Vimentin Human (UniProt ID: P08670) is a 57 kDa type III intermediate filament (IF) protein encoded by the VIM gene. It serves as a structural scaffold in mesenchymal cells and plays critical roles in cellular integrity, organelle positioning, and stress response .
Mechanical resilience: Protects cells from mechanical stress in vivo; vimentin-knockout cells show fragility .
Organelle anchoring: Tethers nucleus, mitochondria, and endoplasmic reticulum .
Cholesterol transport: Facilitates LDL-derived cholesterol trafficking to esterification sites .
Immune modulation: Surface-expressed vimentin interacts with immune receptors (e.g., NKp46) to regulate inflammation .
NSCLC Metastasis: KPV−/− mice (vimentin-deficient) exhibited 80% reduced lung tumor burden and failed to metastasize in allograft models .
EMT Regulation: Vimentin expression in MCF7 cells increases migratory capacity by 2.5-fold and upregulates Slug/ERK signaling .
Sepsis Biomarker: Serum vimentin levels in septic patients were 3.8-fold higher than controls (p < 0.001) .
Macrophage Activity: Phosphorylated vimentin secreted by macrophages enhances oxidative burst against pathogens .
Parameter | Vimentin-Positive Cells | Vimentin-Null Cells |
---|---|---|
Mechanical Stiffness | 1.8 kPa (elastic modulus) | 0.9 kPa (elastic modulus) |
Wound Healing Rate | 95% closure at 24 hrs | 40% closure at 24 hrs |
Colon Cancer: Methylation of the VIM promoter is a fecal DNA biomarker (sensitivity: 72%, specificity: 86%) .
Rheumatoid Arthritis: Anti-citrullinated vimentin antibodies are diagnostic markers .
Vimentin Inhibitors: Withaferin A and simvastatin disrupt filament networks, reducing metastasis in preclinical models .
Gene Therapy: siRNA-mediated vimentin knockdown increases cancer cell apoptosis by 40% .
Human vimentin is a type III intermediate filament protein with a characteristic tripartite domain structure consisting of a central α-helical rod domain flanked by non-α-helical head and tail domains. The rod domain has been extensively characterized through crystallography and EPR spectroscopy, revealing a coiled-coil structure in segments 1B and 2, while segment 1A exhibits a single alpha helix rather than the expected coiled-coil structure .
The molecular structure of vimentin allows remarkable structural plasticity, enabling the protein to respond dynamically to mechanical stress. Using a "Divide and Conquer" approach, researchers have determined the crystal structure of several peptides derived from human vimentin . Recent advancements combine experimental EPR data with molecular modeling to complete the vimentin rod domain structure, particularly the previously unresolved L1-2 linker region (residues 248-263) .
Methodology for structural analysis typically involves:
X-ray crystallography of vimentin peptides
Site-directed spin labeling (SDSL) coupled with electron paramagnetic resonance (EPR) spectroscopy
Molecular dynamics (MD) simulations to validate models and understand dynamic behavior
Construction of full-length models using molecular modeling software like MODELLER and UCSF CHIMERA
Vimentin expression is remarkably widespread in human tissues. According to the Human Protein Atlas database, vimentin protein is expressed in the majority of the 44 tissues analyzed, with 14 tissues showing particularly high expression levels, including skin, lung, kidney, bone marrow, and lymph node . At the RNA level, vimentin is constitutively expressed across all major tissues as documented in the HPA, GTEx, and Fantom databases .
The regulation of vimentin expression involves multiple mechanisms:
A serum response element in the VIM promoter responds to factors present in serum, explaining why many cell types express vimentin when cultured in serum-supplemented media
During embryonic development, vimentin first appears in highly migratory cells when the embryo is still a two-layered epithelium and ectodermal cells begin migrating into the "mesodermal cleft"
Postnatally, vimentin expression becomes restricted to fibroblasts, endothelial cells, lymphocytes, and specialized cells in the thymus and brain
Upregulation occurs during epithelial-mesenchymal transition (EMT) in both cultured epithelial cells and in vivo contexts
Researchers should note that many cell types expressing vimentin in culture may not express it in vivo due to the serum effect, making them potentially problematic models for studying the genuine biological functions of vimentin .
