Fact-checked by Grok 2 weeks ago

Nuclear export signal

A nuclear export signal (NES) is a short, typically leucine-rich sequence, usually 8–15 residues long, that serves as a targeting to direct the export of proteins and certain ribonucleoprotein complexes from the to the via nuclear pore complexes. These signals are recognized by specific nuclear export receptors, known as exportins, which facilitate the selective and energy-dependent transport across the , ensuring proper spatiotemporal regulation of cellular processes. The consensus sequence of classical NESs, often classified as CRM1-dependent or leucine-rich NESs (LR-NESs), follows patterns such as Φ-X₂₋₃-Φ-X₂₋₃-Φ-X-Φ (where Φ represents hydrophobic residues like , , , , or , and X is any ), though structural variations exist that allow flexibility in binding. Exportins, particularly CRM1 (also called exportin 1), bind NES-containing cargoes in the in with Ran-GTP, a GTP-bound form of the Ran that is enriched in the due to its asymmetric distribution across the ; this interaction promotes translocation through the , followed by cargo release in the upon GTP hydrolysis to Ran-GDP. Non-classical NESs, which may lack the strict leucine-rich motif, are recognized by other exportins like exportin-t for tRNAs or exportin-5 for microRNAs. Classical NESs, often via adaptor proteins bearing the signal, also facilitate export of ribonucleoprotein complexes such as ribosomal subunits. NES-mediated export plays a critical role in cellular by controlling the localization of transcription factors, signaling proteins, and viral components, thereby influencing , , and . Dysregulation of NES function, such as through mutations or overexpression of CRM1, has been implicated in diseases including cancers, where altered nuclear-cytoplasmic shuttling of tumor suppressors like disrupts normal cellular responses. Databases like NESdb catalog over 400 experimentally validated NESs from CRM1 cargoes (as of ), aiding in the prediction and analysis of these signals through sequence and structural studies.

Introduction

Definition and Overview

A nuclear export signal (NES) is a short sequence, typically 8–15 in length, embedded within cargo proteins that mediates their from the to the via complexes (NPCs). These signals enable the selective shuttling of proteins and associated cargoes across the , ensuring balanced nucleocytoplasmic distribution that is vital for cellular and function. NESs function in opposition to nuclear localization signals (NLSs), which direct protein import into the ; while both signals exploit the bidirectional permeability of NPCs, NESs drive outward trafficking to regulate , , and stress responses. The concept of NES was first established in 1995 through studies identifying functional export motifs in the HIV-1 Rev protein and the cAMP-dependent (PKIα). At their core, NESs feature a pattern of spaced hydrophobic residues, often leucines, that facilitate recognition and binding by export receptors such as CRM1 (also known as exportin 1).

Biological Importance

(NES) play a pivotal role in gene regulation by facilitating the export of transcription factors from the , thereby deactivating their transcriptional activity and preventing prolonged in response to stimuli. This shuttling mechanism allows for precise control over transcription, as NES-mediated export ensures that factors return to the after their nuclear function, maintaining balanced levels. Similarly, NES enable the export of mRNA-binding proteins, which influences post-transcriptional processes such as mRNA stability and translation efficiency in the . In signaling pathways, are essential for the rapid nucleocytoplasmic shuttling of proteins like kinases and repressors, enabling cells to respond dynamically to external cues such as stress or hormones. This export process terminates nuclear signaling events, preventing aberrant activation and ensuring fidelity. For instance, NES-dependent export silences pathways like those involving proteins after stimulation, restoring cellular equilibrium. NES contribute significantly to RNA export by being present in adaptor proteins that mediate the formation and translocation of ribonucleoprotein complexes, including those for mRNA and rRNA. These signals interact with export receptors like CRM1 to facilitate the directed movement of RNAs from the to the , ensuring proper and . Dysregulation of NES function can lead to nuclear accumulation of proteins, disrupting cellular and impairing processes such as , , and . The presence of NES across eukaryotic organisms underscores their evolutionary conservation, reflecting a fundamental mechanism for nuclear-cytoplasmic compartmentalization that has been preserved to support essential cellular functions. This conservation highlights the indispensable role of NES in maintaining compartmentalized processes from to humans. Viruses, such as , exploit conserved NES pathways, like that of protein, to export their RNAs and hijack host machinery.

Structure and Motifs

Classical Leucine-Rich NES

The classical leucine-rich nuclear export signal (NES) is characterized by a consensus sequence consisting of a hydrophobic core pattern Φ-X_{2-3}-Φ-X_{2-3}-Φ-X-Φ, where Φ represents (L), (I), (V), (M), or (F), and X denotes any residue. This is typically approximately 10 residues in length and serves as the primary recognition element for the exportin CRM1 (also known as XPO1). The key structural features of this NES involve four conserved hydrophobic residues positioned at specific intervals relative to spacer regions: typically at positions 1, 4, 7, and 10 within the 10-residue span. These positions form a critical scaffold that enables interaction with the hydrophobic groove on CRM1. In the unbound state, classical NES sequences are frequently located within intrinsically disordered regions of proteins, providing the necessary flexibility for recognition and binding. Upon engagement with CRM1, the NES adopts an α-helical conformation, which stabilizes the cargo-exportin complex. Experimental validation of the classical NES has been established through studies, where substitution of the key hydrophobic leucines (or equivalent residues) with alanines abolishes nuclear export activity, as demonstrated in reporter assays using proteins like the inhibitor (PKI). The seminal identification of this motif in PKI highlighted its role in rapid nuclear extrusion. In the Eukaryotic Linear Motif () resource, this pattern is designated as the TRG_NES_CRM1_1 motif.

Non-Classical NES

Non-classical nuclear export signals () are protein sequence motifs that facilitate nuclear export through CRM1-dependent mechanisms with non-canonical motifs or via alternative exportin-mediated pathways, often lacking the hydrophobic leucine-rich consensus and instead relying on charged, phosphorylated, or structural features tailored to specific exportins and cargoes such as those involved in processing. These signals are typically shorter or more variable than classical , incorporating elements like phospho-serines or dsRNA-binding domains to enable recognition by alternative receptors including exportin-5 (and its yeast homolog ) or exportin-t. A prominent example is the in the Pho4, which mediates phosphate-dependent export in response to environmental cues. This 35-residue motif (residues 100–134) within Pho4's intrinsically disordered region is enriched in small polar and hydrophobic residues, adopting an extended zig-zagging conformation with three 90° turns upon binding. Phosphorylation at key serines (pS114 and pS128) by kinases like is essential, aligning these sites 30 Å apart to engage specific pockets on the exportin Msn5, forming a high-affinity interface (K_D ≈ 25 nM) spanning 1256.9 Ų across Msn5's concave HEAT repeat surface (h8–h18). A 2025 cryo-EM structure at 3.0–3.2 Å resolution reveals this dynamic assembly with RanGTP, highlighting Msn5's flexibility and the motif's deviation from leucine-rich patterns. Another key instance involves the phosphorylated adaptor for export (PHAX), which directs small nuclear RNAs (snRNAs) via a variant that integrates phosphorylation for complex stability. PHAX's (residues 117–136) features a helical binding CRM1's cleft, but deviates from classical motifs with a conserved (Q127) at the Φ0 position instead of a hydrophobic residue, potentially reducing basal affinity and requiring regulatory inputs. at up to five serine/ sites (e.g., in the ST2 region, residues 64–76) by CK2 enhances electrostatic interactions with RanGTP's basic surface, promoting synergistic assembly of the PHAX-CBC-snRNA subcomplex with CRM1-RanGTP. A 2025 cryo-EM at 2.45 Å elucidates this quaternary complex, showing PHAX bridging capped and exportin components while folding dependently on all partners to reinforce cap recognition and export specificity. In cases like tRNA export, non-classical signals often manifest as structural determinants rather than linear sequences; exportin-t recognizes mature tRNA conformations directly, independent of CRM1, while associated proteins such as eukaryotic elongation factor 1A (eEF1A) rely on exportin-5 binding via aminoacylated tRNA adaptors involving eEF1A's GTP- and tRNA-binding domains. Similarly, exportin-5 exports double-stranded RNA-binding proteins (dsRBPs) like PKR and ILF2 through their dsRNA-binding domains acting as structural export signals, enhanced by RanGTP without a discernible linear NES. These mechanisms underscore how non-classical NES prioritize cargo-specific recognition, often bypassing CRM1 for dedicated pathways in RNA biogenesis.

