Vitronectin (VTN Human), encoded by the VTN gene on chromosome 17, is a multifunctional glycoprotein critical to cell adhesion, complement regulation, and tissue repair. Synthesized primarily in the liver, it circulates in blood plasma at concentrations of 150–300 µg/mL and is a key component of the extracellular matrix (ECM) and bone . VTN Human exists in two forms: a single-chain 75 kDa glycoprotein (478 amino acids) or a two-chain variant linked by disulfide bonds . Its structure includes three domains:
Somatomedin B domain (1–39): Stabilizes plasminogen activator inhibitor-1 (PAI-1) and binds urokinase receptor .
Central hemopexin domain (131–342) and C-terminal domain (347–459): Mediate interactions with integrins and ECM components .
Cell Adhesion & Migration: Binds integrins (αvβ3, αvβ5, αIIbβ3) via the RGD motif (residues 45–47), promoting cell-ECM interactions critical for wound healing and cancer metastasis .
Complement Regulation: Inhibits the terminal cytolytic complement pathway, protecting host cells from immune-mediated damage .
Hemostasis: Stabilizes PAI-1, regulating fibrinolysis and clot formation .
In ischemic stroke, VTN leakage into the brain exacerbates injury in females by inducing interleukin-6 (IL-6), leading to neuroinflammation and poor recovery . Plasma VTN levels correlate with infarct size in females but not males .
Recombinant VTN Human is widely used in biomedical research due to its defined composition and scalability.
Gibco’s CTS™ VTN-N supports long-term (>50 passages) feeder-free culture of human pluripotent stem cells (PSCs) with maintained pluripotency and differentiation capacity .
VTN promotes tumor malignancy by enhancing integrin-mediated metastasis. Anti-VTN aptamers inhibit cancer cell migration by blocking αvβ3/αvβ5 integrin signaling .
In female mice, VTN knockout reduces stroke-induced brain injury by 50% and lowers IL-6 levels, suggesting VTN as a sex-specific therapeutic target .
VTN enhances human PSC differentiation into hematopoietic lineages by activating αvβ3/αvβ5 integrin signaling, outperforming Matrigel in efficiency .
Vitronectin is a multifunctional glycoprotein with a length of 478 amino acids and molecular size of 54306Da. It exists in two main forms: a single-stranded 75-kDa form (V75) and a two-stranded form (65 kDa and 10 kDa). In humans, the VTN gene is located on chromosome 17Q11.2 and consists of eight exons and seven introns. VTN is primarily found in plasma and platelets, with distribution in the liver, spleen, and cardiac tissues. Its conformational variability significantly impacts its functional properties across different tissues .
VTN primarily interacts with integrin receptors, most notably αvβ3 and αvβ5 integrins, through its arginine-glycine-aspartic acid (RGD) sequence. This interaction triggers downstream signaling pathways that regulate various cellular functions including adhesion, migration, and differentiation. The binding specificity between VTN and its integrin receptors depends on the conformational state of VTN and the cellular context in which binding occurs. These interactions are crucial for VTN's role in neural development, stem cell differentiation, and various pathological processes .
VTN serves as a key component of the ECM, facilitating cell-matrix interactions through integrin binding. In the neural context, VTN's ECM interactions support axonal growth and orientation by providing adhesive substrates for growth cones. These interactions activate signaling pathways including PI3K/GSK3β in cerebellar granule cells and integrin-FAK signaling in astrocytes. The interaction between VTN and prion proteins within the ECM also supports axonal growth. When designing experiments to study these interactions, researchers should consider both two-dimensional and three-dimensional culture systems to better recapitulate the in vivo ECM environment .
VTN has multiple functions in neural differentiation. It promotes the differentiation of human embryonic stem cells (hESCs), oligodendrocytes, and mouse cerebellar granule cell precursors. In the developing nervous system, VTN is expressed in the cortex and spinal meninges. It plays a particularly important role in the ventral region of the neural tube, where it promotes motor neuron differentiation in synergy with N-SHH (N-terminal sonic hedgehog). From a methodological perspective, PVDF nanofiber scaffolds coated with VTN can be used to enhance neural differentiation of hESCs in vitro. When designing differentiation protocols, researchers should consider the timing of VTN introduction as it specifically regulates early differentiation stages .
