In this review, Ridge et al. discuss the essential functions of vimentin IFs revealed from studies of Vim−/− mice and cells derived from them.
Keywords: intermediate filaments, vimentin, vimentin-null mouse
Abstract
More than 27 yr ago, the vimentin knockout (Vim−/−) mouse was reported to develop and reproduce without an obvious phenotype, implying that this major cytoskeletal protein was nonessential. Subsequently, comprehensive and careful analyses have revealed numerous phenotypes in Vim−/− mice and their organs, tissues, and cells, frequently reflecting altered responses in the recovery of tissues following various insults or injuries. These findings have been supported by cell-based experiments demonstrating that vimentin intermediate filaments (IFs) play a critical role in regulating cell mechanics and are required to coordinate mechanosensing, transduction, signaling pathways, motility, and inflammatory responses. This review highlights the essential functions of vimentin IFs revealed from studies of Vim−/− mice and cells derived from them.
Of the three cytoskeletal systems, intermediate filament (IF) proteins are encoded by the largest gene family. Comprised of >70 members, IFs show remarkable diversity in their sequences, expression patterns, and distribution in various tissues (Herrmann and Aebi 2016). Vimentin, a type III IF protein, is one of the most well-known and extensively studied members of the IF protein family, reflecting their assembly into major cytoskeletal systems in cells of mesenchymal and ectodermal origin. Vimentin IFs were originally described as 10-nm-diameter filaments in several types of cultured cells, including embryonic chicken cells, HeLa cells, and hamster and mouse embryonic fibroblasts (Taylor 1966; Goldman and Follett 1969; Ishikawa et al. 1969; Goldman 1971). Some early studies in various cell types proposed that IFs were a smaller form of microtubules or “minimicrotubules,” slowing progress in the field for some time (Taylor 1966; Wisniewski et al. 1968). However, by the mid-1970s, fully polymerized vimentin IFs were isolated from cell-free preparations devoid of microtubules and actin filaments (Starger and Goldman 1977). These preparations could be depolymerized and repolymerized in vitro, demonstrating that their biochemical properties and structure were distinctly different from the other cytoskeletal systems (Zackroff and Goldman 1979). In 1983, the hamster vimentin gene was cloned and sequenced, further defining its unique structural properties (Quax et al. 1983). Subsequently, it was shown that vimentin is highly conserved, as it displays extensive sequence homology among species ranging from fish to humans (Herrmann and Aebi 2004; Hyder et al. 2008), suggesting essential evolutionarily conserved functions. However, vimentin's functional significance was seriously challenged when it was reported that genetically modified mice lacking vimentin (Vim−/− mice) develop and reproduce normally without an obvious phenotype (Colucci-Guyon et al. 1994). This outcome has been frequently taken as evidence that vimentin is of little physiological importance. However, soon after this initial report, investigators discovered a remarkable array of disease-related phenotypes linked to the Vim−/− mice (Tables 1, 2). This review intentionally focuses on the results obtained with this mouse model to provide an overview of the impressive diversity of phenotypes attributable to vimentin at the cell, tissue, and organ levels. In addition, it relates these phenotypes to alterations in cellular and molecular mechanisms using cells derived from Vim−/− mice (Fig. 1). Collectively, the information derived from studies involving Vim−/− mice has had a significant impact on the entire IF research field, has opened up new opportunities to study the structure and function of vimentin IFs, and has influenced the understanding of the pathogenesis of several diseases.
Table 1.
A summary of the phenotypes and functions revealed by vimentin-null (Vim−/−) mice
Table 2.
The two-edged sword of vimentin in the nervous system
Figure 1.

Overview of affected organs in mice lacking vimentin. The figure shows the various organs affected by vimentin gene ablation. Some phenotypes are apparent at steady state, but many phenotypes have been discovered when Vim−/− mice are subjected to stress, injury, or the modeling of different diseases.
Vimentin is required for normal wound healing
One of the most profound and major defects analyzed in Vim−/− mice is the inability to heal wounds properly (Fig. 2). In this regard, Vim−/− embryos fail to heal an excision wound within the time frame it takes for wild-type embryos to heal fully. Similar excision wound studies using adult Vim−/− mice also showed delays in the migration of fibroblasts into the wound site and subsequent wound contraction (Eckes et al. 2000). The latter is attributable to the delayed appearance of myofibroblasts. A more recent study showed that the Vim−/− mice display a general wound healing deficiency, occurring irrespectively of whether the wound is caused by excision, incision, or burning (Cheng et al. 2016). These deficiencies include slow scab formation, defective fibroblast functions, impaired inflammatory and immune responses, and faulty angiogenesis (Fig. 2).
Figure 2.
Severely compromised wound healing in mice lacking vimentin. (A) Perturbed wound healing has been one of the more significant phenotypes ascribed to Vim−/− mice. The figure describes normal wound healing (top panel) and the wound healing defects in Vim−/− mice (bottom panel). It has been demonstrated that the inhibited wound healing in Vim−/− mice is due to lost coordination and processing of cellular activities that are key for normal wound healing: fibroblast proliferation, collagen deposition, modeling keratinocyte transdifferentiation, re-epithelialization, angiogenesis, and vascularization. The figure demonstrates that loss of vimentin disrupts all of these functions, leading to slow, poor, and incomplete wound healing. The phenotype is similar for incision, excision, and burn wounds. As these wound models are very different in their character, the observations demonstrate that vimentin has a general role in determining the progression of epidermal regeneration regardless of the type of epidermal injury. The figure also illustrates a loss of fat accumulation in the Vim−/− mice, a constitutive defect in these mice that may have several ramifications for the affected tissues. Active research on Vim−/− mice has recapitulated many of the cellular signaling and other mechanisms underlying the observed defects. These are described in detail in different parts of this review. (B) A close-up of the proliferation stage depicts all the different cells and processes that vimentin is facilitating.
Further insights into the migration defects in wound healing detected in the Vim−/− mice come from comparative studies of mouse embryonic fibroblasts (MEFs) derived from WT and Vim−/− mice. Compared with WT MEFs, the Vim−/− MEFs display reduced mechanical stability, motility, and directional migration toward chemoattractive stimuli (Eckes et al. 1998). From a mechanistic viewpoint, studies of Vim−/− MEFs revealed that the bidirectional interactions between vimentin IFs and the actomyosin network mediate cell motility (Jiu et al. 2015). Specifically, these interactions restrict the retrograde flow of actin and consequently control nuclear positioning during cell migration. This vimentin IF–actomyosin interaction also affects RhoA kinase signaling (Jiu et al. 2017). Vimentin IFs have also been shown to integrate mechanical stimuli from the environment and modulate the dynamics of both the microtubule and actomyosin networks (Gan et al. 2016; Costigliola et al. 2017). During single-cell migration, polymerized elongated vimentin IFs slow the actin retrograde flow rates, which buffer traction stresses. In contrast, actin flows are more than an order of magnitude faster in Vim−/− cells (Costigliola et al. 2017). These findings indicate that vimentin IFs are a load-bearing superstructure that restrains F-actin's retrograde flow and governs the alignment of traction stresses regulating cell migration. Using traction force microscopy and sharp tip atomic force microscopy, it has been shown that Vim−/− MEFs exhibit significant decreases in the stiffness of their cortical regions (Vahabikashi et al. 2019). These findings also help to explain why the migratory activity of Vim−/− MEFs is impaired and the wound fails to generate sufficient force for wound contraction. Apart from defects in fibroblast functions, the wound healing deficiencies in Vim−/− mice have also been related to defects in TGF-β signaling, epithelial-to-mesenchymal transition (EMT) (Cheng et al. 2016; Cheng and Eriksson 2017), and vascularization (Nieminen et al. 2006; Antfolk et al. 2017) as described below.
Vimentin is a key regulator of fibrosis
Given the role of vimentin IFs in wound healing, several laboratories have used Vim−/− mice in studies exploring the process of fibrosis, which is a consequence of dysregulated tissue repair involving the excessive deposition of extracellular matrix (ECM) components by activated fibroblasts. Specifically, WT mice, but not Vim−/− mice, show increased collagen deposition following various pathophysiological insults known to cause fibrosis in WT animals. In general terms, fibrosis is associated with the loss of parenchymal cells and the disruption of tissue architecture, organ dysfunction, and, ultimately, organ failure. For example, in the lungs, fibrosis can be induced by exposure to bleomycin or asbestos. Interestingly, Vim−/− mice are protected from lung fibrosis in part due to a reduced inflammatory response, specifically a decrease in transforming growth factor β (TGF-β) (dos Santos et al. 2015). TGF-β1 is a major profibrogenic cytokine secreted by human and mouse alveolar epithelial cells in response to severe bleomycin- or asbestos-induced injury (Kim et al. 2006; Rogel et al. 2011). The TGF-β1–Smad pathway up-regulates vimentin expression as mouse alveolar epithelial cells undergo the EMT (Rogel et al. 2011). These results indicate that vimentin is required for the mesenchymal cell expansion associated with fibrosis. In complementary studies, human alveolar epithelial cells isolated from patients with idiopathic pulmonary fibrosis (IPF) show increased expression of vimentin, N-cadherin, and α-smooth muscle actin (SMA), all markers of the conversion to mesenchymal cells accompanying the EMT. Inhibition of TGF-β receptor kinase inhibits vimentin expression, causing the alveolar cells to revert to an epithelial phenotype (Kim et al. 2006; Marmai et al. 2011; Reyfman and Gottardi 2019).
The accumulation of fibroblasts leads to excessive collagen deposition and matrix remodeling, which distort the tissue architecture and contribute to the progressive decline in organ function. Notably, Vim−/− mice suppress collagen deposition and thereby maintain lung compliance (e.g., the mice show little change in lung stiffness) and function (dos Santos et al. 2015), protecting the host from lung fibrosis. Mechanistically, Vim−/− mice are protected in part because vimentin IFs stabilize collagen mRNAs, which are needed to promote collagen synthesis and collagen deposition (Challa and Stefanovic 2011). Vim−/− mice also fail to develop the renal fibrosis exhibited by WT mice following unilateral ureteral obstruction (UUO) (Wang et al. 2018). Similar to lungs, interstitial collagen deposition also decreases in Vim−/− mice following UUO.