Export Mechanism

Cargo Recognition by Exportins

Cargo recognition by exportins begins with the specific interaction between the nuclear export signal (NES) on cargo proteins and the exportin receptor, primarily CRM1 (also known as XPO1), which facilitates the export of classical NES-bearing cargoes. CRM1 binds to the hydrophobic residues within the NES through a central hydrophobic groove formed by its HEAT-repeat , enabling selective of leucine-rich motifs. In the interface, the key residues of the insert into a series of hydrophobic pockets (typically four) along the CRM1 groove, with the adopting a predominantly extended conformation that can include helical elements for optimal fit. This interaction is characterized by moderate in the absence of cofactors, but it is significantly enhanced by the of Ran-GTP to a distinct site on CRM1, which allosterically stabilizes the NES association and promotes complex assembly. The resulting export complex forms a quaternary structure comprising the NES-linked , CRM1, and Ran-GTP, with a of 1:1:1, ensuring efficient packaging for translocation. Crystal structures from pre-2020 studies, such as those resolved for various NES peptides bound to CRM1-RanGTP, have elucidated this architecture, revealing how the NES leucines anchor into the groove while the remains flexibly tethered. CRM1 specifically recognizes NES sequences approximately 10 residues long, characterized by spaced hydrophobic elements, whereas non-classical NES cargoes, such as precursor microRNAs (pre-miRNAs), are handled by dedicated exportins like exportin-5 (XPO5), which binds structured dsRNA hairpins rather than linear motifs. Recent cryo-EM structures from , including those of CRM1 complexes with viral or cargoes, further confirm the helical conformation of certain NES peptides within the binding groove, highlighting conformational adaptability in cargo recognition.

Ran-GTP Dependent Transport

The Ran cycle provides the energy and directionality for nuclear export by establishing a steep concentration gradient of Ran-GTP, which is high in the due to the chromatin-bound RCC1 and low in the owing to the GTPase-activating protein RanGAP. This gradient promotes the assembly of the export complex—consisting of the exportin, NES-bearing cargo, and Ran-GTP—within the , where Ran-GTP binding induces a conformational change in the exportin that stabilizes cargo association. In the , RanGAP stimulates GTP on Ran, converting it to Ran-GDP and triggering dissociation of the trimeric complex, thereby releasing the cargo. This step is essential for complex disassembly and ensures the unidirectional flow of transport cycles. The translocation of the export complex through the nuclear pore complex (NPC) occurs via transient, multivalent interactions between hydrophobic patches on the exportin surface and phenylalanine-glycine (FG) repeats of FG-nucleoporins lining the NPC channel. These interactions facilitate rapid, energy-dependent passage across the permeability barrier, with the process being bidirectional in principle but strongly biased toward due to the Ran-GTP gradient, which favors complex stability in the and disassembly in the . Recent three-dimensional MINFLUX studies have revealed that and pathways overlap within the NPC, with NES-dependent export complexes traversing shared central regions of the pore scaffold, underscoring the NPC's efficient multiplexing of traffic directions. Upon reaching the cytoplasmic face of the NPC, the of Ran-GTP to Ran-GDP by RanGAP, often in concert with Ran-binding protein 1 (RanBP1), fully dissociates the exportin from both Ran and the cargo, enabling cargo release into the . The exportin and Ran-GDP are then recycled back to the nucleus via distinct mechanisms, including nuclear import of Ran-GDP by nuclear transport factor 2 (NTF2), which sustains the Ran gradient and allows multiple rounds of export. The primary exportin mediating NES-dependent transport is CRM1 (also known as XPO1). The NES contributes to export directionality by enhancing the bias toward net outward movement, as the affinity of NES for exportins correlates with the stability of the nuclear ternary complex under the Ran-GTP gradient. Transport efficiency through individual NPCs is regulated by this NES affinity, with rates typically on the order of several molecules per second per NPC for NES-bearing complexes, reflecting the balance between and NPC .

Regulation

Masking and Exposure

The activity of nuclear export signals (NES) is often regulated through physical masking, where the NES sequence is concealed within the three-dimensional structure of the protein or by interactions with binding partners, thereby preventing recognition by exportins such as CRM1. One common masking mechanism involves burial of the NES within folded protein domains, which sterically hinders access to the export machinery; for instance, in the tumor suppressor , the leucine-rich NES located in the tetramerization domain is masked in the tetrameric form, retaining in the until conditions favor disassembly. Another mechanism entails binding to nuclear retention factors that occlude the NES; in the transcription factor , association with DNA masks the NES, inhibiting export and promoting transcriptional activity at target sites. These strategies ensure that NES-dependent export occurs only under appropriate cellular contexts. Exposure of masked NES typically arises from triggers such as , which cleaves inhibitory domains to reveal the signal, conformational rearrangements that reposition the NES for accessibility, or dissociation of binding partners that previously shielded it. For example, in the Nrf2 transcription factor, heterodimerization with small Maf proteins masks the NES in the zipper motif, but stimulus-induced dissociation unmasks it, facilitating rapid nuclear exit. In , a shift from tetrameric to monomeric conformation exposes the NES, enabling CRM1-mediated export. Such unmasking events are tightly coupled to cellular signals, allowing precise spatiotemporal control of protein localization. This masking and exposure dynamic critically prevents premature nuclear export, maintaining proteins in the nucleus for their functions until specific cues demand relocation; in signaling pathways, stimulus-induced unmasking enables swift cytoplasmic redeployment, amplifying responses like stress adaptation or immune activation. For instance, in signaling, interactions with the inhibitor IκB contribute to regulated NES accessibility within the complex, ensuring export only post-nuclear activity to terminate transcription. These mechanisms underscore the role of structural control in NES regulation, complementary to other forms like . Experimental validation of masking and exposure often employs fluorescence microscopy to monitor nucleocytoplasmic shuttling in live cells, where disruption of masking interactions—via mutations or binding partner knockdown—results in altered localization patterns that confirm NES functionality. For example, tagging proteins with fluorescent reporters like GFP allows real-time tracking of export kinetics upon induced unmasking, revealing how conformational changes or dissociations accelerate shuttling rates by 2- to 5-fold in responsive systems. fusion assays complement this by quantifying redistribution between nuclei, but microscopy provides direct visualization of dynamic exposure events.