Vimentin plays critical roles in multiple cellular processes:
Cellular mechanics and structure:
Maintains mechanical stability of cells
Integrates mechanical stimuli from the environment
Forms load-bearing superstructures that restrain F-actin retrograde flow
Regulates cell cortical stiffness, which affects migration capacity
Cell migration and motility:
Coordinates with focal adhesions and the actomyosin network to regulate cell motility
Mediates bidirectional interactions with the actomyosin network that control cell migration
Affects RhoA kinase signaling
Modulates the dynamics of both microtubule and actomyosin networks
Additional cellular functions:
Cell adhesion and wound healing
Mechanosensing and mechanotransduction
Cell senescence
Transport of organelles and mRNA
Regulation of protein synthesis
Vimentin's role in cell migration involves complex molecular interactions with multiple cytoskeletal and signaling components:
Interaction with focal adhesion complexes:
Vimentin regulates focal adhesion kinase (FAK), which imparts motile properties to cells
By recruiting VAV2 (a Rac1 guanine nucleotide exchange factor) to FAK, vimentin regulates cell adhesion
Vim−/− cells exhibit elevated levels of phosphorylated FAK and its targets, Src and ERK1/2
Coordination with actin dynamics:
Vimentin intermediate filaments slow actin retrograde flow rates, which buffers traction stresses
In Vim−/− cells, actin flows are more than an order of magnitude faster
Vimentin restrains F-actin's retrograde flow and governs the alignment of traction stresses
Influence on lamellipodium width, a key structure for pseudopodial migration
Mechanical effects:
Cells lacking vimentin show significant decreases in cortical region stiffness
This mechanical deficit contributes to impaired migratory activity and insufficient force generation for wound contraction
Live-cell observations demonstrate that shear stress causes rapid deformation of vimentin networks, suggesting a role in mechanosensing and mechanotransduction
For studying these interactions, researchers employ:
Traction force microscopy
Sharp tip atomic force microscopy
Time-lapse imaging of fluorescently tagged vimentin
Comparative studies between wild-type and Vim−/− cells
Vimentin knockout mice (Vim−/−) exhibit numerous phenotypes across different systems despite initially appearing to develop and reproduce normally. Key phenotypes include:
Vascular system abnormalities:
Poor vascular development with fewer vessels and reduced branching
Defects in arterial remodeling and vascular smooth muscle cell differentiation
Elevated carotid stiffness, contractility, and endothelial dysfunction
Failed vasodilation of the renal vascular system after partial nephrectomy, leading to renal failure
Wound healing and tissue repair defects:
Impaired wound healing and contraction
Defects in TGF-β signaling
Impaired epithelial-to-mesenchymal transition
Nervous system alterations:
Hypermyelination of peripheral nerves
Slower neurite outgrowth
Metabolic and cellular abnormalities:
Defects in lipid droplet organization
Abnormalities in cell size regulation and mTORC1 signaling
Decreased production of corticosterone affecting energy, immune reactions, and stress responses
Sex-specific effects:
Increased mortality in male Vim−/− mice, but not in females
Possibly related to higher susceptibility to environmental stress due to decreased corticosterone production
These diverse phenotypes highlight vimentin's critical roles across multiple physiological systems and provide valuable models for studying vimentin-related diseases.
Vimentin plays multifaceted roles in cancer progression:
EMT and cancer cell plasticity:
Vimentin is a key marker for epithelial-mesenchymal transition (EMT), a critical process in tumor cell dissemination
Experimental co-expression of vimentin and keratin in MCF-7 human breast cancer cells (MoVi clones) leads to phenotypic interconversion between epithelial and mesenchymal states
Enhanced invasion and motility:
Overexpression of vimentin in MCF-7 cells leads to augmented motility and invasiveness in vitro
These activities can be transiently down-regulated by vimentin antisense oligonucleotides
Proliferation and tumorigenicity:
Integrin profile alterations:
Vimentin expression causes a decrease in α2- and α3-containing promiscuous integrins and β1-containing integrins
Concurrent increase in α6-containing laminin receptor integrin
Cell surface vimentin:
Vimentin can be expressed at the cell surface in addition to its cytoplasmic location
Cell surface vimentin serves as a natural human immune response target
Monoclonal antibodies reactive with cell surface vimentin have been obtained from sentinel lymph nodes of cancer patients
Pritumumab, one such vimentin-reactive antibody, has been used to treat brain cancer patients
These findings suggest vimentin confers selective advantages to cancer cells in their interpretation of extracellular matrix signals, though vimentin alone may not be sufficient for metastasis.