Traditionally, aging and changes in inflammatory responses were considered key factors influencing whether wound healing leads to progressive fibrosis or ends with healthy wound repair (see “Vimentin Is Required for Normal Wound Healing” above). Studies using the Vim−/− mice reveal that vimentin can trigger maladaptive fibrotic responses that lead to dysregulated tissue repair and tissue destruction. The development of pharmacologic agents that selectively target vimentin to disrupt EMT, cell migration, and the organization and deposition of extracellular matrix proteins may help tip the balance between healthy wound repair and fibrosis.
Role of vimentin in the metastatic cascade
Vimentin is highly expressed in primary and metastatic tumors derived from epithelial tissues, which normally express keratin intermediate filaments. Vimentin expression in tumor tissue correlates with increased metastatic potential (Kidd et al. 2014; Liu et al. 2016) and poor overall survival in lung, prostate, and breast cancers (Domagala et al. 1990; Burch et al. 2013; Dauphin et al. 2013). Despite the abundance of clinical evidence supporting a link between vimentin expression and the metastatic process, a causal, in vivo link between vimentin and disease progression was not established until genetically engineered mouse cancer models using Vim−/− mice were established. For example, the LSL-KrasG12D/TP53fl/fl/Vim−/− mouse model has been used to demonstrate that vimentin is required for tumor growth and metastasis. Mechanistically, Vim−/− mice display increased survival rates and reduced tumor burden by inducing ferroptosis, an iron-dependent form of programmed cell death (Berr et al. 2020). In addition, the cell-autonomous ability of Luciferase-tagged vimentin-expressing Luc-KPV+/+cells to metastasize can be assessed using an allograft tumor model. Luc-KPV+/+cells metastasize to the lung, while vimentin-null (Luc-KPV−/−) cells or tumor cells expressing a mutant form of vimentin that cannot assemble full-length vimentin IFs (Luc-KPVY117L) (Berr et al. 2020) fail to metastasize. In a complementary lung adenocarcinoma mouse model, KrasG12D/Lkb1fl/fl/Vim−/−, vimentin-expressing, but not Vim−/− cancer-associated fibroblasts surround epithelial-like collective invasion packs to facilitate metastasis (Richardson et al. 2018; Sharma et al. 2019). Using the Vim−/− mice, these lung cancer models provide causal data that vimentin IFs are required for cancer metastasis and validate the robust clinical data linking vimentin to advanced-stage tumors and poor patient outcomes.
Studies using the Vim−/− mouse cancer models validate vimentin IFs’ role in regulating the metastatic cascade: EMT, invasion, and migration (Fig. 3; Kidd et al. 2014). This is also supported by cell biological studies, which demonstrate that the up-regulation of vimentin expression in epithelial-derived tumor cells is a prerequisite for EMT induction (Kidd et al. 2014; Virtakoivu et al. 2015; Cheng et al. 2016; Peuhu et al. 2017; Wang et al. 2018). Vimentin expression and the accompanying EMT are induced by transforming growth factor β1 (TGF-β1) stimulation (Rogel et al. 2011), Snail overexpression (Cano et al. 2000), ZEB2 overexpression (Bindels et al. 2006), and Slug phosphorylation by ERK (Virtakoivu et al. 2015) in mouse and human epithelial-derived carcinoma cell lines. Lung epithelial cells treated with TGF-β1 rapidly induce vimentin expression via a Smad-binding element located in the 5′ promotor region of the VIM gene (Rogel et al. 2011). Upon the assembly of vimentin IFs, these lung epithelial cells adopt a mesenchymal phenotype. Microinjection of vimentin is sufficient to rapidly induce mesenchymal features in epithelial cells, such as a change in cell shape, loss of desmosomes, and increased cell motility (Mendez et al. 2010). Due to their role in the EMT, it is not surprising that vimentin IFs also play pivotal roles in the ability of cells to invade their surrounding matrix. This is supported by the finding that vimentin IFs facilitate the formation of invadopodia required for cancer cell extravasation and invasion for the initiation of metastasis (Sutoh Yoneyama et al. 2014). Mechanistically, intact vimentin IFs are required to elongate invadopodia, since depleting vimentin with siRNA or disrupting filaments with a dominant-negative probe prevents the elongation (Schoumacher et al. 2010). The unique biophysical properties of vimentin IFs likely enable the cancer cell to breach the basement membrane. These properties include strain stiffening and enhanced resistance to bending and stretching mechanical stresses (Janmey et al. 1991; Guzmán et al. 2006; Köster et al. 2015). Vimentin IFs contribute to tumor tissue stiffening (Rathje et al. 2014; Northey et al. 2017). This is supported by the observation that highly invasive breast carcinoma cells devoid of vimentin are more pliant and less contractile, and lose directional persistence of migration (Mendez et al. 2010; Zhu et al. 2011; Liu et al. 2015).
Figure 3.
Vimentin plays a role in the metastatic cascade. (1) Vimentin is expressed in epithelial-derived tumor cells as they undergo the epithelial-to-mesenchymal transition (EMT). EMT is characterized by the down-regulation of epithelial markers (such as E-cadherin and keratins) and the up-regulation of mesenchymal markers (such as vimentin). (2) The migration of metastatic tumor cells away from the primary tumor is mediated by the formation of lamellipodia. Vimentin IFs play an integral role in lamellipodia formation and maintenance of cell polarity in migrating cells. (3) Dynamic reorganization of vimentin IFs in the lamellipodia is necessary for the formation of cellular polarity, leading to an increase in migration. (2, 4, and 6). Vimentin is required for invadopodia maturation. In Vim−/− mice, there was a significant decrease in the formation of mature invadopodia, cell migration, tumor growth, and distant metastasis.
The experimental data obtained from the Vim−/− mouse and various cell culture studies demonstrate that vimentin IFs are unequivocally major initiators of the EMT and, as such, play a crucial role in tumor progression and metastasis. Furthermore, these data support the hypothesis that modifying the dynamics and mechanotransduction of cancer cells by altering the expression and assembly of vimentin IFs impairs their proliferation, migration, and invasion in 2D and 3D environments. To develop targeted cancer therapy, additional research is required to understand the complexity of vimentin IFs’ roles in the metastatic cascade.
Vimentin regulates inflammatory responses
There is evidence that vimentin IFs play essential roles in coordinating signaling pathways that regulate inflammatory response mediator levels in resident tissue recruit inflammatory cells from the blood, including lymphocytes and phagocytes (Cheng and Eriksson 2017). Following leukocyte adhesion to blood vessels, both the leukocytes and the endothelial cells form microvilli that elongate and embrace the cells; these microvilli are enriched in vimentin but not in actin or tubulin. This process is followed by the penetration of leukocytes through the endothelial cell cytoplasm (Nieminen et al. 2006). Given this involvement of vimentin, it has been of interest to study the inflammatory response in Vim−/− mice. The results show that lymphocyte homing at peripheral lymph nodes and spleen is reduced in Vim−/− mice. Furthermore, ICAM-1 and VCAM-1 expression and clustering are strongly reduced in Vim−/− endothelial cells, and Vim−/− lymphocytes cannot correctly polarize and maintain directional movement (Nieminen et al. 2006). Leukocytes derived from Vim−/− mice (Nieminen et al. 2006) also fail to form the docking/transmigration cup as was originally described in normal leukocytes (Carman and Springer 2004).
These defects are likely related to the finding that in normal WT mice, vimentin IFs typically provide lymphocytes with structural support by regulating their mechanical deformations during chemokine-induced polarization and migration through pores in the endothelium. In support of this, there is a pronounced reorganization of vimentin IFs in circulating lymphocytes in WT mice, and this reorganization plays an essential role in extravasation (Brown et al. 2001). In agreement with this finding, the restructuring of vimentin IFs induced by the phosphoinositide 3-kinase γ (PI3Kγ)–Akt signaling pathway is required for the chemokine-induced transmigration of leukocytes to sites of inflammation (Barberis et al. 2009). Additional support for the critical role of vimentin IFs in lymphocyte migration comes from the finding that vimentin IFs are located in macrophage filopodia and podosomes. Podosomes are dynamic cell adhesion structures involved in the directional motility and transmigration of myeloid cells in WT mice (Calle et al. 2006). Moreover, vimentin binds to fimbrin, a major focal adhesion protein in mouse macrophages (Correia et al. 1999). The comparative studies of Vim−/− and WT mice demonstrate the essential roles of vimentin IFs in immune cell functions related to migration, extravasation, homing, and target recognition.
Vim−/− mice have also provided important insights into the role of vimentin IFs in regulating the activation of the NLRP3 inflammasome. This large multiprotein complex regulates proinflammatory cytokines, specifically IL-1β and IL-18. Lipopolysaccharide (LPS) is a potent inducer of inflammation and a known activator of the NLRP3 inflammasome (dos Santos et al. 2012). Interestingly, Vim−/− mice are protected from LPS-induced injury, and this may be related to the fact that they exhibit decreased caspase-1 activity and reduced IL-1β levels (dos Santos et al. 2015). Moreover, experimental reconstitution of bone marrow from Vim−/− donor mice into irradiated WT recipient mice prevents the secretion of IL-1β following the induction of injury. In vitro experiments further demonstrate that vimentin directly interacts with NLRP3 and caspase-1 and serves as a protein scaffold on which the NLRP3 inflammasome is assembled (dos Santos et al. 2015). In addition, disrupting vimentin IF organization in macrophages with withaferin A (a compound known to disrupt vimentin IF organization in fibroblasts) (Grin et al. 2012) abolishes the formation of NLRP3 inflammasomes (dos Santos et al. 2015; Lang et al. 2018). In agreement with these findings, vimentin IFs interact with macrophage inhibitory factor (MIF), which is also required for NLRP3 inflammasome activation (Lang et al. 2018). These studies demonstrate that vimentin IFs are needed for the assembly of the NLRP3 inflammasome protein complex in the innate immune response system (dos Santos et al. 2015; Lang et al. 2018).