Post-Translational Modifications

Post-translational modifications (PTMs) play a crucial role in regulating the activity and accessibility of nuclear export signals (NESs), thereby controlling the nucleocytoplasmic shuttling of proteins. Among these, phosphorylation of serine or residues adjacent to the NES core often inhibits binding to the export receptor CRM1 (also known as exportin 1), preventing nuclear export and promoting nuclear retention. For instance, in hypoxia-inducible factor-1α (HIF-1α), (MAPK) phosphorylates serine residues 641 and 643 within the NES-containing MAPK target domain, which disrupts the interaction between the NES and CRM1, leading to HIF-1α nuclear accumulation and enhanced transcriptional activity under hypoxic conditions. Conversely, by nuclear phosphatases can reverse this inhibition; in the case of (ERKs, members of the MAPK family), dephosphorylation facilitates binding to NES-containing MEK1/2, enabling CRM1-dependent nuclear export and cytoplasmic reactivation. Specific kinases, such as cyclin-dependent kinases (CDKs), target sites flanking NES motifs to modulate export dynamics. CDK of sites within an NLS-NES module in cell-cycle regulators like Mcm3 promotes net nuclear during , as demonstrated by phosphomimetic mutations that enhance and impair replication initiation when mislocalized. This reversibility is maintained by phosphatases, which remove groups to restore NES functionality and allow dynamic of protein localization in response to cellular signals. Beyond phosphorylation, other PTMs fine-tune NES function by altering charge, conformation, or receptor affinity. Sumoylation, the covalent attachment of small ubiquitin-like modifier (SUMO) proteins, can enhance NES-mediated export; in p53, SUMOylation at lysine residues promotes its interaction with CRM1, facilitating nuclear export and cytoplasmic relocation to modulate stress responses. In contrast, cysteine oxidation inhibits NES recognition by CRM1; in the yeast transcription factor Yap1p, oxidative modification of flanking cysteine residues masks the NES, blocking CRM1 binding and causing nuclear accumulation under oxidative stress conditions. These modifications generally alter electrostatic interactions or induce conformational changes that either occlude or expose the hydrophobic NES core for receptor binding. A notable example of phosphorylation-dependent NES activation occurs in non-classical motifs, where phosphate groups directly coordinate with exportins. In the phosphate-sensing Pho4, at serines 114 and 128 within a 35-residue NES enables high-affinity binding to the exportin Msn5 (K_D ≈ 25 nM), as revealed by a 2025 cryogenic-electron showing phospho-serine coordination with conserved and residues in Msn5's binding pocket; weakens this interaction (K_D ≈ 2.4 µM), retaining Pho4 in the to activate phosphate-responsive genes. Thus, PTMs provide a versatile mechanism for signal-responsive control of NES accessibility, distinct from non-covalent masking strategies.

Examples

In Viral Proteins

Nuclear export signals (NES) in viral proteins enable viruses to exploit the host cell's CRM1-dependent export machinery, facilitating the relocation of viral components from the to the to support replication and evade host defenses. A prominent example is the HIV-1 protein, which contains a classical leucine-rich NES (residues 73-83) that binds directly to the exportin CRM1, allowing the export of unspliced and partially spliced viral mRNAs bound to the Rev (RRE). This NES-mediated process is essential for the cytoplasmic translation of structural proteins like and , bypassing the host's splicing-dependent mRNA export restrictions. Although Rev's RNA-binding domain is arginine-rich, its NES functions as a standard hydrophobic leucine-based motif, distinct from arginine-rich variants in other contexts. In adenoviruses, the E1B-55K protein harbors a CRM1-dependent NES that promotes the nuclear export of the E1B- complex, thereby inhibiting p53's transcriptional activity and facilitating viral oncogenesis. This export mechanism relocalizes p53 to cytoplasmic aggresomes for degradation, suppressing host antiviral responses during infection. Similarly, in , the NS2 (nuclear export protein, NEP) contains a conserved N-terminal (residues 12-21) that recruits CRM1 and acts as an adaptor to bridge the matrix protein M1-associated viral ribonucleoproteins (vRNPs), enabling their coordinated nuclear export and relocation to the cytoplasm for virion assembly. Viruses often incorporate multiple NES motifs within polyproteins to ensure synchronized export of viral complexes, as seen in retroviruses like , where the polyprotein's p10 domain NES facilitates the nuclear egress of and associated genomic , coordinating particle assembly. This strategy allows viruses to hijack and overload the host CRM1 pathway, evading regulatory checkpoints such as phosphorylation-dependent masking of cellular NES. In foamy viruses, a similar N-terminal NES in the polyprotein is required for efficient nuclear export and subsequent viral propagation. These NES elements are critical for viral and infectivity; for instance, mutations in the HIV-1 Rev NES disrupt mRNA export, severely impairing late-stage virus production and abolishing replication in . Likewise, alterations in the influenza NS2 NES or RSV Gag p10 NES lead to nuclear retention of vRNPs or polyproteins, resulting in defective virion assembly and loss of infectivity. Recent post-2020 studies have shown that the SARS-CoV-2 accessory protein ORF6 binds the Rae1-Nup98 complex at the , disrupting nucleocytoplasmic transport pathways, including CRM1-mediated nuclear export, thereby inhibiting host mRNA export and signaling to promote . This interference, more potent in SARS-CoV-2 than in , underscores ORF6's contribution to pathogenesis through targeted subversion of nucleocytoplasmic transport.

