increasing its incorporation into virions and reducing Vif+ HIV-1 infectivity by over 60% in TZM-bl reporter assays through blockade of pVHL-mediated degradation. Allosteric APOBEC3B inhibitors from virtual screening similarly curb deamination in biochemical assays, with NMR confirming non-competitive binding that preserves DNA substrate access while inhibiting catalysis.

Clinical Applications

APOBEC mutational signatures, particularly SBS2 and SBS13, serve as biomarkers in liquid biopsies for assessing cancer prognosis, with validation through circulating tumor DNA (ctDNA) analysis in advanced breast cancer to predict immunotherapy responsiveness. In breast cancer, these signatures are enriched in 88% of tumor mutation burden-high metastatic invasive lobular carcinomas, correlating with elevated neoantigen load and improved outcomes in immune checkpoint inhibitor (ICI) therapy. For HIV susceptibility, expression profiling of APOBEC3G mRNA levels in peripheral blood mononuclear cells has been associated with viral load set point and early disease pathogenesis, indicating lower expression correlates with increased infection risk. Similarly, APOBEC3A expression in monocytes links myeloid differentiation states to HIV-1 resistance, supporting its use in profiling innate immune susceptibility. In cancer therapy, APOBEC inhibitors are under investigation to mitigate mutagenesis-driven resistance, particularly in where APOBEC3A and APOBEC3B activity accelerates evolution in hormone receptor-positive and triple-negative subtypes. Research from 2025 highlights APOBEC3 signatures as predictors of shorter in metastatic settings, prompting efforts to develop targeted inhibitors that could extend therapeutic windows by reducing mutation rates. As of November 2025, no phase I/II trials for direct APOBEC inhibitors have commenced, though preclinical studies suggest potential for upcoming biomarker-guided trials to test reduction strategies alongside existing endocrine or targeted therapies. For antiviral applications, enhancers of APOBEC3G activity, such as pathway modulators, show promise in boosting restriction of replication, with preclinical data suggesting reduced viral load through increased A3G expression. Key challenges in APOBEC modulation include off-target effects from inhibitors, which may disrupt normal DNA editing in non-cancerous cells, though next-generation oligonucleotide-based APOBEC3 inhibitors demonstrate improved specificity and stability in 2025 preclinical models. Combination approaches with immunotherapy are advancing, as APOBEC signatures predict ICI efficacy across cancers like non-small cell lung and breast, with high mutation counts (average 38 vs. 10 in responders vs. non-responders) outperforming total burden metrics. In clear cell renal cell carcinoma, APOBEC-high subtypes benefit from ICI paired with anti-angiogenesis agents, enhancing immune infiltration. As of November 2025, FDA approvals for A3B-targeted drugs remain pending, with ongoing research focusing on biomarker-guided trials to address resistance and toxicity.

References

  1. [1]
    The APOBEC Protein Family: United by Structure, Divergent in ... - NIH
    The APOBEC (Apolipoprotein B mRNA Editing Catalytic Polypeptide-like) family of proteins has diverse and important functions in human health and disease.Dna Viruses · Rna Editing Of Cellular... · Apobec And Rna Interactions
  2. [2]
    Functions and consequences of AID/APOBEC-mediated DNA and ...
    Mar 7, 2022 · The AID/APOBEC polynucleotide cytidine deaminases have historically been classified as either DNA mutators or RNA editors based on their first identified ...
  3. [3]
  4. [4]
  5. [5]
  6. [6]
  7. [7]
  8. [8]
  9. [9]
  10. [10]
    An ancient history of gene duplications, fusions and losses in the ...
    May 28, 2012 · The APOBEC3 (A3) genes play a key role in innate antiviral defense in mammals by introducing directed mutations in the DNA. The human genome ...
  11. [11]
    Population Stratification of a Common APOBEC Gene Deletion ... - NIH
    Apr 20, 2007 · The deletion is rare in Africans and Europeans (frequency of 0.9% and 6%), more common in East Asians and Amerindians (36.9% and 57.7%), and ...
  12. [12]
    Mice Deficient in APOBEC2 and APOBEC3 - PMC - PubMed Central
    APOBEC2 is muscle-specific, and APOBEC3 is a DNA deaminase. Both are inessential for mouse development, survival, or fertility. APOBEC3 is abundant in lymphoid ...Missing: non- | Show results with:non-
  13. [13]
    Dimerization regulates both deaminase-dependent and ... - Nature
    Sep 19, 2017 · APOBEC3G (A3G) is particularly effective at suppressing the infectivity of human immunodeficiency virus type 1 (HIV-1). As a result, HIV-1 must ...