Another way vimentin IFs regulate inflammation involves regulatory T cells (Tregs). Tregs are a specialized subpopulation of T cells that function to suppress the immune response, preventing autoimmune and inflammatory disorders (Prigge et al. 2020). When Tregs interact with an antigen-presenting cell (APC), vimentin IFs reorganize to form a superstructure known as the distal pole complex (DPC) that modulates the suppressive activity of Tregs. Although Tregs have not yet been analyzed in Vim−/− mice, it is interesting to note that WT Tregs silenced for vimentin expression were significantly better at suppressing alloreactive T cell priming in a murine model “graft versus host” disease (GVHD) (McDonald-Hyman et al. 2018). These studies provide a proof of principle that disrupting vimentin IFs is a feasible and translationally relevant method to enhance Treg therapy of GVHD (McDonald-Hyman et al. 2018). This link has been reinforced by a study showing clear up-regulation of vimentin in polarized Treg cells (Mohammad et al. 2018).
In addition to releasing cytokines and growth factors, inflammatory macrophages or phagocytes also generate nitric oxide (NO) and reactive oxygen species (ROS), which strongly influence the physiological states of neighboring cells and tissues. Therefore, it has been interesting to study macrophages derived from Vim−/− mice. The results have shown that the Vim−/− macrophages produce elevated amounts of ROS and NO (Mor-Vaknin et al. 2013) when compared with WT macrophages derived from WT mice in which ROS production is suppressed by interaction with the p47phox active subunit of the NADPH oxidase (Sumimoto 2008; Mor-Vaknin et al. 2013). Thus, the loss of vimentin IFs causes enhanced ROS production in macrophages, resulting in oxidative damage in surrounding tissues. ROS promotes proinflammatory signaling in macrophages (Håversen et al. 2018) and is linked to endothelial dysfunction (Langlois et al. 2017). Further evidence that vimentin IFs regulate ROS-mediated inflammatory responses comes from a study of dextran sodium sulfate-induced colitis in Vim−/− mice. The results demonstrate that Vim−/− mouse phagocytes increase the production of ROS and NO, causing more inflammatory damage to the intestines of the Vim−/− mice than WT controls. This same study shows an essential role for vimentin IFs in regulating bacterially induced inflammatory responses (Mor-Vaknin et al. 2013).
Numerous comparative studies of Vim−/− and WT mice reveal roles for vimentin IFs in many important functions of immune cells related to cellular localization and sequestration of signaling and other key molecules. Given that vimentin IFs are regulated by numerous post-translational modifications triggered by immune cell signaling involving factors such as RhoA kinase, PKA, and PKC; and that they interact with key signaling and sensory systems such as the integrins, NLRP3, caspase1, and 14-3-3 (for review, see Hyder et al. 2011), it is not surprising that these type III intermediate filament cytoskeletal components play essential roles in regulating immune cell functions. Therefore, Vim−/− mice have been invaluable in identifying the critical and diverse roles that vimentin IFs play in immune responses. Importantly, these studies provide a rationale for developing novel therapeutic approaches targeting the regulation of vimentin expression to modulate immune cell responses.
Vimentin regulates the vascular endothelium
Early studies of Vim−/− mice show that vimentin IFs are essential for modulating vascular constriction or tone. Evidence supporting this function derives from partial nephrectomy studies in WT and Vim−/− mice. In WT mice, this surgery triggers rapid and sustained vasodilation of the renal vascular system, permitting the animals to survive, while the same surgical procedure results in renal failure and death in 100% of Vim−/− mice (Terzi et al. 1997). This study also describes a concurrent elevation of endothelin-1 (ET-1) synthesis, which causes a reduction in NO levels. Notably, the death of the Vim−/− mice is entirely prevented by administering an endothelin receptor antagonist (Terzi et al. 1997). These data demonstrate that vimentin IFs modulate vascular tone through the endothelin–NO axis.
Related primarily to tissue homeostasis and tissue repair, the vasculature of Vim−/− mouse embryos is poorly developed compared with WT mouse embryos, as evidenced by markedly fewer vessels with less branching. In addition, aortic ring assays prepared from adult Vim−/− mice treated with VEGF show significant decreases in blood vessel sprout number and length compared with WT controls (Antfolk et al. 2017). In response to hemodynamic stress, Vim−/− mice also have defects in arterial remodeling and vascular smooth muscle cell differentiation (van Engeland et al. 2019).
Another phenotype detected in Vim−/− mice is impaired endothelial barrier function, manifested as a leaky and poorly developed vasculature (Nieminen et al. 2006; Antfolk et al. 2017). Not surprisingly, this appears to be related to poor healing of the endothelium of Vim−/− mice (Boraas and Ahsan 2016), which in turn is associated with the dysregulated generation of granulation tissue as well as abnormal angiogenic sprouting (for review, see Dave and Bayless 2014). Similarly, the absence of vimentin in Müller cells of the Vim−/− mouse retina resulted in reduced pathological vascularization after retinal ischemia—specifically, decreased ability of newly formed vessels to traverse the inner limiting membrane of the retina and invade the vitreous body (Lundkvist et al. 2004).
The impairment of angiogenesis and endothelial cell functions in Vim−/− mice has been linked to the role of vimentin in the Notch signaling pathway. Specifically, vimentin IFs bind to and regulate the signal strength of the proangiogenic Notch ligand, Jagged (Antfolk et al. 2017). Mechanistically, this modulation of signal strength has been related to the shear stress-induced phosphorylation of serine-38 in the N-terminal domain of vimentin. This phosphorylated vimentin interacts with Jagged1 and increases Notch activation potential (van Engeland et al. 2019). Therefore, the abnormal remodeling of the arterial walls of Vim−/− mice likely stems from reduced Jagged1–Notch transactivation strength, which in turn disrupts lateral signal induction through the arterial wall (van Engeland et al. 2019).
Apart from Notch signaling, other activities mediated by vimentin IFs may influence the properties of the vasculature in Vim−/− mice. It is known, for example, that vimentin IFs are generally involved in mechanosensing and plasticity through their interactions with focal adhesions (FAs) and the actomyosin network (Gregor et al. 2014; Shen et al. 2021). In addition, live-cell observations of endothelial cells expressing fluorescently tagged vimentin show that shear stress causes rapid deformation of vimentin IF networks, suggesting a role in mechanosensing and mechanotransduction (Helmke et al. 2000, 2001). Indeed, Vim−/− mice have elevated levels of phosphorylated FA kinase and its targets, Src and ERK1/2 (Langlois et al. 2017). This signaling defect, along with reduced Jagged1–Notch transactivation (van Engeland et al. 2019), has been postulated to associate with increased carotid stiffness, contractility, and endothelial dysfunction independent of blood pressure and the collagen/elastin ratio. This increase in arterial stiffness in Vim−/− mice involves changes in vasomotor tone and organization of the endothelial basement membrane (Langlois et al. 2017; van Engeland et al. 2019).
Vimentin plays a role in fat metabolism, lipid droplet homeostasis, and body weight
Vimentin IFs form a complex cage surrounding the lipid droplets in adipocytes. The vimentin IFs associated with this cage are linked to the lipid droplets (LDs) by perilipin to form the “LD–PLIN–vimentin connection,” and vimentin IFs are thought to be involved in the formation of LDs (Franke et al. 1987; Heid et al. 2014). Based on these findings, it was interesting to determine the impact of the complete absence of vimentin expression on the overall physiology of adipocytes and LDs. In this regard, comparative morphological studies of WT and Vim−/− mice show a reduction in the size of adipocytes and their constituent LDs in the absence of vimentin IFs (Shen et al. 2010). Vim−/− mice fed a standard diet gained less body weight compared with WT controls, and the lower body weight and lower body mass index of Vim−/− mice is due primarily to the lower accumulation of body fat (Wilhelmsson et al. 2019c).
The absence of vimentin also is connected to partial resistance to atherosclerosis induced by the transfer of Vim−/− mouse-derived bone marrow to bone marrow-depleted Vim+/+ mice deficient in the low-density lipoprotein receptor (Håversen et al. 2018). After these mice are fed an atherogenic diet, lipid accumulation below the endothelium of the aortic wall decreases despite increased expression of oxidative stress markers and proinflammatory cytokines by the Vim−/− macrophages. Similar results are obtained by feeding an atherogenic diet to Vim−/− mice made hypercholesterolemic by infection with an adeno-associated virus containing a gain-of-function proprotein convertase (PCSK9) (Håversen et al. 2018). These studies suggest that vimentin has a proatherogenic role by regulating inflammatory responses in macrophages during atherogenesis (Håversen et al. 2018).