In Cellular Proteins

Nuclear export signals (NES) are integral to the function of numerous cellular proteins, enabling their shuttling from the to the to regulate diverse processes such as signaling, , and stress responses. Bioinformatic predictions suggest that over 1,000 proteins contain potential NES motifs, with experimental validation confirming their presence in hundreds of cases across functional categories including transcription factors, kinases, and metabolic enzymes. For instance, signal transducer and activator of transcription () family members, such as and , utilize NES to terminate nuclear signaling after stimulation, facilitating and recycling in the to prevent prolonged transcriptional activity. Similarly, metabolic regulators like the M2 isoform incorporate NES to modulate nuclear-cytoplasmic distribution in response to glycolytic demands. A seminal example of NES in cellular proteins is the protein kinase inhibitor (PKI), the first non-viral NES identified, which contains a classical leucine-rich motif that binds CRM1 to actively export the complex of PKI with (PKA) catalytic subunit from the nucleus. This export mechanism inhibits PKA activity in the cytoplasm, preventing ectopic nuclear signaling and maintaining spatial control over cAMP-dependent pathways. The NES in PKI, spanning residues 24-37 (LALKLAGLDI), exemplifies how such signals ensure rapid, energy-dependent translocation, as demonstrated by assays showing net extrusion within minutes. Survivin, an protein (IAP) family member, relies on its (residues 80-88, LPPLP) for dynamic nucleocytoplasmic shuttling that balances its dual roles in and cell survival. During , CRM1-mediated export prevents accumulation of survivin, thereby suppressing its anti-apoptotic function and sensitizing cells to stress-induced death; mutation of this abolishes export, leading to persistent localization and loss of regulation without affecting mitotic chromosome passenger complex assembly. In , transient retention allows survivin to localize to kinetochores, highlighting as a regulatory switch. Nuclear actin, a monomeric form of the cytoskeletal protein, incorporates two functional NES motifs (NES1 at residues 170-181 and NES2 at 211-222) that facilitate its export via Exportin 6, preventing excess accumulation that could disrupt dynamics. This export is crucial for recycling after its role in nuclear processes, such as binding to -remodeling complexes like INO80 and BAF to facilitate ATP-dependent repositioning during transcription and . Dysregulation of actin NES leads to nuclear retention, impairing remodeling efficiency and cellular . Transcription factors like NF-E2-related factor 2 (Nrf2) employ an in their Neh5 domain (residues 175-186, LLSIPELQCLNI) to control responses by enabling post-activation export from the . Upon , Nrf2 translocates to the to bind antioxidant response elements (ARE) and induce protective genes like oxygenase-1; subsequent NES-dependent export, insensitive to changes but modulated by , terminates this response to avoid overactivation. This relocation exemplifies how NES in transcription factors fine-tune temporal for adaptive cellular defense.

Biological Roles and Disease Implications

Cellular Functions

Nuclear export signals (NES) play crucial roles in various physiological processes by facilitating the relocation of regulatory proteins from the to the , thereby modulating their activity and availability for downstream functions. In control, NES-mediated export of proteins such as (PER) and BMAL1 fine-tunes transcriptional rhythms. For instance, mammalian PER1 contains a leucine-rich NES (amino acids 485–495) that promotes its cytoplasmic accumulation, preventing premature nuclear re-entry and ensuring oscillatory patterns essential for the . Similarly, BMAL1 shuttling via its NES motifs regulates CLOCK/BMAL1 heterodimer of E-box-containing genes like Per and Cry, while also controlling heterodimer through ubiquitin-proteasome pathways during active transcription phases. In the stress response, enables the timely relocation of heat shock factors (HSFs) to the post-stress, allowing recovery and preventing prolonged transcriptional activation. Heat shock factor 1 (HSF1), the master regulator of the , undergoes CRM1-dependent nuclear export mediated by its following stress resolution; inhibition of this export with leptomycin B reverses HSF1 cytoplasmic sequestration by 14-3-3ε, highlighting 's role in terminating chaperone gene transcription like Hsp70. This relocation supports the translation of stress-induced proteins in the , maintaining cellular . During the , NES ensures precise spatiotemporal localization of key regulators like , which is essential for mitotic progression. Survivin's , recognized by CRM1, drives its nuclear export to tether the chromosomal passenger complex to mitotic structures; homodimerization antagonizes this export, balancing survivin's nuclear and cytoplasmic pools to promote proper chromosome segregation and . Disruption of NES function impairs survivin's relocation, leading to mitotic defects. In RNA processing, NES in export adaptors such as TAP (also known as NXF1) facilitates bulk mRNA export through nuclear pores. TAP's novel NES, distinct from classical CRM1 pathways, supports its shuttling and binding to processed mRNAs via the constitutive transport element, enabling efficient nucleocytoplasmic transport of cellular and viral transcripts without relying on splicing. NES-mediated shuttling also contributes to embryonic development by regulating signaling pathways like Wnt/β-catenin. Although β-catenin lacks a classical NES, its nuclear export is facilitated by associated factors, allowing dynamic retention and release in the nucleus; Twa1/Gid8 acts as a retention factor that stabilizes nuclear β-catenin during Wnt activation, ensuring proper transcriptional control of developmental genes in dorsal-ventral patterning. For example, the (PKI) exemplifies classical NES function in shuttling cAMP-dependent , influencing developmental signaling.

Role in Pathologies

Dysregulation of nuclear export signals (NES) plays a critical role in cancer by enabling the cytoplasmic retention or enhanced export of oncoproteins, thereby promoting tumor survival and progression. In , an protein overexpressed in many cancers, the NES facilitates CRM1-mediated nuclear export, which is essential for its anti-apoptotic function and resistance to therapy-induced . Mutation or inhibition of this NES leads to nuclear accumulation of survivin, sensitizing cancer cells to , as demonstrated in studies on tumor cell lines where export blockade reduced survivin-mediated protection. A 2024 study revealed a noncanonical oncogenic involving a •GTP:RanGAP1 complex that enhances XPO1-dependent nuclear export of cargo proteins, independent of signaling, thereby driving cytoplasmic mislocalization of tumor suppressors and accelerating tumor growth in RAS-mutant cancers. In neurodegeneration, defects in NES-mediated shuttling of fragile X mental retardation protein (FMRP) contribute to (FXS), the leading genetic cause of , by impairing nuclear export of specific mRNAs essential for synaptic function. FMRP contains a functional NES that enables its nucleocytoplasmic shuttling and regulation of mRNA transport; loss of FMRP due to mutations disrupts this process, leading to aberrant dendritic mRNA localization and translational dysregulation in neurons. A missense variant in the FMRP NES has been linked to and behavioral issues, underscoring how NES impairment exacerbates FXS phenotypes. Recent reviews highlight that FMRP deficiency alters mRNA export pathways, contributing to synaptic deficits and neurodevelopmental abnormalities in FXS. NES in viral proteins facilitate replication by hijacking host nuclear export machinery, with implications for diseases like HIV and adenovirus infections. In HIV-1, the Rev protein's NES binds CRM1 to export unspliced viral mRNAs from the nucleus to the cytoplasm, enabling production of structural proteins and virion assembly critical for replication. Similarly, the adenovirus E4orf6 protein's NES supports CRM1-dependent export, forming complexes that promote viral mRNA cytoplasmic accumulation and efficient virus production during infection. Emerging evidence suggests potential roles for SARS-CoV-2 ORF proteins in modulating nuclear export; for instance, ORF6 disrupts bidirectional nucleocytoplasmic transport more potently than its SARS-CoV-1 counterpart, inhibiting host mRNA export and interferon signaling to favor viral replication in COVID-19. In , dysregulated -dependent export of transcription factors like contributes to by prolonging nuclear activity and excessive proinflammatory gene expression. complexes are exported via CRM1 using in , which sequesters in the under normal conditions; defects in this export lead to sustained nuclear , driving cytokine overproduction in autoimmune disorders such as . This dysregulation amplifies immune responses, linking alterations to inflammatory pathologies where hyperactivity promotes tissue damage and production. A 2025 study identified TSPYL5 as a driver of pathological export in tumors, where it binds G3BP1 to enhance its and nuclear membrane translocation, thereby accelerating RanBP2-mediated SUMOylation and CRM1-dependent export of , sequestering it in the and promoting progression. This mechanism highlights how hyperactivity in components like G3BP1 can suppress tumor suppressor functions, fostering aggressive tumor growth.