Vimentin is implicated in several inflammatory and autoimmune conditions:
Rheumatoid Arthritis (RA):
Biopsies from RA-affected tissues express fibrotic markers including vimentin, while healthy tissues express epithelial-like biomarkers
Approximately 40% of sera from RA patients contain autoantibodies directed toward mutated citrullinated vimentin (MCV)
Anti-MCV antibodies can be detected early in disease progression
Anti-MCV titers correlate closely with disease progression, allowing for early diagnosis, prognosis, and therapeutic monitoring
Citrullination of vimentin during inflammation triggers antigenic properties
Crohn's Disease:
Associated with upregulation of vimentin protein levels
Invasive properties of Crohn's disease cells are linked to vimentin expression
Fibrotic areas show EMT-related markers, particularly vimentin, suggesting EMT involvement in pathogenesis
Vimentin-targeted treatment of Crohn's-disease-associated Escherichia coli with withaferin-A promotes correct functioning of inflammatory response, autophagy, and cell invasion
Other Inflammatory Conditions:
Vimentin serves as an important marker in many inflammatory conditions
It plays roles in delayed wound healing
Involved in cell surface binding and replication of viruses including HIV, SARS-CoV, dengue, and encephalitis viruses
In HIV specifically, vimentin is associated with viral infectivity factor linked to HIV replication
The connection between vimentin and these conditions highlights its potential as both a diagnostic biomarker and therapeutic target.
Researchers employ multiple techniques to assess vimentin:
Immunohistochemistry (IHC):
Standard approach for tissue sections
Can detect spatial distribution within tissues and cells
Allows scoring of expression intensity and percentage of positive cells
Permits co-localization studies with other markers
Immunofluorescence microscopy:
Confocal immunofluorescence microscopy provides superior resolution
Enables visualization of vimentin filament networks
Particularly useful for studying dynamic changes in live cells
Can be combined with computer-assisted imaging to quantify stained areas
Molecular techniques:
Real-time quantitative RT-PCR (RT-qPCR) for mRNA expression
Western blot for protein level detection and semi-quantification
Flow cytometry for quantifying vimentin-positive cell populations
Advanced structural approaches:
Site-directed spin labeling (SDSL) with electron paramagnetic resonance (EPR) spectroscopy
X-ray crystallography of vimentin peptides
Live-cell imaging:
Expression of fluorescently tagged vimentin
Allows for real-time observation of network dynamics under various stimuli
Particularly valuable for mechanosensing studies where rapid deformation can be visualized
When selecting a methodology, researchers should consider:
The specific research question (expression level vs. spatial distribution vs. dynamics)
Available tissue/sample type (fixed vs. fresh vs. cell culture)
Need for quantitative vs. qualitative data
Whether subcellular localization information is required
Several approaches are available for modulating vimentin expression or function:
Genetic manipulation:
Knockout models: Vim−/− mice are widely used to study systemic effects of vimentin loss
siRNA/shRNA for transient or stable knockdown in cell culture models
CRISPR-Cas9 genome editing for targeted modifications of the vimentin gene
Transfection with vimentin expression vectors (as in MoVi clones) to study overexpression effects
Pharmacological interventions:
Withaferin-A: A plant-derived inhibitor that binds to vimentin and disrupts its function
Has been used in Crohn's disease models to target vimentin-mediated processes
Other compounds that target vimentin assembly or stability
Post-translational modification targeting:
Modulating citrullination of vimentin (relevant for autoimmune conditions)
Targeting phosphorylation pathways that affect vimentin organization
Antibody-based approaches:
Vimentin antisense oligonucleotides can transiently down-regulate vimentin-mediated activities
Monoclonal antibodies against cell surface vimentin (like pritumumab) can be used for targeting vimentin-expressing cells
Function-blocking antibodies can interfere with specific vimentin interactions
Experimental considerations:
Cell type selection is crucial as many cultured cells express vimentin due to serum response elements in the VIM promoter
Phenotypic changes may be context-dependent and influenced by compensatory mechanisms
Temporal aspects are important as acute vs. chronic vimentin modulation may yield different results
Investigating vimentin's interactions with other cytoskeletal elements requires specialized techniques:
Colocalization studies:
Dual or multi-color immunofluorescence microscopy
Super-resolution microscopy techniques (STORM, PALM, SIM) for detailed spatial relationships
Live-cell imaging with differently labeled cytoskeletal components
Biochemical interaction assays:
Co-immunoprecipitation to identify protein-protein interactions
Proximity ligation assays to detect close associations in situ
Pull-down assays with purified components to test direct interactions
Functional perturbation approaches:
Selective disruption of specific cytoskeletal elements (e.g., using cytochalasin D for actin, nocodazole for microtubules)
Comparing effects in wild-type vs. Vim−/− cells
Live imaging during cytoskeletal perturbations
Mechanical studies:
Traction force microscopy to measure cell-generated forces
Atomic force microscopy to assess local mechanical properties
Optical tweezers or magnetic tweezers for direct mechanical manipulation
Computational and modeling approaches:
Agent-based modeling of cytoskeletal interactions
Molecular dynamics simulations to predict interaction interfaces
Analysis of simultaneous dynamics of different cytoskeletal elements
Studies have revealed that vimentin interacts with both the actin and microtubule networks. For instance, research has shown that vimentin IFs slow actin retrograde flow rates, with actin flows being more than an order of magnitude faster in Vim−/− cells . These interactions are crucial for cell migration, as vimentin forms a load-bearing superstructure that restrains F-actin's retrograde flow and governs the alignment of traction stresses.
Cell surface vimentin represents an emerging area of vimentin biology with significant therapeutic implications:
Expression and detection:
Vimentin, typically considered a cytoplasmic protein, can also be expressed at the cell surface
Several studies have confirmed this localization using surface biotinylation and non-permeabilized immunostaining approaches
Expression has been detected on various cell types including cancer cells, activated macrophages, and platelets
Natural immune responses:
Natural human monoclonal antibodies reactive with cell surface vimentin have been isolated from sentinel lymph nodes of cancer patients
This suggests an innate auto-antigenic natural human immune response, potentially a function of immunosurveillance
The recognition of cell surface vimentin may be part of a pre-established immune response to vimentin released during cell lysis
Therapeutic applications:
Pritumumab, a vimentin-reactive antibody derived from lymph nodes of cancer patients, has been used to treat brain cancer patients
The natural human origin of these antibodies may confer advantages for therapeutic applications
Cell surface vimentin serves as an accessibility target compared to intracellular vimentin
Research questions under investigation:
How does vimentin—and in what form—enter lymph nodes to stimulate immune responses?
Is surface vimentin by itself, in an altered form, or complexed with other biomolecules?
What role do post-translational modifications play in focusing a human response on particular vimentin epitopes?
How is cell surface vimentin involved in epithelial-mesenchymal transition processes?
The study of cell surface vimentin opens new avenues for therapeutic approaches in cancer and potentially other vimentin-associated diseases.
Post-translational modifications (PTMs) of vimentin critically influence its functions and disease associations:
Citrullination:
Conversion of arginine residues to citrulline by peptidylarginine deiminases
Citrullinated vimentin (MCV) becomes an autoantigen in rheumatoid arthritis
Approximately 40% of RA patient sera contain anti-MCV antibodies
These antibodies serve as valuable diagnostic and prognostic markers
Citrullination during inflammation triggers antigenic properties within the filament
Phosphorylation:
Regulates vimentin assembly/disassembly dynamics
Multiple kinases target vimentin, including PKA, PKC, CDKs, and Rho kinase
Phosphorylation states change during cell cycle progression, cellular stress, and migration
Hyperphosphorylation can lead to filament reorganization or disassembly
Other modifications:
Sumoylation: Affects vimentin solubility and assembly
Glycosylation: May influence stability and interactions
Oxidative modifications: Occur during cellular stress and aging
Methodological approaches to study PTMs:
Mass spectrometry for comprehensive PTM mapping
Phospho-specific or citrulline-specific antibodies
Site-directed mutagenesis to create phosphomimetic or non-phosphorylatable variants
Enzyme inhibitors to modulate specific modifications
Understanding vimentin PTMs provides insights into both normal regulatory mechanisms and pathological processes, particularly in contexts like autoimmunity where modified forms of vimentin become immunogenic.
Vimentin represents a promising therapeutic target across several disease contexts:
Cancer therapy approaches:
Monoclonal antibodies: Pritumumab and other antibodies targeting cell surface vimentin
Small molecule inhibitors: Compounds disrupting vimentin assembly or stability
Vimentin-targeted drug delivery: Exploiting cell surface vimentin for targeted delivery
Gene therapy: Antisense oligonucleotides have shown efficacy in down-regulating vimentin-mediated activities in experimental models
Autoimmune disease strategies:
Targeting citrullinated vimentin in rheumatoid arthritis
Modulating immune responses to vimentin epitopes
Blocking vimentin-mediated signaling in inflammatory contexts
Infectious disease interventions:
Targeting vimentin's role as a receptor or co-receptor for viruses (HIV, SARS-CoV, dengue, encephalitis)
Vimentin-targeted treatment of Crohn's-disease-associated bacteria with withaferin-A
Fibrosis and wound healing approaches:
Modulating vimentin's role in EMT to control fibrotic processes
Targeting vimentin-dependent TGF-β signaling
Enhancing appropriate wound healing responses through vimentin-mediated pathways
Methodological considerations:
Specificity challenges: Vimentin's widespread expression necessitates careful targeting
Delivery strategies: Accessing intracellular vimentin vs. targeting cell surface vimentin
Combination approaches: Vimentin-targeted therapies as part of multi-modal treatment strategies
The remarkable functional diversity of vimentin stems from several factors:
Structural adaptability:
Vimentin intermediate filaments possess inherent structural plasticity
The protein can form different assembly states (soluble, unit-length filaments, mature filaments)
Dynamic reorganization occurs in response to cellular needs and stresses
Molecular dynamics simulations show that the L1-2 linker region exhibits heterogeneity with concerted switching of states among dimer chains
Context-dependent interactions:
Forms different protein complexes depending on cell type and physiological state
Interacts with multiple cytoskeletal elements, including actin and microtubules
Serves as a scaffold for signaling molecules in various pathways
Regulatory diversity:
Subject to numerous post-translational modifications that alter function
Expression levels vary significantly between tissues and developmental stages
Different splice variants may predominate in different contexts
Sex-specific effects suggest hormonal regulation (as seen in male vs. female Vim−/− mice)
Methodological considerations for researchers:
Cell type selection is critical for relevance to in vivo function
Awareness that serum in culture media induces vimentin expression in many cell types
Need for complementary in vitro, cellular, and in vivo approaches
Integration of structural, mechanical, and signaling perspectives
The apparent contradictions in vimentin function likely reflect its role as a cellular integrator—responding to and coordinating diverse inputs to maintain cellular homeostasis across varying conditions and requirements.
Vimentin is a class III intermediate filament protein predominantly found in cells of mesenchymal origin, such as vascular endothelium and blood cells . It plays a crucial role in maintaining cell integrity, providing resistance against stress, and supporting cellular structure . The human Vimentin protein is encoded by the VIM gene and consists of 466 amino acids . Recombinant human Vimentin is produced using advanced biotechnological methods to ensure high purity and functionality .
Recombinant human Vimentin is typically produced in Escherichia coli (E. coli) expression systems . The gene encoding human Vimentin is cloned into an expression vector, which is then introduced into E. coli cells. These cells are cultured under optimal conditions to express the Vimentin protein. After expression, the protein is purified using affinity chromatography techniques, often involving a His-tag for easy purification . The purified protein is then lyophilized and stored under specific conditions to maintain its stability and activity .
Vimentin is known to interact with various cellular components and participate in multiple biochemical processes. One notable interaction is with G-quadruplex (G4) structures, which are tetra-helical arrangements formed within guanine-rich tracts of DNA . Vimentin binds to these G4 structures with high affinity, potentially influencing gene expression and chromatin organization . Additionally, Vimentin plays a role in the epithelial-to-mesenchymal transition (EMT), a process critical for cancer metastasis and drug resistance . It interacts with various signaling molecules and structural proteins, contributing to cellular migration, division, and structural integrity .