ARL5B Human (ADP-ribosylation factor-like protein 5B) is a small GTPase belonging to the RAS superfamily. It shares structural homology with ADP-ribosylation factors (ARFs) and is closely related to ARL5, with which it shares 80% sequence identity . The human protein is 299 amino acids long (full-length) and has a molecular weight of approximately 33 kDa . Recombinant versions are typically produced in E. coli with modifications such as N-terminal His-tags for purification .
Retrograde Transport: ARL5B interacts with effectors to regulate membrane fusion at the TGN .
Viral Modulation: Human rhinovirus 16 (HRV16) upregulates ARL5B in macrophages, impairing phagosome maturation and bacterial clearance .
ARL5B is implicated in breast cancer via the FTO/miR-181b-3p/ARL5B pathway:
Viral Context | ARL5B Activity | Outcome |
---|---|---|
HRV16 Infection | Upregulated in macrophages | Impaired phagosome maturation |
Permissive Cells | Overexpression | Inhibits viral egress |
Key Study: HRV16-induced ARL5B upregulation stalls EEA1+ and CD63+ phagosomes, reducing bacterial clearance .
Tissue | Expression Level | Subcellular Localization |
---|---|---|
Brain | High | Golgi apparatus |
Liver | Moderate | TGN/endosomes |
Cancer Tissues | Variable | Diverse cellular compartments |
ADP-ribosylation factor-like protein 5B, ADP-ribosylation factor-like protein 8, ARL5B, ARL8.
MGSSHHHHHH SSGLVPRGSH MGLIFAKLWS LFCNQEHKVI IVGLDNAGKT TILYQFLMNE VVHTSPTIGS NVEEIVVKNT HFLMWDIGGQ ESLRSSWNTY YSNTEFIILV VDSIDRERLA ITKEELYRML AHEDLRKAAV LIFANKQDMK GCMTAAEISK YLTLSSIKDH PWHIQSCCAL
TGEGLCQGLE WMTSRIGVR.
ARL5B is a human protein belonging to the small GTPase superfamily, specifically within the Arf family . It is a 299-amino acid protein that functions as a molecular switch by binding and exchanging GTP and GDP . The protein contains characteristic GTPase domains that enable it to cycle between active (GTP-bound) and inactive (GDP-bound) states, similar to other members of the Arf family. ARL5B is also known as ADP-ribosylation factor-like protein 5B or sometimes referred to as ADP-ribosylation factor-like protein 8 . In humans, ARL5B has a closely related paralogue called ARL5A, which shares approximately 80% sequence identity . This conservation suggests important cellular functions that have been maintained throughout evolution.
ARL5B primarily localizes to the trans-Golgi network (TGN) in human cells . For visualization of ARL5B, researchers typically employ fluorescent protein fusions (such as ARL5B-GFP) or immunofluorescence techniques using specific antibodies against ARL5B. The localization can be confirmed through co-staining with established TGN markers. When performing subcellular fractionation experiments, ARL5B predominantly appears in the Golgi-enriched fractions. For more precise localization studies, immunogold electron microscopy can determine the exact positioning of ARL5B within the Golgi subcompartments. Notably, the localization pattern of ARL5B appears to be conserved across species, as both human ARL5B and the Drosophila Arl5 orthologue demonstrate trans-Golgi localization .
Researchers can produce recombinant human ARL5B protein using bacterial expression systems, particularly Escherichia coli . For optimal production, the full-length human ARL5B sequence (1-299 amino acids) can be cloned into appropriate expression vectors, such as those with GST or His-tags for purification purposes. Commercial recombinant proteins are available with features such as His-tags (e.g., M G S S H H H H H H S S G L V P R G S H...) and can achieve >90% purity suitable for various applications including SDS-PAGE and mass spectrometry . For functional studies, site-directed mutagenesis can generate specific variants of ARL5B, such as T30N (GDP-locked, inactive) and Q70L (GTP-locked, active) forms that are valuable for affinity chromatography experiments to identify protein interactors . When using E. coli expression systems, optimizing induction conditions (temperature, IPTG concentration, duration) can significantly improve protein yield and solubility.