Therapeutic Targeting

CRM1 Inhibitors

CRM1 inhibitors target the nuclear export receptor CRM1 (also known as XPO1) to block the recognition and export of proteins bearing nuclear export signals (), thereby accumulating tumor suppressor proteins in the nucleus and promoting in cancer cells. These agents have been particularly explored for their therapeutic potential in malignancies characterized by dysregulated nuclear transport. Leptomycin B (LMB), a natural fungal derived from species, was the first identified CRM1 inhibitor. It covalently binds to the NES-binding groove of CRM1 at cysteine residue 528 (Cys528), irreversibly blocking the interaction with NES-bearing cargoes and halting nuclear export. Despite demonstrating potent preclinical anticancer activity at nanomolar concentrations, LMB's clinical development was halted due to severe , including gastrointestinal distress and , limiting its use to experimental settings. Selinexor (KPT-330), an orally bioavailable selective inhibitor of nuclear export (SINE), represents a more clinically viable CRM1 . Approved by the FDA in for use in with dexamethasone for relapsed or refractory after at least four prior therapies, selinexor traps NES-bound cargoes in the by forming a slowly reversible with Cys528 in CRM1's cargo-binding groove. In December 2020, selinexor received full approval for after at least one prior in with bortezomib and dexamethasone. In 2020, selinexor received accelerated approval for relapsed or refractory after at least two prior systemic therapies. This mechanism selectively impairs highly proliferative tumor cells, which often exhibit elevated CRM1 expression and nuclear export activity compared to normal cells, leading to nuclear retention of tumor suppressors like and IκB, and subsequent induction of . In clinical trials, selinexor has shown meaningful efficacy in hematologic malignancies. The phase 2b STORM trial reported an overall response rate of 26% and a median progression-free survival of 3.7 months in triple-class refractory multiple myeloma patients, with improved overall survival in responders. Common adverse effects include thrombocytopenia (73% incidence), fatigue (73%), and nausea (72%), often manageable with dose adjustments and supportive care. As of 2025, ongoing trials continue to evaluate its role in solid tumors such as endometrial cancer (e.g., phase 3 XPORT-EC-042 trial showing PFS benefits in TP53 wild-type advanced disease) and pediatric solid tumors including sarcomas, aiming to address unmet needs in advanced settings. Lower-dose and weekly schedules of selinexor have shown improved tolerability while maintaining efficacy in multiple myeloma.

Recent Developments

Recent advancements in nuclear export signal (NES) targeting have focused on developing more selective and safer inhibitors beyond traditional covalent binders, with emphasis on reversible CRM1 modulators entering clinical evaluation. The small-molecule LFS-1107 represents a promising next-generation reversible CRM1 , demonstrating potent antiproliferative effects in extranodal NK/T cell lymphoma models through non-covalent binding and a clear profile that may reduce off-target toxicities compared to earlier agents. Building on the foundation of , which established CRM1 inhibition as a viable anticancer , LFS-1107 remains in preclinical optimization as of 2025, highlighting improved safety in hematologic malignancies. Structural biology has accelerated NES-targeted drug design through high-resolution cryo-EM studies of NES-CRM1 complexes. In 2025, cryo-EM structures revealed the atomic details of HIV-1 Rev NES interactions within the CRM1-Ran nuclear export complex, enabling rational design of allosteric inhibitors that disrupt RanGTP-dependent cargo release with sub-nanomolar affinity. These insights have informed the development of conformation-specific binders for enhanced selectivity.

Resources

Databases

NESbase version 1.0, released in 2004 by researchers at the (DTU) and the (UCPH), serves as a foundational curated repository of experimentally validated leucine-rich nuclear export signals (NESs). The database compiles data from over 200 published articles, focusing on NESs not annotated in major protein databases like SWISS-PROT at the time, and includes 75 proteins with 80 distinct NESs confirmed through experimental methods such as mutational analysis and shuttling assays. Each entry in NESbase provides detailed information on the NES sequence, its position within the parent protein, a conservation score derived from a analysis, the associated protein (cross-referenced to SWISS-PROT/TrEMBL accessions), organism of origin, experimental evidence, and literature references. Users can search the database by organism (e.g., Homo sapiens or ) or by motif sequence to retrieve matching entries, facilitating targeted queries for specific biological contexts. Notable examples include the NES from HIV-1 Rev protein (positions 75-84: LQLPPLERLT) and the (PKI) NES (positions 37-46: LALKLAGLDI), both verified for CRM1-dependent export. Maintenance of NESbase has been limited, with no updates since its 2004 release, though the resource remains accessible via the DTU Health Tech services for download in text format. NESdb, developed by the Chook Laboratory and first released in 2012, is a comprehensive database of CRM1-dependent NES-containing cargoes, updated in 2021 to include 399 experimentally validated entries from proteins and some RNAs. It catalogs NES sequences, their positions, experimental validation methods (e.g., shuttling assays, CRM1 binding), associated cargoes, organisms, and references, serving as a key resource for nuclear export research. Users can browse or search by cargo name, NES sequence, or organism; examples include NESs from (positions 305-317: LRLRQALGELRL) and HIV-1 . The database is accessible online and supports downloads. Complementary to these, the Eukaryotic Linear Motif () resource catalogs NES motifs under the identifier TRG_NES_CRM1_1, offering a pattern (L-x[2,3]-[LIVFM]-x[2,3]-L-x-[LI]) for classical leucine-rich NESs recognized by CRM1/exportin-1, while also integrating instances of non-classical variants through curated instances across eukaryotic proteomes. is actively maintained, with updates as of 2024. These databases support annotation of NESs in newly sequenced proteins and validation of predictions from computational tools, enabling researchers to cross-reference experimental data for functional studies in nuclear transport.