  14. [14]
    Association of a germline copy number polymorphism of ... - Nature
    Apr 13, 2014 · The results suggest that the APOBEC3A-APOBEC3B germline deletion allele confers cancer susceptibility through increased activity of APOBEC- ...
  15. [15]
    Analysis of Human APOBEC3H Haplotypes and Anti-Human ... - NIH
    All of them have antiretroviral activity, although the level of this activity can vary among different retroviral targets (26, 36, 38). In particular, A3B, A3DE ...
  16. [16]
    Functions and Regulation of the APOBEC Family of Proteins - NIH
    Abstract. APOBEC1 is a cytidine deaminase that edits messenger RNAs and was the first enzyme in the APOBEC family to be functionally characterized.1. Introduction · Figure 2. Apobec Family... · 2.3. Apobec3
  17. [17]
    APOBEC-1 and AID are Nucleo-cytoplasmic Trafficking Proteins but ...
    Along with an N-terminal NLS, both APOBEC-1 and AID have C-terminal leucine-rich, nuclear export signal (NES) domains (Fig. 1). In fact, for APOBEC-1 the NES is ...
  18. [18]
    Crystal structures of APOBEC3G N-domain alone and its complex ...
    Aug 2, 2016 · The dimerization interface of rA3G-CD1 is important for oligomerization, nucleic acid binding and Vif-mediated degradation.Results · Soluble A3g-Cd1 Dimers After... · Dimer Formation Is Important...
  19. [19]
    Crystal Structure of the DNA Deaminase APOBEC3B Catalytic Domain
    The mode of zinc coordination resembles those of other A3 members and zinc-dependent nucleoside/mononucleotide deaminase enzymes. The single zinc ion in ...In Vitro Dna Deamination... · A3b Catalytic Domain Crystal... · Nucleotide Binding Site And...
  20. [20]
    Reaction Mechanism of Zinc-Dependent Cytosine Deaminase from ...
    This zinc-dependent enzyme catalyzes the deamination of cytosine to form uracil and ammonia. The calculations give a detailed description of the catalytic ...
  21. [21]
    APOBEC3s: DNA‐editing human cytidine deaminases - Silvas - 2019
    Jun 26, 2019 · Deamination reaction mechanism. (a) Schematic of deoxycytidine conversion to deoxyuridine by ssDNA cytidine deaminases. Leaving NH2 group of ...
  22. [22]
    Review APOBEC Enzymes as Targets for Virus and Cancer Therapy
    Jan 18, 2018 · The A3s contain a consensus Zn-binding motif of histidine(His)-X-glutamic acid(Glu)-X23-28-proline(Pro)-cysteine(Cys)-X2-4-cysteine(Cys), where ...
  23. [23]
    Structural determinants of human APOBEC3A enzymatic and ...
    The cysteine and histidine residues coordinate a zinc metal ion (Zn2+), and ... APOBEC3G catalytic domain reveals potential oligomerization interfaces. ,.
  24. [24]
    Molecular cloning of an apolipoprotein B messenger RNA editing ...
    A full-length complementary DNA clone encoding an apo B messenger RNA editing protein (REPR) was isolated from rat small intestine.
  25. [25]
    An auxiliary factor containing a 240-kDa protein complex is involved in apolipoprotein B RNA editing. | PNAS
    ### Summary of ACF Identification and Role in APOBEC1 RNA Editing Mechanism
  26. [26]
    The apolipoprotein B mRNA editing complex performs a ... - NIH
    Introduction. Cytidine (C) to uridine (U) RNA editing was first discovered in apolipoprotein B (apoB) mRNA where deamination of a C to U in mammalian apoB100 ...
  27. [27]
    Transcriptome-wide sequencing reveals numerous APOBEC1 ... - NIH
    Editing results in translation of a truncated apoB isoform with distinct functions in lipid transport. To address the possibility that APOBEC1 edits additional ...
  28. [28]
    Characterisation of APOBEC3B-Mediated RNA editing in breast ...
    Sep 25, 2024 · Our findings indicate that A3B engages in selective RNA editing including targeting NEAT1 and MALAT1 long non-coding RNAs that are often highly expressed in ...
  29. [29]
    The evolution of apolipoprotein B and its mRNA editing complex ...