ARL5B expression can be studied at both RNA and protein levels. For RNA analysis, RT-PCR provides a reliable method for detecting ARL5B transcripts in different cell types or tissues . When designing RT-PCR experiments, researchers should consider using random primers for the reverse transcription step, followed by amplification with gene-specific primers . For quantitative assessment, qRT-PCR allows precise measurement of ARL5B expression levels, as demonstrated in studies examining the threefold upregulation of ARL5B transcripts in HRV16-treated compared to control macrophages . At the protein level, Western blotting using specific antibodies can detect endogenous ARL5B. Proteomic approaches have provided insights into ARL5B abundance, with one study estimating approximately 6,885 copies per HeLa cell, while its paralogue ARL5A was below detection limits in the same analysis . For tissue-specific expression patterns, immunohistochemistry or in situ hybridization can reveal spatial distribution across different cell types.
ARL5B plays a critical role in endosome-to-Golgi trafficking primarily through its interaction with the Golgi-associated retrograde protein (GARP) complex . The GARP complex functions as a tethering factor for vesicles moving from endosomes to the trans-Golgi network (TGN), and ARL5B appears to be one of the key factors directing the recruitment of this complex to the trans-Golgi .
To investigate this function, researchers can employ several methodologies:
Affinity chromatography approaches: Both GST-fusion protein columns and liposome-based systems with GTP-locked (active) or GDP-locked (inactive) ARL5B mutants can isolate interacting partners from cell lysates. These techniques have successfully identified GARP complex components (Vps51, Vps52, Vps53, and Vps54/Scat) as specific binding partners of GTP-bound ARL5B .
Immunofluorescence co-localization studies: These can determine the effect of ARL5B depletion on GARP complex localization. In both Drosophila tissues and human cells, loss of ARL5B results in partial displacement of GARP components from the Golgi to the cytosol .
Functional rescue experiments: Expression of siRNA-resistant forms of ARL5B can restore GARP localization in knockdown cells, confirming specificity. Interestingly, expression of the paralogue ARL5A can also rescue GARP recruitment in cells depleted of ARL5B, suggesting functional redundancy despite differences in expression levels .
Endosomal compartment analysis: In ARL5B-deficient systems, the late endosomal compartment becomes enlarged, consistent with impaired endosome-to-Golgi traffic . This phenotype can be quantified through morphometric analysis of labeled endosomal compartments.
Recent research has uncovered an unexpected role for ARL5B in viral immunity and bacterial clearance pathways. Human rhinovirus 16 (HRV16) significantly upregulates ARL5B expression in human macrophages at both transcript and protein levels . This upregulation appears to be a specific response to viral replication, as UV-inactivated virus does not induce similar effects.
To investigate this function, researchers have employed several approaches:
Transcriptomic analysis: RNA sequencing identified ARL5B as one of the most consistently and significantly upregulated genes in HRV16-challenged human monocyte-derived macrophages (hMDMs) compared to control conditions . This finding was validated using RT-qPCR, which confirmed a threefold increase in ARL5B transcripts in virus-treated cells .
Protein quantification: Western blotting confirmed that the transcriptional upregulation translates to increased ARL5B protein levels .
siRNA-mediated depletion: Knockdown of ARL5B using specific siRNAs (siARL5b.1 or siARL5b.2) restored phagosome maturation in HRV16-treated cells, as evidenced by proper recruitment of Lamp1 to phagosomes . This suggests that ARL5B functions as a mediator of the virus-induced impairment of bacterial clearance.
Functional assays in permissive cells: In cells that support HRV16 replication (HeLa Ohio), ARL5B appears to act as a restriction factor for viral propagation , suggesting a complex relationship between the virus and this host protein.
These findings suggest a model where HRV16 induces ARL5B expression, which in turn impairs phagosome maturation and bacterial clearance while potentially restricting viral replication in certain cell types.
Despite sharing approximately 80% sequence identity, ARL5A and ARL5B exhibit interesting functional differences that can be investigated through several methodological approaches:
Expression analysis: Proteomic studies have detected differential expression patterns, with ARL5B being more readily detectable in HeLa cells (estimated at 6,885 copies per cell) while ARL5A levels were below detection limits . RT-PCR analysis confirms that both genes are expressed in human cells, but potentially at different levels across tissues .
Knockdown studies: siRNA-mediated depletion experiments reveal that knockdown of ARL5B, but not ARL5A, results in substantial redistribution of GARP complex components (e.g., myc-Vps54) from the TGN to the cytoplasm in HeLa cells . Specifically, only 30% of cells maintained myc-Vps54 at the TGN after ARL5B depletion, compared to 94% in the case of ARL5A depletion .