Prediction Tools

Several computational tools have been developed to predict nuclear export signals () in protein sequences, primarily focusing on classical leucine-rich motifs recognized by the CRM1/exportin-1 pathway. These tools employ approaches, such as neural networks and hidden Markov models, to scan sequences for patterns characterized by hydrophobic residues (especially leucines) spaced at specific intervals, typically following the consensus Φ-X_{2-3}-Φ-X_{2-3}-Φ-X-Φ (where Φ is a hydrophobic residue). Predictions help annotate potential export activity but are probabilistic and require experimental verification. NetNES, a hosted by DTU Tech, uses a combination of neural networks and hidden Markov models to identify leucine-rich NES in eukaryotic proteins. The tool generates scores based on hydrophobicity and residue spacing, reflecting the biophysical properties essential for CRM1 binding; higher scores indicate stronger predicted export potential. Trained on experimentally validated NES, NetNES has been widely applied since its release, demonstrating utility in analyzing viral and cellular proteins. NESmapper is a web-based predictor that enhances accuracy by using activity-based profiles derived from optimization of properties and positional weights. It maps potential while assigning flexibility scores to account for variability in disordered protein regions, where many NES occur, thereby reducing false positives compared to earlier methods. This approach yields improved sensitivity for classical NES, making it suitable for . Machine learning models since 2020 have begun incorporating structural predictions, such as from , and post-translational modifications like to refine NES identification in research contexts, often trained on databases like NESdb. However, dedicated NES-specific tools remain limited. Tool predictions are validated through experiments, where key residues are altered to assess export disruption, or nuclear shuttling assays monitoring protein localization. Accuracies for classical NES typically range from 70-80% in benchmark tests against verified datasets, with tools like NESmapper and LocNES outperforming older consensus-based methods in . A key limitation of these tools is their poor performance on non-classical NES, which deviate from leucine-rich patterns and rely on alternative pathways or structures, often resulting in high false-negative rates. All predictions necessitate experimental confirmation, as sequence-based methods cannot fully capture contextual factors like or interactions.