    Jan 30, 2018 · This study focused on the molecular evolution of Apobec1 and Apob in order to ascertain if apob mRNA editing occurs in eels, a basal teleost.Missing: primary | Show results with:primary
  30. [30]
    APOBEC ENZYMES AS TARGETS FOR VIRUS AND CANCER ...
    APOBEC3 enzymes are tantalizing targets for developing chemical probes and therapeutic molecules to harness mutational processes in human disease.Apobec Enzymes As Targets... · Apobec Structure And... · Apobec Inhibition: Therapy...
  31. [31]
    Structural basis for targeted DNA cytosine deamination and ...
    A3A is a globular enzyme with a single zinc-coordinating active site, and it has proven to be the most potent human DNA cytosine deaminase. To elucidate the ...Results · A3a-Ssdna Structure Reveals... · A3b-Ssdna Structure And The...Missing: composition | Show results with:composition
  32. [32]
    APOBEC-Induced Mutagenesis in Cancer - Annual Reviews
    Aug 26, 2022 · However, tumors with APOBEC-induced mutations also have higher instances of oncogenic PIK3CA mutations, which are more likely to occur at TCW.
  33. [33]
    Different Mutagenic Potential of HIV-1 Restriction Factors ...
    Using synthetic DNA substrates, we characterized APOBEC3F and found that similar to APOBEC3G; it is a processive enzyme and can deaminate at least two cytosines ...
  34. [34]
    Biochemical Analysis of Hypermutation by the Deoxycytidine ...
    We find that APOBEC3A, unlike the highly processive APOBEC3G, exhibits low or no processivity when deaminating synthetic ssDNA substrates with two cytosines ...
  35. [35]
    Repair of APOBEC3G-Mutated Retroviral DNA In Vivo Is ... - NIH
    These data show that UNG plays a critical role in the repair of the damage inflicted by APOBEC3 deamination of reverse-transcribed DNA.
  36. [36]
    Uracil DNA glycosylase counteracts APOBEC3G-induced ... - PubMed
    We investigated whether uracil residues in hepadnavirus DNA were excised by uracil-DNA glycosylase (UNG), a host factor for base excision repair (BER).
  37. [37]
    The deaminase APOBEC3B triggers the death of cells lacking uracil ...
    Oct 14, 2019 · APOBEC3B (A3B) is overexpressed in multiple tumor types and a major source of mutation by converting DNA cytosines to uracils.
  38. [38]
    Perturbation of base excision repair sensitizes breast cancer cells to ...
    Jan 6, 2020 · Abundant APOBEC3 (A3) deaminase-mediated mutations can dominate the mutational landscape ('mutator phenotype') of some cancers, however, ...
  39. [39]
    Isolation of a human gene that inhibits HIV-1 infection and ... - Nature
    Jul 14, 2002 · Sheehy, A., Gaddis, N., Choi, J. et al. Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein.Missing: APOBEC3G | Show results with:APOBEC3G
  40. [40]
    Resistance of human T cell leukemia virus type 1 to APOBEC3G ...
    Human T cell leukemia virus type 1 (HTLV-1) has evolved a remarkable strategy to thwart the antiviral effects of the cellular cytidine deaminase APOBEC3G ...
  41. [41]
    Human and Rhesus APOBEC3D, APOBEC3F, APOBEC3G ... - PMC
    Unlike its human homolog, rhesus A3B was unable to significantly restrict Vif-deficient HIV.
  42. [42]
    Interferon-inducible expression of APOBEC3 editing enzymes in ...
    We show that normal human liver expresses the mRNAs of APOBEC3B (A3B), APOBEC3C (A3C), A3F, and A3G. In primary human hepatocytes, ...
  43. [43]
    Sustained high expression of multiple APOBEC3 cytidine ... - Nature
    Apr 12, 2021 · Interferon-inducible expression of APOBEC3 editing enzymes in human hepatocytes and inhibition of hepatitis B virus replication. Hepatology ...
  44. [44]
    APOBEC3H polymorphisms and susceptibility to HIV-1 infection in ...
    Nov 12, 2015 · Human APOBEC3H (A3H) is a member of APOBEC cytidine deaminase family intensively constraining the HIV-1 replication.
  45. [45]
    Class Switch Recombination and Hypermutation Require Activation ...
    These results suggest that AID may be involved in regulation or catalysis of the DNA modification step of both class switching and somatic hypermutation.