The loss of the vimentin IF cage surrounding LDs in Vim−/− mice also affects lipolysis. In WT cells, energy stored in fat is released as fatty acids and cholesterol through lipolysis, controlled mainly by the sympathetic nervous system and insulin. Thus, catecholamines, acting through β-adrenergic receptors, activate adenylyl cyclase, raise intracellular concentrations of cyclic AMP, and stimulate cyclic AMP-dependent protein kinase (PKA) (Duncan et al. 2007). In turn, PKA phosphorylates hormone-sensitive lipase (HSL), the main fatty acid-mobilizing enzyme that facilitates the transfer of cholesterol to mitochondria (Kraemer and Shen 2002). Importantly, there is evidence that vimentin IFs participate in lipolysis through direct, hormonally regulated interactions with HSL (Shen et al. 2003, 2010). Studies of Vim−/− mice also lend significant support for the function of vimentin IFs in lipolysis, as they have a reduced influx of cholesterol into the mitochondria located in both the adrenal glands and ovaries, causing decreases in corticosterone and progesterone (Shen et al. 2012). Vimentin IFs influence lipolysis by interacting with the β3-adrenergic receptor, influencing the stimulation and subsequent activation of ERK. In this regard, a proteomics study showed vimentin IFs associated with the β3-adrenergic receptor following its activation. In addition, depletion of vimentin with shRNA completely inhibited β3-adrenergic receptor-mediated ERK activation and significantly reduced lipolysis in WT mice (Kumar et al. 2007). Thus, vimentin IFs appear to be necessary for the mobilization of cholesterol from lipid droplets to mitochondria for steroidogenesis and overall lipid droplet homeostasis. It is possible that the smaller body size of Vim−/− mice (Wilhelmsson et al. 2019c) is connected to the defect in cell size regulation and mTORC1 signaling recently reported in cells from Vim−/− mice (Mohanasundaram et al. 2021). The functional connections between lipid storage and vimentin IFs revealed by comparative studies of WT and Vim−/− mice may be related to the first vimentin mutation reported in humans, one manifestation of which is lipodystrophy (Cogné et al. 2020; Eriksson 2020).
Interestingly, male Vim−/− mice, but not female Vim−/− mice, show increased mortality, but the cause of death is unknown (Wilhelmsson et al. 2019c). However, since Vim−/− mice have a decreased production of corticosterone (Shen et al. 2012), the main glucocorticoid regulating energy, immune reactions, and stress responses, it is interesting to speculate that the increased mortality of Vim−/− males reflects their higher susceptibility to environmental stress (Wilhelmsson et al. 2019c).
The two-edged sword of vimentin in the central nervous system
Vimentin is expressed in multiple cell types of the nervous system, including neurons, astrocytes, other glial cell types, endothelial cells, neural progenitor cells, and immature neurons. Vim−/− mice show hypermyelination of peripheral nerves and, despite the fact that myelin-forming Schwann cells normally express vimentin, this phenotype seems to be caused by Schwann cell-extrinsic mechanisms that remain to be identified (Triolo et al. 2012). Vim−/− mice also show slower neurite outgrowth and a slower recovery of sensation following sciatic nerve crush (Perlson et al. 2005).
Vimentin is an important component of the cytoplasmic IFs of astrocytes. These cells play key roles in the organization of the CNS and control many functions of the brain, spinal cord, and retina in health and disease (Pekny and Pekna 2014; Pekny et al. 2016, 2019). The up-regulation of vimentin and glial fibrillary acidic protein (GFAP; another type III IF protein), as well as nestin and synemin, the IF proteins of astrocytes, is the hallmark of astrocyte reactivity and reactive gliosis in response to injury, ischemia, or neurodegeneration (Hol and Pekny 2015). The reactivity of astrocytes in Vim−/− mice is attenuated after neurotrauma (Wilhelmsson et al. 2004), and the migratory speed of Vim−/− astrocytes is decreased (Lepekhin et al. 2001).
In Vim−/− mice, GFAP can partially compensate for the absence of vimentin (Eliasson et al. 1999; Pekny et al. 1999b), and thus many revealing phenotypes of vimentin deficiency become apparent only in Vim−/− mice on a GFAP−/− background; i.e., in mice with astrocytes that completely lack cytoplasmic IFs (nestin or synemin cannot form IFs in GFAP−/−Vim−/− astrocytes).
In GFAP−/−Vim−/− mice, astrocytes are present in normal numbers and form normally tiled cellular domains. Upon injury, however, GFAP−/−Vim−/− astrocytes fail to develop their typical hypertrophy of main astrocyte processes (Wilhelmsson et al. 2004) and exhibit other signs of attenuated reactive gliosis, including decreased ERK and c-FOS activation (Nakazawa et al. 2007) or less prominent up-regulation of the 14-3-3 adapter proteins (Sihlbom et al. 2007). While retinal ischemia induces increased stiffness of endfeet and inner processes of retinal Müller cells in WT mice, it fails to do so in GFAP−/−Vim−/− mice (Lu et al. 2011).
The finding that astrocyte reactivity is attenuated in GFAP−/−Vim−/− mice with slower wound healing after brain and spinal cord trauma (Pekny et al. 1999b; Wilhelmsson et al. 2004) has raised the question of whether the slower healing/restorative process might support better regeneration and lead to a better functional outcome (Pekny and Pekna 2014). Indeed, GFAP−/−Vim−/− mice are both worse and better off depending on the specific situation (Table 2).
On the one hand, GFAP−/−Vim−/− mice show lower resistance of the CNS to mechanical stress (Lundkvist et al. 2004; Verardo et al. 2008) and ischemia (Ding et al. 1998; Li et al. 2008; de Pablo et al. 2013; Wunderlich et al. 2015). Ischemic stroke, when induced in GFAP−/−Vim−/− mice, results in a larger tissue loss, with less efficient endothelin-3-induced blockage of gap junction communication among astrocytes (Li et al. 2008), decreased astrocyte glutamine levels (Pekny et al. 1999a), decreased glutamate transport capacity (Li et al. 2008) and lower resistance to oxidative stress (de Pablo et al. 2013), showing that both vimentin and GFAP are important for the neuroprotective functions of astrocytes in acute ischemic stroke. Similarly, fewer cells of the inner retina of GFAP−/−Vim−/− mice survive after retinal ischemia reperfusion (Wunderlich et al. 2015). Interestingly, the absence of vimentin and GFAP does not affect the tissue loss in photothrombotic brain injury (Liu et al. 2014) or neonatal hypoxic–ischemic brain injury (Järlestedt et al. 2010), indicating that the effect of reactive gliosis at the acute stage after injury depends on the lesion type and might differ between immature and adult brains. GFAP−/−Vim−/− mice show an altered response of astrocytes to neuronal degeneration and exhibit a faster progression of Alzheimer's and Batten disease in mouse models (Macauley et al. 2011; Kraft et al. 2013; Kamphuis et al. 2015). The regeneration after sciatic nerve crush in GFAP−/−Vim−/− mice takes longer, but the resulting outcome is not worse (Berg et al. 2013). Remodeling of the corticospinal tract and axonal regeneration after a photothrombotic stroke takes longer, and it was unclear whether it reaches the same level as in WT mice (Liu et al. 2014). The most recent study demonstrated that after brain injury, GFAP−/−Vim−/− mice showed maladaptive neuronal connectivity associated with impaired functional recovery; i.e., increased synaptic plasticity in the perilesional region and increased loss and gain of neuronal connections in sensorimotor networks (Aswendt et al. 2022).
On the other hand, pathological neovascularization that accompanies oxygen-induced retinopathy is reduced in GFAP−/−Vim−/− mice (Lundkvist et al. 2004), and so is photoreceptor cell death and monocyte infiltration in GFAP−/−Vim−/− mice subjected to sodium hyaluronate-induced retinal degeneration (Nakazawa et al. 2007). Moreover, GFAP−/−Vim−/− mice show increased hippocampal neurogenesis in an unchallenged situation (Larsson et al. 2004; Wilhelmsson et al. 2012) with an effect on memory extinction, potentially attributable to the increased rate of reorganization of the hippocampal circuitry (Wilhelmsson et al. 2019b), increased neurogenesis after ischemia (Järlestedt et al. 2010) and neurotrauma (Wilhelmsson et al. 2012), and improved posttraumatic regeneration of neuronal axons and synapses (Menet et al. 2003; Wilhelmsson et al. 2004; Cho et al. 2005). GFAP−/−Vim−/− mice show reduced Notch signaling from astrocytes to neural stem cells (Wilhelmsson et al. 2012; Lebkuechner et al. 2015), a major inhibitory axis controlling adult neurogenesis, and it has been shown that intracellular vesicle trafficking in astrocytes depends on vimentin, GFAP, and also nestin (Potokar et al. 2007, 2010; Vardjan et al. 2012; Wilhelmsson et al. 2019a; Lasič et al. 2020). Interestingly, the CNS environment of GFAP−/− mice is also more supportive of integration and survival of neural grafts (Kinouchi et al. 2003) and supports increased neuronal and astrocyte differentiation of transplanted neural stem cells (Widestrand et al. 2007), indicating that a reduction in the graft-induced reactive gliosis might be the way to improve the success of transplantation of neural grafts or neural progenitor cells in the CNS. However, it remains to be established to what extent the improved graft integration results from attenuated reactive gliosis and to what extent it is the direct consequence of the altered Notch signaling (and possibly other signaling) between host astrocytes and grafted progenitor cells.
Conclusion
It is quite obvious that the Vim−/− mouse model has dramatically advanced our understanding of the molecular and cellular functions of not only vimentin IFs but of cytoplasmic IFs in general. As vimentin is expressed in multiple cell types of mesodermal and ectodermal origin, it is perhaps not surprising that the Vim−/− mice show a remarkable repertoire of phenotypes. Vim−/− mice have been instrumental in linking vimentin IFs to physiologic and pathophysiologic functions related to tissue repair and wound healing; fibrosis; angiogenesis; tumorigenesis; digestive diseases, including Crohn's disease and colitis; inflammatory functions; and host response to infections. Changes in cell morphology, adhesion, stiffness, and migration are commonly reported phenotypes associated with Vim−/− mice. These phenotypes have been linked to reduced wound healing capacity and an inability to properly remodel capillaries and arteries, leading to impaired vascular functions. Some of these phenotypes reflect altered cell activation and cell dynamics, as well as a compromised ability to respond to environmental and physiologic stresses that disrupt tissue integrity. Other phenotypes have positive outcomes, such as improved regenerative responses in the CNS. Thus, by observing stress resilience and tissue repair in the Vim−/− mouse model, important information has been gained on the functions of vimentin and the mechanisms underlying stress and regenerative responses. While the impairment in vimentin-mediated signaling and other functions often results in detrimental phenotypes, depending on the context, elimination of vimentin has also had favorable impacts on certain conditions. For example, certain diseases involve vimentin-mediated maladaptive responses and have consequences that are inhibited or blunted by the absence of vimentin. This review is focused on the impact of the absence of vimentin. In the future, it will be of great interest to compare the effects of loss- or gain-of-function mutations in mouse models. Examples include engineering mice carrying mutations in the vimentin gene at one or more of its numerous phosphorylation sites involved in regulating its assembly states; e.g., phosphovimentin-deficient mice. This approach has already provided insights into the regulatory function of vimentin during development (Matsuyama et al. 2013; Tanaka et al. 2015; Chen et al. 2018; de Pablo et al. 2019). In addition, studies of mice carrying the vimentin mutations known to cause human diseases could also prove to be very useful in defining specific pathways involved in the vimentin-associated pathologies (Muller et al. 2009; Cogné et al. 2020). Notably, there is renewed interest in extracellular vimentin, first reported over two decades ago (Evans et al. 1993), and its ability to act as a danger-associated molecular pattern (DAMP) receptor (Yu et al. 2018). Extracellular vimentin represents an understudied but important area of investigation.