VTN supports and guides neurite extension through multiple mechanisms. It promotes the growth of hippocampal neurites and enhances branching of mouse cortical neurons in vitro. The interaction between VTN and cellular prion proteins supports axonal growth through mechanisms that can be compensated by integrins. VTN also participates in neurogenesis through interactions with prion protein and stress inducer protein 1 (STI1). For experimental approaches, artificial nerve guidance conduits incorporating VTN have been developed to support and guide neurite extension. When studying these processes, researchers should consider using time-lapse microscopy to monitor growth cone dynamics in the presence of VTN .
In cerebellar granule cell precursors, VTN regulates axon specification through the β5 integrin/PI3K/GSK3β pathway during differentiation. VTN also induces phosphorylation of cyclic-AMP responsive element-binding protein (CREB), which regulates granule neuron differentiation. In astrocytes, VTN activates the integrin-FAK signaling pathway, enhancing the expression of ciliary neurotrophic factor (CNTF) to promote subventricular zone (SVZ) neurogenesis. When designing experiments to study these pathways, researchers should consider using specific inhibitors at different points in the signaling cascade to determine the precise mechanisms of VTN action .
VTN plays a detrimental role in ischemic stroke specifically in female mice. Following middle cerebral artery occlusion (MCAO), VTN enters the brain through the disrupted blood-brain barrier in both sexes equally, but VTN knockout females show smaller injuries compared to wild-type, while male outcomes remain unaffected by VTN status. Plasma VTN levels correlate with injury severity only in females. Mechanistically, VTN induces interleukin-6 (IL-6) in female brains after stroke, leading to increased inflammation, cell loss, and poorer neurological recovery. This sexual dimorphism appears independent of gonadal sex hormones, suggesting other mechanisms may be involved. For researchers studying these phenomena, it's essential to include both sexes in experimental designs and to consider VTN as a potential biomarker for stroke outcomes specifically in females .
To study VTN crossing the blood-brain barrier (BBB), researchers can employ multiple complementary approaches. In animal models, fluorescently-labeled or tagged VTN can be administered systemically followed by confocal microscopy to visualize its distribution in brain tissue. Co-localization studies with endothelial markers can help determine the precise route of entry. For quantitative assessment, western blotting or ELISA of brain tissue at various time points after stroke or injury can track VTN accumulation. In vitro BBB models using transwell systems with brain endothelial cells can measure VTN translocation under inflammatory or hypoxic conditions. When designing such experiments, researchers should include appropriate controls to distinguish between endogenous brain VTN and that derived from plasma, and consider the timing of sampling since VTN shows dynamic changes post-stroke, with levels peaking at 24 hours and returning to baseline by 7 days .
VTN is widely used for feeder-free culture of human pluripotent stem cells (hPSCs), providing a defined substrate that supports self-renewal. Different substrates like VTN or matrigel can induce distinct substates of hPSCs. While VTN effectively maintains pluripotency, it also influences differentiation potential. The choice between VTN and other substrates should be considered carefully depending on the experimental goals. For maintenance protocols, recombinant VTN is typically used at concentrations of 0.5-5 μg/cm², while differentiation protocols may require different concentrations or combinatorial approaches with other factors. Researchers should monitor pluripotency markers and differentiation potential regularly when using VTN-based culture systems .
When utilizing VTN for neural tissue engineering, several factors require careful optimization. The concentration and conformational state of VTN will affect cellular responses. PVDF nanofiber scaffolds coated with VTN have proven effective for neural differentiation of hESCs. The topographical features of scaffolds, combined with VTN's biochemical signals, synergistically influence neural differentiation and axonal guidance. For experiments involving 3D culture systems, VTN can be incorporated into hydrogels or electrospun nanofibers at concentrations typically ranging from 1-10 μg/mL. Researchers should systematically test multiple concentrations and perform time-course analyses to determine optimal conditions for their specific applications. Additionally, the mechanical properties of the substrate should be tuned to mimic those of native neural tissue for more physiologically relevant results .
The female-specific detrimental effects of VTN in stroke present unique opportunities for sex-specific therapeutic strategies. For translational studies, researchers should consider several approaches: (1) Develop VTN-targeting therapies specifically for female stroke patients, such as inhibitors of VTN-integrin binding or antibodies that neutralize leaked VTN in the brain; (2) Investigate the efficacy of inhibiting downstream IL-6 signaling as a female-specific neuroprotective strategy; (3) Evaluate plasma VTN levels as a biomarker for stroke risk and outcomes specifically in women; (4) Explore combination therapies that address both VTN-mediated and other sex-specific pathways simultaneously. When designing clinical trials or animal studies, accounting for estrous/menstrual cycle phases is crucial as hormonal status may modify VTN-related outcomes. The timing of intervention is also critical, as VTN levels peak 24 hours post-stroke and return to baseline by 7 days .