References

  1. [1]
    The Transport of Molecules between the Nucleus and the Cytosol
    These proteins contain nuclear export signals that are recognized by export receptors that guide the RNA out of the nucleus through nuclear pore complexes. Upon ...
  2. [2]
    Nuclear Transport
    Nuclear export is mediated by different transport receptors called exportins, which recognize proteins containing a Nuclear Export Signal, or NES. mRNA and ...
  3. [3]
    Sequence and structural analyses of nuclear export signals in the ...
    Abstract. We compiled >200 nuclear export signal (NES)–containing CRM1 cargoes in a database named NESdb. We analyzed the sequences and three-dimensional ...
  4. [4]
    The CRM1 nuclear export protein in normal development and disease
    ... nuclear export signal (NES). Included in the cadre of cargo are multiple tumor suppressor and oncoproteins as p53, BRCA1, Survivin, NPM, and APC, which ...
  5. [5]
    Identification of a signal for rapid export of proteins from the nucleus
    Aug 11, 1995 · The heat-stable inhibitor (PKl) of cAPK contains a nuclear export signal (NES) that triggers rapid, active net extrusion of the C-PKl complex from the nucleus.
  6. [6]
    Prediction of leucine-rich nuclear export signal containing proteins ...
    The classical nuclear export signal (NES), also known as the leucine-rich NES, is a protein localization signal often involved in important processes such as ...
  7. [7]
    Nuclear export signal located within the DNA‐binding ... - EMBO Press
    This family of transcription factors was first identified in the interferon signal pathway.Missing: discovered | Show results with:discovered
  8. [8]
    Regulation of Stat3 nuclear export - JCI
    Upon cytokine stimulation, they rapidly translocate to the nucleus, where they promote the expression of target genes. During the subsequent period of signal ...
  9. [9]
    A nuclear export signal and phosphorylation regulate Dok1 ...
    Nuclear export modulated by external stimuli and phosphorylation may be a mechanism by which Dok1 is maintained in the cytoplasm and membrane, thus regulating ...Missing: importance | Show results with:importance
  10. [10]
    Inhibition of mRNA export in vertebrate cells by nuclear ... - PNAS
    These results provide direct evidence that mRNA export in vertebrates depends on interactions between an NES and its cognate NES receptors. PKI NES conjugates ...<|separator|>
  11. [11]
    The nuclear export receptor CRM1/XPO1 and its diverse cargoes
    Sep 27, 2025 · CRM1 (Exportin 1, XPO1), the best-characterized nuclear export receptor, exports hundreds of proteins and various RNA species.
  12. [12]
    Altered Nuclear Export Signal Recognition as a Driver of Oncogenesis
    This discovery that mutant XPO1 can functionally drive tumorigenesis may be particularly useful for the development of targeted therapies for malignancies ...
  13. [13]
    An evolutionarily conserved nuclear export signal ... - PubMed - NIH
    An evolutionarily conserved nuclear export signal facilitates cytoplasmic localization of the Tbx5 transcription factor. Mol Cell Biol. 2008 Mar;28(5):1553-64.
  14. [14]
    Sequence and structural analyses of nuclear export signals in the ...
    Jul 25, 2012 · Nuclear localization signals (NLSs) direct proteins into the nucleus, whereas nuclear export signals (NESs) direct export of proteins from the ...
  15. [15]
    Nuclear Export Signal Consensus Sequences Defined Using a ...
    Nov 20, 2008 · NESs that are recognized by CRM1 (i.e., leucine-rich NESs) typically contain large hydrophobic conserved residues separated by a variable number ...<|control11|><|separator|>
  16. [16]
    The NES–Crm1p export pathway is not a major mRNA export route ...
    It can also be considered an RNA exporter: Rev, the founding NES family member, exports viral RNAs via the CRM1 pathway (Stutz and Rosbash, 1998). In Xenopus ...
  17. [17]
    Accurate Prediction of Leucine-Rich Nuclear Export Signals Using ...
    Sep 18, 2014 · The proposed consensus sequence, designated the “classical consensus”, is Φ–X2,3–Φ–X2,3–Φ–X–Φ, where Φ represents L, I, V, M, or F and X2,3 ...
  18. [18]
    a database of validated leucine-rich nuclear export signals - PMC
    Some previous work has defined a consensus sequence for NESs as [LIVFM]-x-(2,3)-[LIVFM]-x(2,3)-[LIVFM]-x-[LIVFM], where x is any amino acid (12). However, we ...
  19. [19]
    Nuclear export receptor CRM1 recognizes diverse conformations in ...
    Mar 10, 2017 · NESs adopt diverse conformations to bind CRM1, sharing only a small one turn of the helix structural element. · The Lys568 of HsCRM1 is important ...
  20. [20]
    Prediction of nuclear export signals using weighted regular ...
    Finally ELM (Gould et al., 2010) is a database of eukaryotic linear motifs described as regular expressions, including the 'TRG_NES_CRM1_1' that allows ...
  21. [21]
    Phosphate-dependent nuclear export via a non-classical NES class ...
    Mar 16, 2025 · These findings characterize a mechanism of phosphate-specific recognition mediated by a non-classical signal distinct from that for Exportin-1.
  22. [22]
  23. [23]
    Phosphate-dependent nuclear export via a non-classical NES class ...
    Phosphate-dependent nuclear export via a non-classical NES class recognized by exportin Msn5. Nat Commun. 2025 Mar 16;16(1):2580. doi: 10.1038/s41467-025- ...
  24. [24]
    Structural basis for the synergistic assembly of the snRNA export ...
    Jul 3, 2025 · Here we present the cryo-electron microscopy structure of the human snRNA export complex comprising phosphorylated PHAX, CBC, CRM1–RanGTP and capped RNA.
  25. [25]
    Structural basis for the synergistic assembly of the snRNA export ...
    Jul 3, 2025 · 2025 ... Here we present the cryo-electron microscopy structure of the human snRNA export complex comprising phosphorylated PHAX, CBC, CRM1-RanGTP ...
  26. [26]
  27. [27]
    Structural basis for leucine-rich nuclear export signal recognition by ...
    The LR-NES interface explains the consensus hydrophobic pattern, preference for intervening electronegative residues and inhibition by leptomycin B. The second ...
  28. [28]
    Structural basis for cooperativity of CRM1 export complex formation
    Dec 31, 2012 · The extended conformation of free CRM1, and, consequently, the closed state of the NES binding cleft, seems to be stabilized by the rearranged C ...Results · Crystal Structure Analysis · Md Simulations
  29. [29]
    Exportin-5 mediates the nuclear export of pre-microRNAs and short ...
    We demonstrate that human pre-miRNA nuclear export, and miRNA function, are dependent on Exportin-5. Exportin-5 can bind pre-miRNAs specifically in vitro.Missing: classical | Show results with:classical
  30. [30]
    The nuclear export receptor CRM1/XPO1 and its diverse cargoes
    Sep 27, 2025 · The SPN1 NES (1MEELSQALASSFSV14) binds the CRM1 groove at repeats h11 and h12. The NES adopts a helix-strand conformation: residues 1–12 form an ...
  31. [31]
    The HIV-1 nuclear export complex reveals the role of RNA in CRM1 ...
    Aug 21, 2025 · Our studies demonstrate that the binding of two NES peptides from the Rev dimer largely drives the dimeric form of CRM1. The CRM1 dimer ...
  32. [32]
    Ran and Nuclear Transport* - Journal of Biological Chemistry
    Sep 4, 1998 · Ran, a small GTPase, plays a central role in nuclear transport, coordinating traffic and triggering transport complex assembly/disassembly.
  33. [33]
    Ran-dependent nuclear export mediators: a structural perspective
    Nuclear export is an essential eukaryotic activity. It proceeds through nuclear pore complexes (NPCs) and is mediated by soluble receptors that shuttle between ...Introduction · Ran-Controlled Cargo Loading... · Rna Export Mediated By Exp5
  34. [34]
    RanGTPase: A Key Regulator of Nucleocytoplasmic Trafficking - PMC
    To summarize, RanGTPase plays a role in the nuclear export in such a way that nuclear GTP-bound Ran triggers the aggregation of Ran:exportin:cargo trimeric ...Missing: seminal papers
  35. [35]
    Nuclear export of the pre-60S ribosomal subunit through single ...
    Oct 27, 2021 · Here we analyse the export kinetics of the large ribosomal subunit (pre-60S particle) through single NPCs in human cells.
  36. [36]
    Overlapping nuclear import and export paths unveiled by two-colour ...
    Mar 19, 2025 · Here we used 3D MINFLUX 2 to identify the nuclear pore scaffold and then to simultaneously monitor both nuclear import and nuclear export.
  37. [37]
    A Role for RanBP1 in the Release of CRM1 from the Nuclear Pore ...
    Taken together, our results indicate that RanGTP is important for the targeting of export complexes to the cytoplasmic side of the NPC and that RanBP1 and ...
  38. [38]
    The RanGTP Pathway: From Nucleo-Cytoplasmic Transport to ...
    In this mini-review we will describe briefly how the RanGTP system regulates the nucleo-cytoplasmic shuttling of components in interphase.Abstract · The Nucleo-cytoplasmic... · Understanding the RanGTP... · ConclusionsMissing: seminal papers
  39. [39]
    The direction of transport through the nuclear pore can be inverted
    Ran-GTP is predominantly located in the nucleus and has been shown to regulate cargo binding and release of import and export receptors in their respective ...Missing: papers | Show results with:papers
  40. [40]
    A leucine-rich nuclear export signal in the p53 tetramerization domain
    We show that p53 export is mediated by a highly conserved leucine-rich nuclear export signal (NES) located in its tetramerization domain.
  41. [41]
    Nuclear export signal located within theDNA-binding domain of the ...
    We identify a functional NES in STAT1 and provide evidence for a novel mechanism that could regulate export by masking the NES when STAT1 is associated with DNA ...
  42. [42]
    Heterodimerization with Small Maf Proteins Enhances Nuclear ...