Functional complementation assays: Despite the apparent dispensability of ARL5A for GARP localization under normal conditions, expression of ARL5A-GFP can fully restore targeting of myc-Vps54 in cells depleted of ARL5B . This suggests that both proteins possess the intrinsic ability to direct GARP recruitment, but their relative contributions may depend on their expression levels in specific cell types.
Binding partner analysis: Comparative affinity chromatography using GTP-locked forms of both proteins could identify unique binding partners that might explain functional differences beyond GARP complex recruitment.
These findings suggest a model where ARL5B serves as the predominant form directing GARP recruitment in many cell types due to higher expression levels, while ARL5A possesses similar biochemical capabilities but may play more specialized roles or function redundantly as a backup system.
The interaction between ARL5B and the GARP complex represents a critical aspect of membrane trafficking regulation. Several experimental approaches can help elucidate the molecular details of this interaction:
Structural studies: X-ray crystallography or cryo-electron microscopy of ARL5B-GTP in complex with GARP components could reveal the precise binding interface and conformational changes involved in recognition.
Domain mapping: Generation of truncation or deletion mutants of both ARL5B and GARP subunits can identify the minimal regions required for interaction. This can be combined with pull-down assays or yeast two-hybrid screens to systematically map the binding domains.
Site-directed mutagenesis: Beyond the GTP/GDP-locked mutations (T30N and Q70L) , targeted mutations at potential interface residues can identify key amino acids mediating the interaction. Mutations that disrupt binding without affecting GTP loading would be particularly informative.
In vitro reconstitution: Purified components can be used to determine if the interaction is direct or requires additional factors. Techniques such as surface plasmon resonance or isothermal titration calorimetry can provide quantitative binding parameters.
Live-cell imaging: Fluorescently tagged ARL5B and GARP components can be used to visualize the dynamics of recruitment in real-time, potentially revealing the temporal sequence of complex assembly.
Cross-linking mass spectrometry: Chemical cross-linking followed by mass spectrometry analysis can identify residues in close proximity at the binding interface, providing constraints for molecular modeling of the complex.
The combination of these approaches would provide comprehensive insights into how ARL5B-GTP specifically recognizes and recruits the GARP complex to the trans-Golgi membrane.
ARL5B depletion leads to cellular phenotypes that extend beyond GARP complex mislocalization, providing insights into its broader functional roles:
Endosomal enlargement: In Drosophila tissues lacking Arl5, the late endosomal compartment becomes enlarged , suggesting accumulation of cargo that would normally be recycled to the Golgi. This can be quantified through morphometric analysis of labeled endosomes.
Phagosome maturation: In human macrophages, ARL5B depletion restores Lamp1 recruitment to phagosomes in HRV16-treated cells , indicating a role in regulating phagosome maturation. This can be assessed through co-localization analysis of phagosomal and lysosomal markers.
Bacterial clearance: The impact on phagosome maturation suggests potential effects on bacterial clearance capacity. Functional assays measuring the killing of phagocytosed bacteria could reveal whether ARL5B depletion enhances antimicrobial functions in infected cells.
Viral replication: In permissive cell types, ARL5B appears to act as a restriction factor for HRV16 , suggesting that its depletion might enhance viral replication. Viral titer assays or plaque formation tests could quantify this effect.
Golgi morphology: Since ARL5B localizes to the trans-Golgi and affects trafficking pathways, detailed ultrastructural analysis of Golgi morphology in ARL5B-depleted cells might reveal subtle alterations in Golgi architecture.
Secretory pathway function: Broader effects on the secretory pathway could be assessed through cargo trafficking assays, measuring the transport kinetics of model proteins that traverse the Golgi en route to the plasma membrane or extracellular space. Comprehensive phenotypic analysis using these approaches would provide a more complete picture of ARL5B's roles in cellular homeostasis.
ARL5B, like other members of the ARF family, is structurally similar to ADP-ribosylation factors. These proteins are involved in various cellular processes, including vesicular trafficking and actin cytoskeleton remodeling . ARL5B specifically binds and exchanges GTP and GDP, playing a crucial role in intracellular signaling pathways .
Human recombinant ARL5B is used in various research applications to study its function and role in cellular processes. Understanding the mechanisms of ARL5B can provide insights into its involvement in diseases and potential therapeutic targets.