  46. [46]
    AID function in somatic hypermutation and class switch recombination
    Activation-induced cytidine deaminase (AID) initiates somatic hypermutation of immunoglobulin (Ig) gene variable regions and class switch recombination (CSR)Missing: seminal papers
  47. [47]
    Regulation of AID, the B-cell genome mutator - Genes & Development
    Both processes require the activity of the B-cell-expressed mutator enzyme activation-induced cytidine deaminase (AID), which is expressed by DZ B cells and ...
  48. [48]
    Interplay between Target Sequences and Repair Pathways ...
    We find that the switch (S) region is a much more efficient AID deamination target than the V region. Igh locus AID-initiated lesions are processed by error- ...
  49. [49]
    A Common Mechanism that Underpins Antibody Diversification
    Nov 19, 2015 · Targeting of AID to antibody variable (V) regions results in somatic hypermutation, whereas its recruitment to switch (S) regions leads to class-switch ...
  50. [50]
    AID expression in B-cell lymphomas causes accumulation ... - PubMed
    We found that clustered mutations (kataegis) in lymphoma and chronic lymphocytic leukemia predominantly carry AID-hotspot mutational signatures.
  51. [51]
    AID-associated DNA repair pathways regulate malignant ...
    Although off-target AID activity also contributes to oncogenic point mutations and chromosome translocations associated with GC and post-GC B-cell lymphomas, ...
  52. [52]
    The complex regulation and function of activation-induced cytidine ...
    Activation-induced cytidine deaminase (AID) instigates mutations and DNA breaks in Ig genes undergoing somatic hypermutation (SHM) and class switch ...
  53. [53]
    Potential roles of Activation-Induced cytidine Deaminase in ...
    Because AID is responsible for both CSR and SHM, AID may therefore promote the development of autoimmune diseases by allowing the production of mutated ...
  54. [54]
    AID dysregulation in lupus-prone MRL/Faslpr/lpr mice increases ...
    Thus, AID dysregulation in autoimmune MRL/Faslpr/lpr mice results in high levels of interchromosomal translocations involving the IgH locus. HoxC4 deficiency ...
  55. [55]
    Mechanisms of APOBEC3 mutagenesis in human cancer cells | Nature
    Jul 20, 2022 · The APOBEC3 family of cytosine deaminases has been implicated in some of the most prevalent mutational signatures in cancer.
  56. [56]
    APOBEC3-mediated mutagenesis in cancer: causes, clinical ...
    Mar 28, 2023 · Apolipoprotein B mRNA-editing enzyme, catalytic polypeptides (APOBECs) are cytosine deaminases involved in innate and adaptive immunity.<|control11|><|separator|>
  57. [57]
    Mapping clustered mutations in cancer reveals APOBEC3 ... - Nature
    Feb 9, 2022 · Our results reveal the diversity of clustered mutational processes in human cancer and the role of APOBEC3 in recurrently mutating and fuelling the evolution ...
  58. [58]
    The PKC-NFκB Signaling Pathway Induces APOBEC3B Expression ...
    Sep 29, 2015 · Overexpression of the antiviral DNA cytosine deaminase APOBEC3B has been linked to somatic mutagenesis in many cancers.
  59. [59]
    APOBEC3 mutagenesis drives therapy resistance in breast cancer
    May 16, 2025 · These studies reveal APOBEC3 mutagenesis to be a frequent mediator of therapy resistance in breast cancer and highlight its potential as a biomarker and target ...<|separator|>
  60. [60]
    APOBEC in breast cancer: a dual player in tumor evolution ... - NIH
    Apr 28, 2025 · APOBEC-driven mutagenesis correlates with elevated tumor mutational burden (TMB), replication stress vulnerability, and immune checkpoint inhibitor (ICI) ...
  61. [61]
    APOBEC in breast cancer: a dual player in tumor evolution and ...
    Apr 27, 2025 · In breast cancer, APOBEC contributes to immunogenic mutations, potentially enhancing response to immune checkpoint inhibitors in tumors with ...Missing: recent | Show results with:recent
  62. [62]
    Pembrolizumab monotherapy for previously treated metastatic ...
    A common germline deletion polymorphism in the APOBEC3B gene increases the rate of somatic hypermutation in breast cancer, which in turn is associated with ...Pembrolizumab Monotherapy... · Study Design And Patient... · Evaluating Germline Apobec3b...
  63. [63]
    Comprehensive Insights into APOBEC Mutations in Thyroid Cancer
    Jul 26, 2025 · High APOBEC mutation enrichment scores were strongly associated with poor prognosis, immune evasion, and increased risk of malignant progression ...
  64. [64]
    APOBEC mutational signature predicts prognosis and ...
    Mar 31, 2023 · APOBEC3 score provides a novel means of predicting prognosis and improving the immunotherapy response for future applications.
  65. [65]
    Zinc chelation inhibits HIV Vif activity and liberates antiviral function ...
    Zinc chelation inhibits HIV Vif activity and liberates antiviral function of the cytidine deaminase APOBEC3G · Authors · Affiliation.
  66. [66]
    Structure-Based Design of First-Generation Small Molecule ...
    Jul 19, 2021 · We identified small molecules that inhibit purified AID, AID in cell extracts, and endogenous AID of lymphoma cells.Missing: binders | Show results with:binders
  67. [67]
    Structure-guided inhibition of the cancer DNA-mutating enzyme ...
    Oct 11, 2023 · APOBEC3A inhibitors may therefore comprise a unique class of anti-cancer agents that work by blocking mutagenesis, slowing tumor evolvability, and preventing ...Missing: chelators binders
  68. [68]
    Development of Allosteric Small Molecule APOBEC3B Inhibitors ...
    Apr 25, 2024 · Our studies represent the first virtual screening effort against A3Bctd that has yielded candidate inhibitors suitable for further development.Missing: chelators | Show results with:chelators
  69. [69]
    Development of APOBEC3B inhibitors: Recent advances and ...
    Sep 27, 2025 · Presently, although research on A3B inhibitors is still in its early stages, various selective inhibitors have already been reported. ... A3B ...
  70. [70]
    Identification of natural product 3, 5-diiodotyrosine as APOBEC3B ...
    Nov 2, 2022 · Conclusions This is the first prove-of-concept study to elucidate that the natural product 3, 5-diiodotyrosine could prevent somatic mutation ...
  71. [71]
    Next generation APOBEC3 inhibitors: optimally designed for ...
    Our optimally designed A3 oligonucleotide inhibitors show improved potency and stability compared to previous inhibitors targeting these critical enzymes.Missing: chelators binders
  72. [72]
    Degradation-Independent Inhibition of APOBEC3G by the HIV-1 Vif ...
    In this review, we describe our current knowledge regarding the degradation-independent inhibition of A3s, and A3G in particular, by the HIV-1 Vif protein.Missing: aptamers | Show results with:aptamers
  73. [73]
    Inhibiting APOBEC3 Activity with Single-Stranded DNA Containing 2
    Nov 12, 2018 · APOBEC3 enzymes form part of the innate immune system by deaminating cytosine to uracil in single-stranded DNA (ssDNA) and thereby ...
  74. [74]
    Design and Characterization of Inhibitors of Cell-Mediated ... - MDPI
    Design and Characterization of Inhibitors of Cell-Mediated Degradation of APOBEC3G That Decrease HIV-1 Infectivity. Viruses 2025, 17, 514. https://doi.org ...
  75. [75]
    APOBEC3G Expression is Dysregulated in Primary HIV-1 Infection ...
    We investigated the association of APOBEC3G mRNA levels and APOBEC3G genetic variants on HIV-1 susceptibility, and early disease pathogenesis using viral load ...
  76. [76]
    Myeloid differentiation and susceptibility to HIV-1 are linked to ... - NIH
    We uncovered APOBEC3A, not previously associated with anti-HIV activity, as being critical in monocyte resistance.
  77. [77]
    BCRF-Backed Study Identifies Process Linked to Drug Resistance in ...
    Jul 9, 2025 · Abnormal elevation of APOBEC3 activity has been observed in bladder, head and neck, and breast cancers. Prior studies have associated higher ...
  78. [78]
    APOBEC3G Is a p53-Dependent Restriction Factor in Respiratory ...
    Nov 27, 2023 · Clinically, the opportunity to boost p53 activity and levels of A3G might decrease the viral load. These approaches may also help in developing ...
  79. [79]
    APOBEC3B and APOBEC mutational signature as potential ...
    Apr 26, 2018 · APOBEC mutation count can be a novel predictive biomarker for immunotherapy efficacy. APOBEC activation in cancer leads to elevated levels of ...
  80. [80]
    APOBEC family reshapes the immune microenvironment and ...
    Sep 9, 2024 · The APOBEC family plays a critical role in regulating the progression and metastasis of cancer by inducing mutations. In addition, these ...