By cataloging the characteristics of Vim−/− mice, researchers have provided a biological context in which drugs and other therapies targeting intermediate filaments can be developed and tested. However, a limitation of the constitutive ablation of vimentin in mice has been the inability to assess the effects of tissue-specific gene ablation. Therefore, an important next step for the field will be the generation of tissue-specific and inducible ablation of vimentin in mice. Another area of research yet to be explored is that vimentin is frequently expressed in early development and later replaced with cell-specific IF proteins such as desmin in muscle (Vaittinen et al. 1999) or neurofilament triplet proteins in neurons (Kirkcaldie and Dwyer 2017). This transition is a gradual process. Moreover, while purified vimentin can self-assemble in vitro, it should be pointed out that it is frequently found as part of IF heteropolymers in the native state of cells and tissues (Herrmann and Aebi 2000). Therefore, it would be interesting to determine whether there is compensatory up-regulation or early onset of expression of other IF types during differentiation in the Vim−/− mouse model. The physiological consequences of the changes in IF gene expression at the wrong times in development may ultimately reflect alterations in the specific mechanical and signaling properties of different IF cytoskeletal networks (Chang and Goldman 2004; Etienne-Manneville 2018; Patteson et al. 2020).
It is interesting to note that reports of human diseases associated with vimentin mutants are strikingly rare. This may partly reflect dire consequences resulting in early termination. There has been little information reflecting on the possible phenotypes one should expect when screening for vimentin mutations. The data from vimentin-deficient mice should be invaluable in determining the types of phenotypes that might be expected in any future screening approaches.
Although the primary goal of this review has been to demonstrate the consequences of knocking out vimentin expression at a more organismic level, it is worthwhile to speculate on the basic cellular and biochemical mechanisms that might help explain the broad range of phenotypes reported in Vim−/− mice. One of the most obvious functions attributed to vimentin IFs at the cellular level is their role in regulating cells’ mechanical and motile properties. For example, it has been shown that vimentin-null MEFs exhibit defective directional cell motility in 2D culture assays (Mendez et al. 2010). In contrast, in 3D transwell migration assays, the Vim−/− MEFs move faster through small pores but experience a dramatic increase in nuclear rupture and DNA damage repair (Patteson et al. 2019). It has also been demonstrated that the force exerted on soft substrates is greatly diminished in Vim−/− MEFs and that both their cytoplasmic stiffness and cortical stiffness are significantly reduced (Guo et al. 2014; Hu et al. 2019; Vahabikashi et al. 2019). These alterations are likely related to the recent discovery that vimentin IFs and F-actin form interpenetrating networks in the cell cortex, and that vimentin IFs regulate the dynamics of actin subunit exchange (Wu et al. 2022). These findings could have significant impacts during early development, when individual cells participate in various forms of active migration through various types of 3D networks. The biochemical and physical properties of vimentin IFs assembled in vitro from purified vimentin have also revealed remarkable strain-stiffening properties (Janmey et al. 1991; Qin et al. 2009) when subjected to forces that would typically rupture F-actin and microtubules. Therefore, it is likely that a combination of these basic properties of vimentin IF contributes significantly to the wide range of phenotypes seen in Vim−/− mice.
In almost three decades since the Vim−/− mouse model was introduced, many investigators have defined functional roles for vimentin in numerous organ, tissue, and cell systems using Vim−/− mice. The large amount of data that has accumulated should put to rest any remaining notions that vimentin IFs are dispensable members of the cytoskeletal repertoire of mammalian systems.
Acknowledgments
We thank Dr. Edward Kuczmarski, Dr. Marcela Pekna, and Dr. Ulrika Wilhelmsson for their help with the manuscript. We thank Henok Karvonen (Turku Bioscience Center) and Jennifer Davis (Northwestern University) for their help with the graphics design work. K.M.R. is supported by the National Institutes of Health (NIH)-National Institute of General Medical Sciences (NIGMS) (GM096971); NIH-National Heart, Lung, and Blood Institute (HL154998 and HL128194); and NIH-National Institute on Aging (AG049665). J.E.E. supported by Academy of Finland, Sigrid Jusélius Foundation, Magnus Ehrnrooth Foundation, the Endowment of the Åbo Akademi University, and the Foundation “Konung Gustaf V:s och Drottning Victorias Frimurarestiftelse”. M.P. is supported by the Swedish Medical Research Council (2017-02255), ALF Gothenburg (146051), AFA (Arbetsmarknadens Försäkringsaktiebolag) Research Foundation, Söderbergs Foundations, Sten A. Olsson Foundation for Research and Culture, Hjärnfonden, Hagströmer's Foundation Millennium, Amlöv's Foundation, E. Jacobson's Donation Fund, VINNOVA (Sweden's Innovation Agency), the Swedish Stroke Foundation, La Caixa Foundation, EU FP 7 Program EduGlia (237956), EU FP 7 Program TargetBraIn (279017), and EU Horizon Europe European Joint Programme on Rare Diseases ALEXANDER (The astrocyte nanofilament system in Alexander disease—from molecules to function, uncovering new leads for therapy). J.E.E. and M.P. have been part of the COST Actions NanoNet (BM1002) and EuroCellNet (CA15214). R.D.G. is supported by the NIH-NIGMS (GM096971 and GM140108).
Footnotes
Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.349358.122.
Freely available online through the Genes & Development Open Access option.
Competing interest statement
The authors declare no competing interests.
References
- Antfolk D, Sjöqvist M, Cheng F, Isoniemi K, Duran CL, Rivero-Muller A, Antila C, Niemi R, Landor S, Bouten CVC, et al. 2017. Selective regulation of Notch ligands during angiogenesis is mediated by vimentin. Proc Natl Acad Sci 114: E4574–E4581. 10.1073/pnas.1703057114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aswendt M, Wilhelmsson U, Wieters F, Stokowska A, Schmitt FJ, Pallast N, de Pablo Y, Mohammed L, Hoehn M, Pekna M, et al. 2022. Reactive astrocytes prevent maladaptive plasticity after ischemic stroke. Prog Neurobiol 209: 102199. 10.1016/j.pneurobio.2021.102199 [DOI] [PubMed] [Google Scholar]
- Barberis L, Pasquali C, Bertschy-Meier D, Cuccurullo A, Costa C, Ambrogio C, Vilbois F, Chiarle R, Wymann M, Altruda F, et al. 2009. Leukocyte transmigration is modulated by chemokine-mediated PI3Kγ-dependent phosphorylation of vimentin. Eur J Immunol 39: 1136–1146. 10.1002/eji.200838884 [DOI] [PubMed] [Google Scholar]
- Bargagna-Mohan P, Paranthan RR, Hamza A, Zhan CG, Lee DM, Kim KB, Lau DL, Srinivasan C, Nakayama K, Nakayama KI, et al. 2012. Corneal antifibrotic switch identified in genetic and pharmacological deficiency of vimentin. J Biol Chem 287: 989–1006. 10.1074/jbc.M111.297150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bargagna-Mohan P, Lei L, Thompson A, Shaw C, Kasahara K, Inagaki M, Mohan R. 2015. Vimentin phosphorylation underlies myofibroblast sensitivity to withaferin a in vitro and during corneal fibrosis. PLoS One 10: e0133399. 10.1371/journal.pone.0133399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berg A, Zelano J, Pekna M, Wilhelmsson U, Pekny M, Cullheim S. 2013. Axonal regeneration after sciatic nerve lesion is delayed but complete in GFAP- and vimentin-deficient mice. PLoS One 8: e79395. 10.1371/journal.pone.0079395 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berr AL, Wiese K, dos Santos G, Davis JM, Koch CM, Anekalla KR, Kidd M, Cheng Y, Hu Y-S, Ridge KM. 2020. Vimentin is required for tumor progression and metastasis in a mouse model of non-small cell lung cancer. bioRxiv 10.1101/2020.06.04.130963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bindels S, Mestdagt M, Vandewalle C, Jacobs N, Volders L, Noël A, van Roy F, Berx G, Foidart JM, Gilles C. 2006. Regulation of vimentin by SIP1 in human epithelial breast tumor cells. Oncogene 25: 4975–4985. 10.1038/sj.onc.1209511 [DOI] [PubMed] [Google Scholar]
- Boraas LC, Ahsan T. 2016. Lack of vimentin impairs endothelial differentiation of embryonic stem cells. Sci Rep 6: 30814. 10.1038/srep30814 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown MJ, Hallam JA, Colucci-Guyon E, Shaw S. 2001. Rigidity of circulating lymphocytes is primarily conferred by vimentin intermediate filaments. J Immunol 166: 6640–6646. 10.4049/jimmunol.166.11.6640 [DOI] [PubMed] [Google Scholar]