    Heterodimerization with Small Maf Proteins Enhances Nuclear Retention of Nrf2 via Masking the NESzip Motif ... A NES motif is characterized in the tetramerization ...
  43. [43]
    The NF-κB Family of Transcription Factors and Its Regulation - PMC
    NF-κB consists of a family of transcription factors that play critical roles in inflammation, immunity, cell proliferation, differentiation, and survival.
  44. [44]
    Nuclear export of actin: a novel mechanism regulating the ... - NIH
    In this paper we report that actin contains two leucine-rich type nuclear export signal (NES) sequences in the middle part of the molecule, which are both shown ...Missing: masking exposure
  45. [45]
    Fluorescence-based Quantification of Nucleocytoplasmic Transport
    Nov 10, 2018 · Here we present some important technical considerations for studying nuclear and cytoplasmic localization, and provide guidance for quantifying protein levels.2. Materials And Methods · 2.1. Microscope... · 3. Calculations
  46. [46]
    Optogenetic control of nuclear protein export - PMC - PubMed Central
    Feb 8, 2016 · Regulation of protein import and export is achieved mainly by masking and unmasking of nuclear import and nuclear export signals (NLSs and NESs) ...<|control11|><|separator|>
  47. [47]
    Atypical CRM1-dependent Nuclear Export Signal Mediates ...
    Localization, reporter gene, and co-immunoprecipitation assays demonstrate that the identified NES interacts with CRM1 in a phosphorylation-sensitive manner.
  48. [48]
    Nuclear Export of ERK3 by a CRM1-dependent Mechanism ...
    The cytoplasmic localization of ERK3 requires active nuclear export by a CRM1-dependent mechanism. Contrary to other MAP kinase family members, ERK3 binds ...
  49. [49]
    CDK Phosphorylation of a Novel NLS-NES Module Distributed ...
    Phosphomimic mutations of the Mcm3 CDK consensus sites promote net nuclear export of Mcm proteins in G1 phase and this mislocalization impairs replication ...
  50. [50]
    p53 SUMOylation promotes its nuclear export by facilitating its ...
    Jul 3, 2013 · Here we show that tethering of a small, ubiquitin-like modifier (SUMO) moiety to p53 markedly increases its cytoplasmic localization. SUMO ...
  51. [51]
    Crm1p mediates regulated nuclear export of a yeast AP‐1‐like ...
    Recognition of Yap1p by Crm1p is inhibited by oxidation, and this inhibition requires at least one of the three cysteine residues flanking the NES. These ...
  52. [52]
    Phosphorylation meets nuclear import: a review
    Dec 23, 2010 · It is unclear whether the ERK5 NES region recruits Crm1 directly, or via another NES-containing binding partner. Undoubtedly, phosphorylation ...
  53. [53]
    Identification of a signal for rapid export of proteins from the nucleus
    The heat-stable inhibitor (PKI) of cAPK contains a nuclear export signal (NES) that triggers rapid, active net extrusion of the C-PKI complex from the nucleus.
  54. [54]
    The Survivin–Crm1 interaction is essential for chromosomal ...
    Nov 10, 2006 · We show that the nuclear export receptor Crm1 is crucially involved in tethering the CPC to the centromere by interacting with a leucine‐rich nuclear export ...
  55. [55]
    Active maintenance of nuclear actin by importin 9 supports ... - PNAS
    Feb 9, 2012 · Actin is a component of many chromatin remodelling complexes (2, 3) ... actin would contain nuclear export signal (NES) sequences (26).Results · Nuclear Export Of Actin Is... · Nuclear Import Of Actin Is...
  56. [56]
    Mechanisms of Nuclear Actin in Chromatin Remodeling Complexes
    Moreover, actin has nuclear export signals (NES1 and NES2) in its primary sequence, which were shown to be functional and used to remove excess actin ...Missing: NES | Show results with:NES
  57. [57]
    Nuclear import and export signals in control of Nrf2 - PubMed - NIH
    In this study, we have identified nuclear import and export signals of Nrf2 and show that the nuclear import and export of Nrf2 is regulated by antioxidants.
  58. [58]
    Nuclear export of mammalian PERIOD proteins - PubMed - NIH
    Dec 7, 2001 · In mammals, nuclear localization of the circadian regulators PER1-3 is controlled by multiple mechanisms, including multimerization with PER and ...
  59. [59]
    BMAL1 shuttling controls transactivation and degradation ... - PubMed
    We show here that nucleocytoplasmic shuttling of BMAL1 is essential for transactivation and for degradation of the CLOCK/BMAL1 heterodimer.
  60. [60]
    Regulation of molecular chaperone gene transcription involves the ...
    Leptomycin B, a specific inhibitor of nuclear export receptor CRM1, was found to reverse the cytoplasmic sequestration of HSF1 mediated by 14-3-3 epsilon, ...
  61. [61]
    Homodimerization antagonizes nuclear export of survivin - PubMed
    In mitosis, Survivin is a key regulator of cell division, while in interphase, Survivin is able to protect cells from apoptosis. Survivin shuttles between ...
  62. [62]
    The human Tap protein is a nuclear mRNA export factor ... - PubMed
    Together, these data validate Tap as the first human sequence-specific nuclear mRNA export factor and identify a novel type of NES that can support nuclear mRNA ...
  63. [63]
    Twa1/Gid8 is a β-catenin nuclear retention factor in Wnt signaling ...
    Aug 22, 2017 · Here, we report that Twa1/Gid8 is a key nuclear retention factor for β-catenin during Wnt signaling and colorectal carcinogenesis.
  64. [64]
    Inhibition of CRM1-dependent nuclear export sensitizes malignant ...
    Clinical trials using the classic CRM1 inhibitor leptomycin B proved too toxic for patients; however, a new generation of less toxic small molecule inhibitors ...
  65. [65]
    [PDF] XPOVIO (Selinexor) MULTI-DISCIPLINE REVIEW - accessdata.fda.gov
    Mar 13, 2019 · XPOVIO is indicated in combination with dexamethasone, for the treatment of patients with relapsed refractory multiple myeloma who have received ...
  66. [66]
    Molecular mechanism and therapeutic implications of selinexor ...
    These inhibitors specifically and reversibly bind to residue Cys528 in the cargo-binding groove of XPO1. Selinexor (KPT-330) is the most advanced oral inhibitor ...
  67. [67]
    Oral Selinexor–Dexamethasone for Triple-Class Refractory Multiple ...
    Aug 21, 2019 · The most common adverse events that emerged during treatment were thrombocytopenia (in 73% of the patients), fatigue (in 73%), nausea (in 72%), ...
  68. [68]
    Study Details | NCT05611931 | Selinexor in Maintenance Therapy ...
    The purpose of this study is to evaluate the efficacy and safety of selinexor as a maintenance treatment in patients with p53 wt endometrial carcinoma (EC).<|control11|><|separator|>
  69. [69]
    Discovery and biological evaluation of a potent small molecule ...
    Furthermore, the results revealed that LFS-1107 is a reversible CRM1 inhibitor with clear dissociation process which may implicate a low toxicity profile. This ...
  70. [70]
    Exportin-T Promotes Breast Cancer Progression via PI3K/AKT ... - NIH
    May 21, 2025 · Studies have shown that XPOT promotes tumor progression in several cancers, including pleural mesothelioma, hepatocellular carcinoma, and ...
  71. [71]
    In vivo human T cell engineering with enveloped delivery vehicles
    Jan 11, 2024 · We find that Cas9-EDVs achieve targeted genome editing within in vivo-generated chimeric antigen receptor (CAR) T cells in mice with a humanized immune system.
  72. [72]
    Structural basis for the synergistic assembly of the snRNA export ...
    Dubiez and colleagues present a cryo-EM structure of the complex responsible for nuclear export of pre-small nuclear RNAs, comprising CBC–PHAX–CRM1–RanGTP. The ...
  73. [73]
    Targeting the SHOC2–RAS interaction in RAS-mutant cancers | Nature
    May 7, 2025 · In vitro high-throughput screening enabled the discovery of small molecules that bind to SHOC2 and disrupt the interaction with NRAS(Q61*).Missing: NES crosstalk
  74. [74]
    NXT2 is a key component of the RNA nuclear export factor complex ...
    Jul 7, 2025 · The study introduces NXT2 as a candidate gene for male infertility and shows that the encoded protein is involved in RNA nucleocytoplasmic ...Missing: modulators | Show results with:modulators
  75. [75]
    NESbase 1.0 - DTU Health Tech
    NESbase is a database of experimentally validated Leucine-rich Nuclear Export Signals (NES) curated from literature. These signals are not annotated in ...
  76. [76]
    Analysis and prediction of leucine-rich nuclear export signals
    Nuclear export signals (NESs) are extremely important regulators of the subcellular location of proteins. This regulation has an impact on transcription and ...
  77. [77]
    NetNES 1.1 - DTU Health Tech - Bioinformatic Services
    The NetNES 1.1 server predicts leucine-rich nuclear export signals (NES) in eukaryotic proteins using a combination of neural networks and hidden Markov models.
  78. [78]
    NESmapper: Accurate Prediction of Leucine-Rich Nuclear Export ...
    Sep 18, 2014 · A computational tool, NESmapper, to predict NESs by using profiles that had been further optimized by training and combining the amino acid properties of the ...
  79. [79]
    NESbase version 1.0: a database of nuclear export signals
    The compiled information was used to make a sequence logo of the Leucine-rich NESs, displaying the conservation of amino acids within a window of 25 ...
  80. [80]
  81. [81]
    LocNES: a computational tool for locating classical NESs in CRM1 ...
    NES peptides are usually 8–15 amino acids long with regularly spaced conserved hydrophobic residues. The first NES consensus of L-X2,3-[LIVFM]-X2,3-L-X-[LI] was ...
  82. [82]
    Prediction of leucine-rich nuclear export signal containing proteins ...
    The classical nuclear export signal (NES), also known as the leucine-rich NES, is a protein localization signal often involved in important processes such ...