VTN exists in multiple conformational states that may differentially affect its functions in the nervous system. This presents several methodological challenges for researchers: (1) Developing conformation-specific antibodies or probes to distinguish between the single-stranded 75-kDa form and the two-stranded form (65 kDa and 10 kDa) in different neural contexts; (2) Engineering recombinant VTN variants that maintain specific conformational states to isolate their functional effects; (3) Employing advanced biophysical techniques such as circular dichroism spectroscopy or hydrogen-deuterium exchange mass spectrometry to monitor conformational changes in response to neural microenvironmental factors; (4) Using site-directed mutagenesis to determine which domains are critical for specific neural functions. Researchers should consider that conformational changes may occur dynamically in response to local factors in the neural environment, necessitating in situ analysis techniques rather than relying solely on in vitro approaches .
VTN interactions with glial cells, particularly astrocytes, present promising therapeutic targets. VTN activates integrin-FAK signaling in astrocytes, enhancing CNTF expression and promoting neurogenesis. Several research directions hold promise: (1) Developing small molecules or peptides that modulate specific VTN-integrin interactions on astrocytes to enhance their neurotrophic functions; (2) Engineering VTN variants with enhanced binding to specific integrin subtypes expressed by supportive glial populations while minimizing interaction with detrimental inflammatory cell types; (3) Creating conditional knockout models to selectively disrupt VTN-integrin signaling in specific glial subtypes to determine cell-specific functions; (4) Exploring the potential of astrocyte-targeted gene therapy to modify VTN-responsive pathways. When designing such studies, researchers should consider regional differences in glial response to VTN and the timing of intervention relative to injury or disease progression. Additionally, potential interactions between VTN-mediated signaling and other glial inflammatory pathways should be characterized to avoid unintended consequences of therapeutic interventions .
Advanced genomic and proteomic methodologies can significantly enhance our understanding of VTN in neural contexts. Single-cell RNA sequencing could identify cell-specific responses to VTN across neural cell types. Spatial transcriptomics could map VTN and receptor expression patterns across brain regions during development and in disease states. Chromatin immunoprecipitation sequencing (ChIP-seq) could identify transcription factors regulating VTN expression in neural tissues. Proteomic approaches such as proximity labeling (BioID or APEX) could identify novel VTN-interacting proteins in neural cells. Phosphoproteomics might uncover the complete signaling cascade activated by VTN-integrin binding in different neural cell types. When designing such studies, researchers should include appropriate controls for sex differences given the known sexual dimorphism in VTN function. Integration of multi-omics data will be essential to fully characterize the VTN interactome in neural contexts .
The influence of environmental factors on VTN biology remains largely unexplored but presents an important research frontier. Inflammation induced by environmental toxins, stress, or infection might alter hepatic VTN production or its plasma levels. Aging likely affects the conformational stability and function of VTN, potentially contributing to neurodegenerative processes. Diet and metabolic factors could influence VTN glycosylation patterns, affecting its binding properties and clearance. Researchers should consider designing longitudinal studies that track VTN levels in response to environmental exposures and incorporate metabolomic analyses to identify factors that modify VTN structure or function. Animal models exposed to various environmental conditions with subsequent analysis of VTN expression, conformation, and function in neural tissues could provide valuable insights. Additionally, epidemiological studies comparing VTN levels across populations with different environmental exposures might identify previously unrecognized risk factors for VTN-associated neurological conditions .
Vitronectin is a large glycoprotein found in blood and the extracellular matrix (ECM). It plays a crucial role in various physiological processes, including cell adhesion, migration, and tissue repair. The recombinant form of vitronectin, known as Vitronectin (Human Recombinant), is produced using advanced biotechnological methods to ensure high purity and functionality.
Vitronectin is composed of 459 amino acids and includes several functional domains:
These domains enable vitronectin to interact with a variety of cell surface receptors and ECM components, thereby influencing cell behavior and tissue organization .
Recombinant human vitronectin is typically produced in mammalian cell lines, such as Chinese Hamster Ovary (CHO) cells or human embryonic kidney (HEK) cells. The gene encoding vitronectin is inserted into these cells, which then express and secrete the protein. The recombinant protein is subsequently purified using techniques like affinity chromatography to achieve high purity levels .
Recombinant human vitronectin is widely used in biomedical research and clinical applications: