RHOD regulates endosomal trafficking and alignment through interactions with Diaphanous-related formin (DRF) proteins and Src tyrosine kinase. It coordinates early endosome movement and internalization of tyrosine kinase receptors (e.g., PDGFRB) by modulating actin filament dynamics .
RHOD antagonizes RhoA-mediated stress fiber formation and promotes actin filament bundling via effector proteins:
WHAMM: Activates Arp2/3-dependent actin nucleation, driving filopodia formation and stress fiber dissolution .
FILIP1: Binds filamin A (FLNA), modulating cell adhesion and migration .
Overexpression of constitutively active RHOD induces thin, motile protrusions (cytoneme-like structures) .
RHOD knockdown increases focal adhesion size and cell attachment, impairing migration .
Recombinant RHOD is produced in E. coli for functional studies, enabling analysis of its GTPase activity and effector interactions .
RHOD expression levels correlate with cancer risk and prognosis:
Breast Cancer: RHOD may promote oncogenic signaling via cytoskeletal remodeling, enhancing tumor progression .
AML: RHOD overexpression correlates with aggressive subtypes (e.g., TP53-mutated cases) and reduced response to chemotherapy .
RHOD activation by fibroblast growth factor (FGF) induces cytoneme-like protrusions in mesenchymal cells:
Morphology: Thin, actin-rich extensions (20–30 μm) with microtubule colocalization .
Function: Mediate long-range signaling and nutrient uptake (e.g., transferrin internalization) .
GTPase | Key Functions | Downstream Effectors |
---|---|---|
RhoD | Endosome dynamics, actin bundling | WHAMM, FILIP1, Src |
RhoA | Stress fiber formation, cell contraction | ROCK, mDia1 |
Rac1 | Lamellipodia formation, cell migration | WASP, PAK |
Cdc42 | Filopodia formation, cell polarity | WASP, MRCK |
RHOD uniquely antagonizes RhoA activity, suppressing stress fibers while promoting actin bundles .
Therapeutic Targeting: Inhibiting RHOD in cancers where it promotes aggressive phenotypes (e.g., AML) .
Signaling Pathways: Elucidating cross-talk between RHOD and other Rho GTPases in disease contexts.
Structural Studies: Resolving RHOD-effector complexes (e.g., WHAMM or FILIP1) to guide drug design.
Rho-related GTP-binding protein RhoD, Rho-related protein HP1, RhoHP1, RHOD, ARHD, Rho, RHOM.
MGSSHHHHHH SSGLVPRGSH MVKVVLVGDG GCGKTSLLMV FADGAFPESY TPTVFERYMV NLQVKGKPVH LHIWDTAGQD DYDRLRPLFY PDASVLLLCF DVTSPNSFDN IFNRWYPEVN HFCKKVPIIV VGCKTDLRKD KSLVNKLRRN GLEPVTYHRG QEMARSVGAV AYLECSARLH
DNVHAVFQEA AEVALSSRGR NFWRRITQGF C.
RHOD (ras homolog gene family, member D) is a 210 amino acid protein with a molecular weight of approximately 23 kDa that belongs to the small GTPase superfamily, Rho family . It functions as a molecular switch that cycles between active GTP-bound and inactive GDP-bound states to regulate various cellular processes.
RHOD is primarily involved in:
Endosome dynamics and vesicular trafficking
Coordination of membrane transport with cytoskeletal function
Internalization and trafficking of activated tyrosine kinase receptors such as PDGFRB
Reorganization of actin cytoskeleton, particularly filopodia formation and actin filament bundling
Modulation of DAPK3's effect in reorganizing actin cytoskeleton and focal adhesion dissolution
To study RHOD's cellular functions, researchers typically employ combinations of:
Live-cell imaging with fluorescently tagged RHOD constructs
Co-immunoprecipitation to identify binding partners
siRNA knockdown or CRISPR/Cas9 knockout experiments
Overexpression of wild-type, constitutively active, or dominant-negative RHOD mutants
RHOD has distinct roles compared to better-studied Rho members like Cdc42, RhoA, and Rac1. While most Rho GTPases promote stress fiber formation and focal adhesion assembly, RHOD exhibits several unique characteristics:
RHOD uniquely binds to WHAMM (WASp homologue associated with actin Golgi membranes and microtubules) and FILIP1, which distinguishes its signaling pathway from other Rho GTPases . This interaction establishes a distinct regulatory mechanism where RHOD coordinates Arp2/3-dependent and filamin A-dependent processes to control actin dynamics .
To investigate structure-function relationships in RHOD research, consider these methodological approaches:
Site-directed mutagenesis: Create point mutations in conserved domains (e.g., GTP-binding pocket, effector binding regions) to analyze their impact on RHOD function. The recombinant human RHOD protein (aa 18-207) can serve as a reference for designing mutations .
Domain swapping experiments: Exchange domains between RHOD and other Rho GTPases to identify regions responsible for specific functions.
Structural biology techniques:
X-ray crystallography of RHOD alone or in complex with effectors
NMR spectroscopy for dynamic structural information
Cryo-EM for visualization of larger complexes
Biophysical interaction analysis:
Surface plasmon resonance (SPR) to measure binding kinetics
Isothermal titration calorimetry (ITC) for thermodynamic parameters
Fluorescence resonance energy transfer (FRET) to study protein-protein interactions in living cells
Advanced imaging:
Super-resolution microscopy to visualize RHOD localization with nanometer precision
Single-molecule tracking to follow individual RHOD molecules in living cells
RHOD forms a functional complex with WHAMM that plays a critical role in actin nucleation and cytoskeletal organization. This interaction represents a distinct regulatory pathway compared to other Rho GTPases .
The mechanism follows these steps:
RHOD binds directly to WHAMM, which acts as an effector protein
WHAMM then binds to and activates the Arp2/3 complex
The activated Arp2/3 complex promotes actin nucleation and branched filament formation
This process contributes to filopodia formation and actin filament bundling
WHAMM acts downstream of RHOD in regulating these cytoskeletal dynamics
Experimental evidence shows that cells with reduced levels of RHOD and WHAMM (through siRNA treatment) exhibit increased cell attachment and decreased cell migration, highlighting the physiological importance of this pathway .
Research approaches to study this interaction include:
Co-immunoprecipitation to verify complex formation
In vitro actin polymerization assays with purified components
Live-cell imaging with fluorescently tagged proteins
siRNA-mediated knockdown of either protein to establish functional relationships
Researchers investigating RHOD's impact on cytoskeletal dynamics can employ these methodological approaches:
Immunofluorescence microscopy:
Live-cell imaging:
Express fluorescently-tagged RHOD (e.g., GFP-RHOD) in cells
Use SiR-Actin for live F-actin visualization
Employ spinning disk confocal microscopy for rapid acquisition
Apply TIRF microscopy to focus on cytoskeletal events near the plasma membrane
Quantitative analysis of cytoskeletal parameters:
Measure filopodia number, length, and dynamics
Quantify actin filament organization using FilamentTracker or similar tools
Analyze focal adhesion turnover in relation to RHOD activity
Advanced biophysical approaches:
Atomic force microscopy to measure cell stiffness
Traction force microscopy to quantify cell-generated forces
Laser ablation to study tension in actin networks
Biochemical assays:
Actin co-sedimentation to measure F-actin binding
Pyrene-actin assays to measure polymerization kinetics
Pull-down assays using GST-RHOD to identify new binding partners
RHOD binds not only to WHAMM but also to FILIP1 (Filamin A-interacting protein 1), establishing a connection to filamin A (FLNa)-dependent mechanisms . This interaction represents an important pathway through which RHOD influences cytoskeletal architecture.
The mechanism involves:
RHOD binding to FILIP1
FILIP1 interacting with filamin A
Filamin A functioning as an actin-crosslinking protein that organizes actin filaments into orthogonal networks
This pathway influencing cell adhesion, spreading, and migration properties
Research evidence suggests that RHOD coordinates both Arp2/3-dependent mechanisms (via WHAMM) and FLNa-dependent processes (via FILIP1) to comprehensively control the actin filament system . This dual regulatory capability explains RHOD's distinctive impact on cell morphology and behavior.
To investigate this pathway, researchers can:
Perform co-immunoprecipitation studies with RHOD, FILIP1, and filamin A
Use proximity ligation assays to visualize protein interactions in situ
Conduct FRET analysis to measure binding dynamics in living cells
Employ filamin A-null cells to assess RHOD-dependent phenotypes
Analyze filamin A phosphorylation status in response to RHOD activation
RHOD localizes to early endosomes and recycling endosomes, indicating its important role in endosomal trafficking . Its function in this context appears to be coordinating membrane transport with cytoskeletal dynamics.
The mechanism includes:
RHOD association with endosomal membranes in its active GTP-bound state
Recruitment of effector proteins that connect endosomes to the actin cytoskeleton
Regulation of endosome motility and positioning within the cell
Facilitation of internalization and trafficking of activated tyrosine kinase receptors, particularly PDGFRB
To study RHOD's role in endosomal trafficking, researchers can:
Track fluorescently labeled cargo proteins (e.g., transferrin, EGF) in cells with modified RHOD expression
Use live-cell imaging with dual-color labeling of RHOD and endosomal markers
Employ super-resolution microscopy to resolve endosomal subdomains
Conduct pulse-chase experiments to follow receptor trafficking pathways
Analyze endosomal pH and maturation in RHOD-depleted cells
Distinguishing direct from indirect effects of RHOD on vesicular transport requires sophisticated experimental designs:
Acute inactivation/activation approaches:
Rapamycin-inducible dimerization to rapidly recruit or remove RHOD from membranes
Optogenetic control of RHOD activity with light-sensitive domains
Small molecule inhibitors specific to RHOD (though these are currently limited)
In vitro reconstitution assays:
Mutational analysis:
Create RHOD mutants that selectively disrupt binding to specific effectors
Generate endosomal targeting mutants that maintain cytoskeletal function
Use these tools to dissect which interactions are essential for vesicular transport
High-resolution temporal studies:
Perform time-resolved proteomics of RHOD-associated complexes
Conduct live-cell imaging with high temporal resolution
Correlate RHOD activation with vesicular movement events
Selective inhibition of downstream pathways:
Use specific inhibitors of actin dynamics, microtubules, or motor proteins
Determine if RHOD effects persist when these pathways are blocked
Apply computational modeling to predict direct versus feedback effects
CRISPR/Cas9 provides powerful tools for investigating RHOD function through precise genetic manipulation:
Knockout strategies:
Design gRNAs targeting early exons of the RHOD gene
Create complete knockout cell lines to observe loss-of-function phenotypes
Generate conditional knockouts using Cre-lox or similar systems for temporal control
Knockin approaches:
Create endogenously tagged RHOD (e.g., RHOD-GFP) to observe native expression levels
Introduce specific point mutations to study structure-function relationships
Generate reporter lines where fluorescent proteins are expressed under the RHOD promoter
Domain-specific editing:
Target specific functional domains (GTP-binding, effector interaction regions)
Create truncation mutants to identify minimal functional units
Perform precise base editing to introduce subtle modifications
Screening applications:
Conduct CRISPR screens targeting potential RHOD regulators or effectors
Use CRISPRi/CRISPRa to modulate RHOD expression without genetic alterations
Perform synthetic lethality screens in RHOD-depleted backgrounds
Optimization considerations:
Validate editing efficiency using sequencing and Western blotting
Confirm phenotypes with rescue experiments using wild-type RHOD
Create multiple independent clones to control for off-target effects
Discovering new RHOD binding partners requires specialized proteomics techniques:
Proximity-based approaches:
BioID or TurboID: Fuse RHOD to a biotin ligase to label proximal proteins
APEX: Use RHOD-APEX fusion to biotinylate nearby proteins upon H₂O₂ addition
These methods identify both stable and transient interactions in living cells
Traditional affinity purification:
Quantitative comparison strategies:
SILAC, TMT, or label-free quantification to compare:
Active vs. inactive RHOD mutants
Stimulated vs. basal conditions
Wild-type vs. RHOD knockout cells
Protein array screening:
Computational prediction and validation:
Use protein-protein interaction prediction algorithms
Validate top candidates with co-immunoprecipitation
Map interaction networks using systems biology approaches
Investigating RHOD in clinical contexts requires specialized approaches for patient samples:
Tissue analysis techniques:
Patient-derived cell models:
Primary cell cultures from patient samples
Patient-derived xenografts (PDX) in immunocompromised mice
Induced pluripotent stem cells (iPSCs) from patient fibroblasts
Differentiation into relevant cell types (e.g., cardiomyocytes if studying heart function)
Organoid applications:
Clinical correlation studies:
Measure RHOD expression/activity in patient cohorts
Correlate with clinical parameters (disease progression, treatment response)
Identify potential biomarker applications
Perform genetic association studies for RHOD variants
Ethical and practical considerations:
RHOD shows distinctive effects on cell migration compared to canonical Rho GTPases:
Unique migration phenotypes:
Mechanistic differences:
Context-dependent effects:
RHOD's impact may vary by cell type and microenvironment
The balance between RHOD and other Rho GTPases likely determines migration outcomes
RHOD's endosomal localization suggests it may regulate receptor recycling during migration
To study these distinctive effects, researchers can:
Perform wound healing assays comparing RHOD, RhoA, Rac1, and Cdc42 manipulations
Use microfluidic devices to measure directed migration in controlled gradients
Conduct single-cell tracking to quantify migration parameters
Employ traction force microscopy to measure mechanical forces during migration
To investigate RHOD in cancer invasion contexts, researchers can employ these approaches:
3D invasion assays:
Spheroid invasion into collagen or Matrigel matrices
Transwell invasion assays with different extracellular matrix components
Organotypic cultures mimicking tissue-specific microenvironments
Real-time imaging of invasion dynamics with fluorescently labeled cells
Molecular manipulation strategies:
Analysis of invasion mechanisms:
Assess invadopodia formation and matrix degradation
Quantify expression of matrix metalloproteinases
Evaluate changes in cell-cell and cell-matrix adhesions
Measure effects on epithelial-mesenchymal transition markers
In vivo metastasis models:
Intravital imaging of RHOD-manipulated cancer cells
Orthotopic implantation to study tissue-specific invasion
Circulating tumor cell analysis after RHOD modification
Lung colonization assays following tail vein injection
Clinical correlation:
Analyze RHOD expression in invasive tumor fronts
Compare RHOD levels between primary tumors and metastases
Correlate RHOD expression with patient outcomes
Investigate RHOD in therapy-resistant populations
Researchers have several sophisticated options for visualizing RHOD activity:
FRET-based biosensors:
Design intramolecular FRET sensors with RHOD flanked by fluorescent proteins
The sensor changes conformation upon GTP binding, altering FRET efficiency
Allows real-time visualization of RHOD activation in specific subcellular locations
Can be combined with other fluorescent markers for multiplexed imaging
Effector domain-based reporters:
Pull-down assays for biochemical quantification:
Use the RHOD-binding domain of effectors in GST-tagged form
Perform pull-downs at different timepoints after stimulation
Quantify active RHOD by Western blotting
Combine with phospho-specific antibodies to correlate with downstream signaling
Advanced microscopy approaches:
Fluorescence correlation spectroscopy (FCS) to measure RHOD diffusion
Fluorescence recovery after photobleaching (FRAP) to assess membrane association
Single-molecule tracking to follow individual RHOD molecules
Super-resolution microscopy to visualize RHOD nanoclusters
Proteomic readouts:
Proximity labeling at different activation states
Phosphoproteomic analysis of downstream signaling events
Targeted mass spectrometry to quantify RHOD PTMs
Correlation with functional readouts like cytoskeletal reorganization
Ras Homolog Gene Family Member D (RHOD) is a protein encoded by the RHOD gene in humans. It belongs to the Rho family of GTPases, which are part of the larger Ras superfamily of small GTP-binding proteins. These proteins play crucial roles in various cellular processes, including cytoskeletal dynamics, cell migration, and intracellular trafficking.
The RHOD gene is located on chromosome 11 and encodes a protein that is approximately 21 kDa in size. The RHOD protein contains a GTPase domain, which is responsible for binding and hydrolyzing GTP. This domain is highly conserved among members of the Rho family, indicating its importance in the protein’s function .
RHOD is involved in several cellular processes, primarily related to the actin cytoskeleton and membrane trafficking. It plays a significant role in endosome dynamics, which are essential for the internalization and trafficking of activated tyrosine kinase receptors such as the platelet-derived growth factor receptor beta (PDGFRB) .
One of the key functions of RHOD is to coordinate membrane transport with the function of the cytoskeleton. This coordination is crucial for maintaining cellular structure and facilitating various cellular activities, including cell division and migration .
RHOD interacts with several other proteins to carry out its functions. It has been shown to interact with WHAMM (WASP Homolog Associated with Actin, Golgi Membranes, and Microtubules), which is involved in the regulation of actin filament bundling and filopodia formation . Additionally, RHOD can modulate the effect of DAPK3 (Death-Associated Protein Kinase 3) in the reorganization of the actin cytoskeleton and focal adhesion dissolution .
Mutations or dysregulation of RHOD have been associated with various diseases. For instance, alterations in RHOD expression have been linked to Wiskott-Aldrich Syndrome and Anal Canal Squamous Cell Carcinoma . Understanding the role of RHOD in these diseases could provide insights into potential therapeutic targets.
Research on RHOD continues to uncover its diverse roles in cellular processes and its potential implications in disease. The recombinant form of RHOD is used in various experimental settings to study its function and interactions with other proteins. These studies are crucial for developing targeted therapies for diseases associated with RHOD dysregulation.