- Burch TC, Watson MT, Nyalwidhe JO. 2013. Variable metastatic potentials correlate with differential plectin and vimentin expression in syngeneic androgen independent prostate cancer cells. PLoS One 8: e65005. 10.1371/journal.pone.0065005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calle Y, Carragher NO, Thrasher AJ, Jones GE. 2006. Inhibition of calpain stabilises podosomes and impairs dendritic cell motility. J Cell Sci 119: 2375–2385. 10.1242/jcs.02939 [DOI] [PubMed] [Google Scholar]
- Cano A, Pérez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ, del Barrio MG, Portillo F, Nieto MA. 2000. The transcription factor snail controls epithelial–mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2: 76–83. 10.1038/35000025 [DOI] [PubMed] [Google Scholar]
- Carman CV, Springer TA. 2004. A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them. J Cell Biol 167: 377–388. 10.1083/jcb.200404129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Challa AA, Stefanovic B. 2011. A novel role of vimentin filaments: binding and stabilization of collagen mRNAs. Mol Cell Biol 31: 3773–3789. 10.1128/MCB.05263-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang L, Goldman RD. 2004. Intermediate filaments mediate cytoskeletal crosstalk. Nat Rev Mol Cell Biol 5: 601–613. 10.1038/nrm1438 [DOI] [PubMed] [Google Scholar]
- Chen M, Puschmann TB, Marasek P, Inagaki M, Pekna M, Wilhelmsson U, Pekny M. 2018. Increased neuronal differentiation of neural progenitor cells derived from phosphovimentin-deficient mice. Mol Neurobiol 55: 5478–5489. 10.1007/s12035-017-0759-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng F, Eriksson JE. 2017. Intermediate filaments and the regulation of cell motility during regeneration and wound healing. Cold Spring Harb Perspect Biol 9: a022046. 10.1101/cshperspect.a022046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng F, Shen Y, Mohanasundaram P, Lindstrom M, Ivaska J, Ny T, Eriksson JE. 2016. Vimentin coordinates fibroblast proliferation and keratinocyte differentiation in wound healing via TGF-β–Slug signaling. Proc Natl Acad Sci 113: E4320–E4327. 10.1073/pnas.1519197113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cho KS, Yang L, Lu B, Feng Ma H, Huang X, Pekny M, Chen DF. 2005. Re-establishing the regenerative potential of central nervous system axons in postnatal mice. J Cell Sci 118: 863–872. 10.1242/jcs.01658 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cogné B, Bouameur JE, Hayot G, Latypova X, Pattabiraman S, Caillaud A, Si-Tayeb K, Besnard T, Küry S, Chariau C, et al. 2020. A dominant vimentin variant causes a rare syndrome with premature aging. Eur J Hum Genet 28: 1218–1230. 10.1038/s41431-020-0583-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colucci-Guyon E, Portier MM, Dunia I, Paulin D, Pournin S, Babinet C. 1994. Mice lacking vimentin develop and reproduce without an obvious phenotype. Cell 79: 679–694. 10.1016/0092-8674(94)90553-3 [DOI] [PubMed] [Google Scholar]
- Correia I, Chu D, Chou YH, Goldman RD, Matsudaira P. 1999. Integrating the actin and vimentin cytoskeletons. adhesion-dependent formation of fimbrin-vimentin complexes in macrophages. J Cell Biol 146: 831–842. 10.1083/jcb.146.4.831 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costigliola N, Ding L, Burckhardt CJ, Han SJ, Gutierrez E, Mota A, Groisman A, Mitchison TJ, Danuser G. 2017. Vimentin fibers orient traction stress. Proc Natl Acad Sci 114: 5195–5200. 10.1073/pnas.1614610114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dauphin M, Barbe C, Lemaire S, Nawrocki-Raby B, Lagonotte E, Delepine G, Birembaut P, Gilles C, Polette M. 2013. Vimentin expression predicts the occurrence of metastases in non small cell lung carcinomas. Lung Cancer 81: 117–122. 10.1016/j.lungcan.2013.03.011 [DOI] [PubMed] [Google Scholar]
- Dave JM, Bayless KJ. 2014. Vimentin as an integral regulator of cell adhesion and endothelial sprouting. Microcirculation 21: 333–344. 10.1111/micc.12111 [DOI] [PubMed] [Google Scholar]
- de Pablo Y, Nilsson M, Pekna M, Pekny M. 2013. Intermediate filaments are important for astrocyte response to oxidative stress induced by oxygen-glucose deprivation and reperfusion. Histochem Cell Biol 140: 81–91. 10.1007/s00418-013-1110-0 [DOI] [PubMed] [Google Scholar]
- de Pablo Y, Marasek P, Pozo-Rodrigalvarez A, Wilhelmsson U, Inagaki M, Pekna M, Pekny M. 2019. Vimentin phosphorylation is required for normal cell division of immature astrocytes. Cells 8: 1016. 10.3390/cells8091016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding M, Eliasson C, Betsholtz C, Hamberger A, Pekny M. 1998. Altered taurine release following hypotonic stress in astrocytes from mice deficient for GFAP and vimentin. Brain Res Mol Brain Res 62: 77–81. 10.1016/S0169-328X(98)00240-X [DOI] [PubMed] [Google Scholar]
- Domagala W, Lasota J, Dukowicz A, Markiewski M, Striker G, Weber K, Osborn M. 1990. Vimentin expression appears to be associated with poor prognosis in node-negative ductal NOS breast carcinomas. Am J Pathol 137: 1299–1304. [PMC free article] [PubMed] [Google Scholar]
- dos Santos G, Kutuzov MA, Ridge KM. 2012. The inflammasome in lung diseases. Am J Physiol Lung Cell Mol Physiol 303: L627–L633. 10.1152/ajplung.00225.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- dos Santos G, Rogel MR, Baker MA, Troken JR, Urich D, Morales-Nebreda L, Sennello JA, Kutuzov MA, Sitikov A, Davis JM, et al. 2015. Vimentin regulates activation of the NLRP3 inflammasome. Nat Commun 6: 6574. 10.1038/ncomms7574 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duncan RE, Ahmadian M, Jaworski K, Sarkadi-Nagy E, Sul HS. 2007. Regulation of lipolysis in adipocytes. Annu Rev Nutr 27: 79–101. 10.1146/annurev.nutr.27.061406.093734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eckes B, Dogic D, Colucci-Guyon E, Wang N, Maniotis A, Ingber D, Merckling A, Langa F, Aumailley M, Delouvee A, et al. 1998. Impaired mechanical stability, migration and contractile capacity in vimentin-deficient fibroblasts. J Cell Sci 111: 1897–1907. 10.1242/jcs.111.13.1897 [DOI] [PubMed] [Google Scholar]
- Eckes B, Colucci-Guyon E, Smola H, Nodder S, Babinet C, Krieg T, Martin P. 2000. Impaired wound healing in embryonic and adult mice lacking vimentin. J Cell Sci 113: 2455–2462. 10.1242/jcs.113.13.2455 [DOI] [PubMed] [Google Scholar]
- Eliasson C, Sahlgren C, Berthold CH, Stakeberg J, Celis JE, Betsholtz C, Eriksson JE, Pekny M. 1999. Intermediate filament protein partnership in astrocytes. J Biol Chem 274: 23996–24006. 10.1074/jbc.274.34.23996 [DOI] [PubMed] [Google Scholar]
- Eriksson JE. 2020. Harmful vimentin manifests itself as multiorgan failure. Eur J Hum Genet 28: 1139–1140. 10.1038/s41431-020-0684-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Etienne-Manneville S. 2018. Cytoplasmic intermediate filaments in cell biology. Annu Rev Cell Dev Biol 34: 1–28. 10.1146/annurev-cellbio-100617-062534 [DOI] [PubMed] [Google Scholar]
- Evans DL, Harris DT, Leary JH III, St John AL, Jaso-Friedman L. 1993. Identification of a vimentin-like function associated molecule (FAM) on rat NK cells: evidence for receptor function. Scand J Immunol 37: 131–142. 10.1111/j.1365-3083.1993.tb01748.x [DOI] [PubMed] [Google Scholar]
- Franke WW, Hergt M, Grund C. 1987. Rearrangement of the vimentin cytoskeleton during adipose conversion: formation of an intermediate filament cage around lipid globules. Cell 49: 131–141. 10.1016/0092-8674(87)90763-X [DOI] [PubMed] [Google Scholar]
- Gan Z, Ding L, Burckhardt CJ, Lowery J, Zaritsky A, Sitterley K, Mota A, Costigliola N, Starker CG, Voytas DF, et al. 2016. Vimentin intermediate filaments template microtubule networks to enhance persistence in cell polarity and directed migration. Cell Syst 3: 252–263.e8. 10.1016/j.cels.2016.08.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geerts A, Eliasson C, Niki T, Wielant A, Vaeyens F, Pekny M. 2001. Formation of normal desmin intermediate filaments in mouse hepatic stellate cells requires vimentin. Hepatology 33: 177–188. 10.1053/jhep.2001.21045 [DOI] [PubMed] [Google Scholar]
- Goldman RD. 1971. The role of three cytoplasmic fibers in BHK-21 cell motility. I. Microtubules and the effects of colchicine. J Cell Biol 51: 752–762. 10.1083/jcb.51.3.752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldman RD, Follett EA. 1969. The structure of the major cell processes of isolated BHK21 fibroblasts. Exp Cell Res 57: 263–276. 10.1016/0014-4827(69)90150-5 [DOI] [PubMed] [Google Scholar]
- Gregor M, Osmanagic-Myers S, Burgstaller G, Wolfram M, Fischer I, Walko G, Resch GP, Jörgl A, Herrmann H, Wiche G. 2014. Mechanosensing through focal adhesion-anchored intermediate filaments. FASEB J 28: 715–729. 10.1096/fj.13-231829 [DOI] [PubMed] [Google Scholar]
- Grin B, Mahammad S, Wedig T, Cleland MM, Tsai L, Herrmann H, Goldman RD. 2012. Withaferin a alters intermediate filament organization, cell shape and behavior. PLoS One 7: e39065. 10.1371/journal.pone.0039065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo M, Ehrlicher AJ, Jensen MH, Renz M, Moore JR, Goldman RD, Lippincott-Schwartz J, Mackintosh FC, Weitz DA. 2014. Probing the stochastic, motor-driven properties of the cytoplasm using force spectrum microscopy. Cell 158: 822–832. 10.1016/j.cell.2014.06.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guzmán C, Jeney S, Kreplak L, Kasas S, Kulik AJ, Aebi U, Forró L. 2006. Exploring the mechanical properties of single vimentin intermediate filaments by atomic force microscopy. J Mol Biol 360: 623–630. 10.1016/j.jmb.2006.05.030 [DOI] [PubMed] [Google Scholar]
- Håversen L, Sundelin JP, Mardinoglu A, Rutberg M, Ståhlman M, Wilhelmsson U, Hultén LM, Pekny M, Fogelstrand P, Bentzon JF, et al. 2018. Vimentin deficiency in macrophages induces increased oxidative stress and vascular inflammation but attenuates atherosclerosis in mice. Sci Rep 8: 16973. 10.1038/s41598-018-34659-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heid H, Rickelt S, Zimbelmann R, Winter S, Schumacher H, Dörflinger Y, Kuhn C, Franke WW. 2014. On the formation of lipid droplets in human adipocytes: the organization of the perilipin-vimentin cortex. PLoS One 9: e90386. 10.1371/journal.pone.0090386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Helmke BP, Goldman RD, Davies PF. 2000. Rapid displacement of vimentin intermediate filaments in living endothelial cells exposed to flow. Circ Res 86: 745–752. 10.1161/01.RES.86.7.745 [DOI] [PubMed] [Google Scholar]
- Helmke BP, Thakker DB, Goldman RD, Davies PF. 2001. Spatiotemporal analysis of flow-induced intermediate filament displacement in living endothelial cells. Biophys J 80: 184–194. 10.1016/S0006-3495(01)76006-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henrion D, Terzi F, Matrougui K, Duriez M, Boulanger CM, Colucci-Guyon E, Babinet C, Briand P, Friedlander G, Poitevin P, et al. 1997. Impaired flow-induced dilation in mesenteric resistance arteries from mice lacking vimentin. J Clin Invest 100: 2909–2914. 10.1172/JCI119840 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herrmann H, Aebi U. 2000. Intermediate filaments and their associates: multi-talented structural elements specifying cytoarchitecture and cytodynamics. Curr Opin Cell Biol 12: 79–90. 10.1016/S0955-0674(99)00060-5 [DOI] [PubMed] [Google Scholar]
- Herrmann H, Aebi U. 2004. Intermediate filaments: molecular structure, assembly mechanism, and integration into functionally distinct intracellular scaffolds. Annu Rev Biochem 73: 749–789. 10.1146/annurev.biochem.73.011303.073823 [DOI] [PubMed] [Google Scholar]
- Herrmann H, Aebi U. 2016. Intermediate filaments: structure and assembly. Cold Spring Harb Perspect Biol 8: a018242. 10.1101/cshperspect.a018242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hol EM, Pekny M. 2015. Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system. Curr Opin Cell Biol 32: 121–130. 10.1016/j.ceb.2015.02.004 [DOI] [PubMed] [Google Scholar]
- Hu J, Li Y, Hao Y, Zheng T, Gupta SK, Parada GA, Wu H, Lin S, Wang S, Zhao X, et al. 2019. High stretchability, strength, and toughness of living cells enabled by hyperelastic vimentin intermediate filaments. Proc Natl Acad Sci 116: 17175–17180. 10.1073/pnas.1903890116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang SH, Chi F, Peng L, Bo T, Zhang B, Liu LQ, Wu X, Mor-Vaknin N, Markovitz DM, Cao H, et al. 2016. Vimentin, a novel NF-κB regulator, is required for meningitic escherichia coli K1-induced pathogen invasion and PMN transmigration across the blood-brain barrier. PLoS One 11: e0162641. 10.1371/journal.pone.0162641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hyder CL, Pallari HM, Kochin V, Eriksson JE. 2008. Providing cellular signposts—post-translational modifications of intermediate filaments. FEBS Lett 582: 2140–2148. 10.1016/j.febslet.2008.04.064 [DOI] [PubMed] [Google Scholar]
- Hyder CL, Isoniemi KO, Torvaldson ES, Eriksson JE. 2011. Insights into intermediate filament regulation from development to ageing. J Cell Sci 124: 1363–1372. 10.1242/jcs.041244 [DOI] [PubMed] [Google Scholar]
- Ishikawa H, Bischoff R, Holtzer H. 1969. Formation of arrowhead complexes with heavy meromyosin in a variety of cell types. J Cell Biol 43: 312–328. 10.1083/jcb.43.2.312 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishola T, Da Q, Marrelli SP, Cruz MA. 2015. Vimentin is a novel molecule required for the formation of Von Willebrand factor strings from the vascular endothelium. Blood 126: 2237. 10.1182/blood.V126.23.2237.2237 [DOI] [Google Scholar]
- Janmey PA, Euteneuer U, Traub P, Schliwa M. 1991. Viscoelastic properties of vimentin compared with other filamentous biopolymer networks. J Cell Biol 113: 155–160. 10.1083/jcb.113.1.155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Järlestedt K, Rousset CI, Faiz M, Wilhelmsson U, Ståhlberg A, Sourkova H, Pekna M, Mallard C, Hagberg H, Pekny M. 2010. Attenuation of reactive gliosis does not affect infarct volume in neonatal hypoxic–ischemic brain injury in mice. PLoS One 5: e10397. 10.1371/journal.pone.0010397 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiu Y, Lehtimäki J, Tojkander S, Cheng F, Jäälinoja H, Liu X, Varjosalo M, Eriksson JE, Lappalainen P. 2015. Bidirectional interplay between vimentin intermediate filaments and contractile actin stress fibers. Cell Rep 11: 1511–1518. 10.1016/j.celrep.2015.05.008 [DOI] [PubMed] [Google Scholar]
- Jiu Y, Peranen J, Schaible N, Cheng F, Eriksson JE, Krishnan R, Lappalainen P. 2017. Vimentin intermediate filaments control actin stress fiber assembly through GEF-H1 and RhoA. J Cell Sci 130: 892–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamphuis W, Kooijman L, Orre M, Stassen O, Pekny M, Hol EM. 2015. GFAP and vimentin deficiency alters gene expression in astrocytes and microglia in wild-type mice and changes the transcriptional response of reactive glia in mouse model for Alzheimer's disease. Glia 63: 1036–1056. 10.1002/glia.22800 [DOI] [PubMed] [Google Scholar]
- Kidd ME, Shumaker DK, Ridge KM. 2014. The role of vimentin intermediate filaments in the progression of lung cancer. Am J Respir Cell Mol Biol 50: 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, Sheppard D, Chapman HA. 2006. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci 103: 13180–13185. 10.1073/pnas.0605669103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim S, Kim I, Cho W, Oh GT, Park YM. 2021. Vimentin deficiency prevents high-fat diet-induced obesity and insulin resistance in mice. Diabetes Metab J 45: 97–108. 10.4093/dmj.2019.0198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kinouchi R, Takeda M, Yang L, Wilhelmsson U, Lundkvist A, Pekny M, Chen DF. 2003. Robust neural integration from retinal transplants in mice deficient in GFAP and vimentin. Nat Neurosci 6: 863–868. 10.1038/nn1088 [DOI] [PubMed] [Google Scholar]
- Kirkcaldie MTK, Dwyer ST. 2017. The third wave: intermediate filaments in the maturing nervous system. Mol Cell Neurosci 84: 68–76. 10.1016/j.mcn.2017.05.010 [DOI] [PubMed] [Google Scholar]
- Koch CM, Kishore R, Anekalla KR, Hu Y-S, Davis JM, Ciesielski M, Gadhvi G, Chen S-Y, Turner M, Cheng Y, et al. 2020. Influenza-induced activation of recruited alveolar macrophages during the early inflammatory phase drives lung injury and lethality. bioRxiv 10.1101/2020.06.08.141309 [DOI] [Google Scholar]
- Köster S, Weitz DA, Goldman RD, Aebi U, Herrmann H. 2015. Intermediate filament mechanics in vitro and in the cell: from coiled coils to filaments, fibers and networks. Curr Opin Cell Biol 32: 82–91. 10.1016/j.ceb.2015.01.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kraemer FB, Shen WJ. 2002. Hormone-sensitive lipase: control of intracellular tri-(di-)acylglycerol and cholesteryl ester hydrolysis. J Lipid Res 43: 1585–1594. 10.1194/jlr.R200009-JLR200 [DOI] [PubMed] [Google Scholar]
- Kraft AW, Hu X, Yoon H, Yan P, Xiao Q, Wang Y, Gil SC, Brown J, Wilhelmsson U, Restivo JL, et al. 2013. Attenuating astrocyte activation accelerates plaque pathogenesis in APP/PS1 mice. FASEB J 27: 187–198. 10.1096/fj.12-208660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar N, Robidoux J, Daniel KW, Guzman G, Floering LM, Collins S. 2007. Requirement of vimentin filament assembly for β3-adrenergic receptor activation of ERK MAP kinase and lipolysis. J Biol Chem 282: 9244–9250. 10.1074/jbc.M605571200 [DOI] [PubMed] [Google Scholar]
- Lang T, Lee JPW, Elgass K, Pinar AA, Tate MD, Aitken EH, Fan H, Creed SJ, Deen NS, Traore DAK, et al. 2018. Macrophage migration inhibitory factor is required for NLRP3 inflammasome activation. Nat Commun 9: 2223. 10.1038/s41467-018-04581-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langlois B, Belozertseva E, Parlakian A, Bourhim M, Gao-Li J, Blanc J, Tian L, Coletti D, Labat C, Ramdame-Cherif Z, et al. 2017. Vimentin knockout results in increased expression of sub-endothelial basement membrane components and carotid stiffness in mice. Sci Rep 7: 11628. 10.1038/s41598-017-12024-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larsson A, Wilhelmsson U, Pekna M, Pekny M. 2004. Increased cell proliferation and neurogenesis in the hippocampal dentate gyrus of old GFAP−/−Vim−/− mice. Neurochem Res 29: 2069–2073. 10.1007/s11064-004-6880-2 [DOI] [PubMed] [Google Scholar]
- Lasič E, Trkov Bobnar S, Wilhelmsson U, de Pablo Y, Pekny M, Zorec R, Stenovec M. 2020. Nestin affects fusion pore dynamics in mouse astrocytes. Acta Physiol 228: e13399. 10.1111/apha.13399 [DOI] [PubMed] [Google Scholar]
- Lebkuechner I, Wilhelmsson U, Möllerström E, Pekna M, Pekny M. 2015. Heterogeneity of Notch signaling in astrocytes and the effects of GFAP and vimentin deficiency. J Neurochem 135: 234–248. 10.1111/jnc.13213 [DOI] [PubMed] [Google Scholar]
- Lepekhin EA, Eliasson C, Berthold CH, Berezin V, Bock E, Pekny M. 2001. Intermediate filaments regulate astrocyte motility. J Neurochem 79: 617–625. 10.1046/j.1471-4159.2001.00595.x [DOI] [PubMed] [Google Scholar]
- Li L, Lundkvist A, Andersson D, Wilhelmsson U, Nagai N, Pardo AC, Nodin C, Ståhlberg A, Aprico K, Larsson K, et al. 2008. Protective role of reactive astrocytes in brain ischemia. J Cereb Blood Flow Metab 28: 468–481. 10.1038/sj.jcbfm.9600546 [DOI] [PubMed] [Google Scholar]
- Li FJ, Surolia R, Li H, Wang Z, Liu G, Liu RM, Mirov SB, Athar M, Thannickal VJ, Antony VB. 2017. Low-dose cadmium exposure induces peribronchiolar fibrosis through site-specific phosphorylation of vimentin. Am J Physiol Lung Cell Mol Physiol 313: L80–L91. 10.1152/ajplung.00087.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Z, Li Y, Cui Y, Roberts C, Lu M, Wilhelmsson U, Pekny M, Chopp M. 2014. Beneficial effects of gfap/vimentin reactive astrocytes for axonal remodeling and motor behavioral recovery in mice after stroke. Glia 62: 2022–2033. 10.1002/glia.22723 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu CY, Lin HH, Tang MJ, Wang YK. 2015. Vimentin contributes to epithelial–mesenchymal transition cancer cell mechanics by mediating cytoskeletal organization and focal adhesion maturation. Oncotarget 6: 15966–15983. 10.18632/oncotarget.3862 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu S, Liu L, Ye W, Ye D, Wang T, Guo W, Liao Y, Xu D, Song H, Zhang L, et al. 2016. High vimentin expression associated with lymph node metastasis and predicated a poor prognosis in oral squamous cell carcinoma. Sci Rep 6: 38834. 10.1038/srep38834 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu YB, Iandiev I, Hollborn M, Körber N, Ulbricht E, Hirrlinger PG, Pannicke T, Wei EQ, Bringmann A, Wolburg H, et al. 2011. Reactive glial cells: increased stiffness correlates with increased intermediate filament expression. FASEB J 25: 624–631. 10.1096/fj.10-163790 [DOI] [PubMed] [Google Scholar]
- Lund N, Henrion D, Tiede P, Ziche M, Schunkert H, Ito WD. 2010. Vimentin expression influences flow dependent VASP phosphorylation and regulates cell migration and proliferation. Biochem Biophys Res Commun 395: 401–406. 10.1016/j.bbrc.2010.04.033 [DOI] [PubMed] [Google Scholar]
- Lundkvist A, Reichenbach A, Betsholtz C, Carmeliet P, Wolburg H, Pekny M. 2004. Under stress, the absence of intermediate filaments from Müller cells in the retina has structural and functional consequences. J Cell Sci 117: 3481–3488. 10.1242/jcs.01221 [DOI] [PubMed] [Google Scholar]
- Macauley SL, Pekny M, Sands MS. 2011. The role of attenuated astrocyte activation in infantile neuronal ceroid lipofuscinosis. J Neurosci 31: 15575–15585. 10.1523/JNEUROSCI.3579-11.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marmai C, Sutherland RE, Kim KK, Dolganov GM, Fang X, Kim SS, Jiang S, Golden JA, Hoopes CW, Matthay MA, et al. 2011. Alveolar epithelial cells express mesenchymal proteins in patients with idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 301: L71–L78. 10.1152/ajplung.00212.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matsuyama M, Tanaka H, Inoko A, Goto H, Yonemura S, Kobori K, Hayashi Y, Kondo E, Itohara S, Izawa I, et al. 2013. Defect of mitotic vimentin phosphorylation causes microophthalmia and cataract via aneuploidy and senescence in lens epithelial cells. J Biol Chem 288: 35626–35635. 10.1074/jbc.M113.514737 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDonald-Hyman C, Muller JT, Loschi M, Thangavelu G, Saha A, Kumari S, Reichenbach DK, Smith MJ, Zhang G, Koehn BH, et al. 2018. The vimentin intermediate filament network restrains regulatory T cell suppression of graft-versus-host disease. J Clin Invest 128: 4604–4621. 10.1172/JCI95713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mendez MG, Kojima S, Goldman RD. 2010. Vimentin induces changes in cell shape, motility, and adhesion during the epithelial to mesenchymal transition. FASEB J 24: 1838–1851. 10.1096/fj.09-151639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Menet V, Prieto M, Privat A, Gimenez y Ribotta M. 2003. Axonal plasticity and functional recovery after spinal cord injury in mice deficient in both glial fibrillary acidic protein and vimentin genes. Proc Natl Acad Sci 100: 8999–9004. 10.1073/pnas.1533187100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mohammad I, Nousiainen K, Bhosale SD, Starskaia I, Moulder R, Rokka A, Cheng F, Mohanasundaram P, Eriksson JE, Goodlett DR, et al. 2018. Quantitative proteomic characterization and comparison of T helper 17 and induced regulatory T cells. PLoS Biol 16: e2004194. 10.1371/journal.pbio.2004194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mohanasundaram P, Coelho Rato LS, Modi M, Urbanska M, Lautenschläger F, Cheng F, Eriksson J. 2021. Cytoskeletal vimentin regulates cell size and autophagy through mTORC1 signaling. bioRxiv 10.1101/2021.04.19.440145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mor-Vaknin N, Legendre M, Yu Y, Serezani CH, Garg SK, Jatzek A, Swanson MD, Gonzalez-Hernandez MJ, Teitz-Tennenbaum S, Punturieri A, et al. 2013. Murine colitis is mediated by vimentin. Sci Rep 3: 1045. 10.1038/srep01045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muller M, Bhattacharya SS, Moore T, Prescott Q, Wedig T, Herrmann H, Magin TM. 2009. Dominant cataract formation in association with a vimentin assembly disrupting mutation. Hum Mol Genet 18: 1052–1057. 10.1093/hmg/ddn440 [DOI] [PubMed] [Google Scholar]
- Nakazawa T, Takeda M, Lewis GP, Cho KS, Jiao J, Wilhelmsson U, Fisher SK, Pekny M, Chen DF, Miller JW. 2007. Attenuated glial reactions and photoreceptor degeneration after retinal detachment in mice deficient in glial fibrillary acidic protein and vimentin. Invest Ophthalmol Vis Sci 48: 2760–2768. 10.1167/iovs.06-1398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nieminen M, Henttinen T, Merinen M, Marttila-Ichihara F, Eriksson JE, Jalkanen S. 2006. Vimentin function in lymphocyte adhesion and transcellular migration. Nat Cell Biol 8: 156–162. 10.1038/ncb1355 [DOI] [PubMed] [Google Scholar]
- Northey JJ, Przybyla L, Weaver VM. 2017. Tissue force programs cell fate and tumor aggression. Cancer Discov 7: 1224–1237. 10.1158/2159-8290.CD-16-0733 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patteson AE, Vahabikashi A, Pogoda K, Adam SA, Mandal K, Kittisopikul M, Sivagurunathan S, Goldman A, Goldman RD, Janmey PA. 2019. Vimentin protects cells against nuclear rupture and DNA damage during migration. J Cell Biol 218: 4079–4092. 10.1083/jcb.201902046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patteson AE, Vahabikashi A, Goldman RD, Janmey PA. 2020. Mechanical and non-mechanical functions of filamentous and non-filamentous vimentin. Bioessays 42: e2000078. 10.1002/bies.202000078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pekny M, Pekna M. 2014. Astrocyte reactivity and reactive astrogliosis: costs and benefits. Physiol Rev 94: 1077–1098. 10.1152/physrev.00041.2013 [DOI] [PubMed] [Google Scholar]
- Pekny M, Eliasson C, Siushansian R, Ding M, Dixon SJ, Pekna M, Wilson JX, Hamberger A. 1999a. The impact of genetic removal of GFAP and/or vimentin on glutamine levels and transport of glucose and ascorbate in astrocytes. Neurochem Res 24: 1357–1362. 10.1023/A:1022572304626 [DOI] [PubMed] [Google Scholar]
- Pekny M, Johansson CB, Eliasson C, Stakeberg J, Wallén A, Perlmann T, Lendahl U, Betsholtz C, Berthold CH, Frisén J. 1999b. Abnormal reaction to central nervous system injury in mice lacking glial fibrillary acidic protein and vimentin. J Cell Biol 145: 503–514. 10.1083/jcb.145.3.503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pekny M, Pekna M, Messing A, Steinhäuser C, Lee JM, Parpura V, Hol EM, Sofroniew MV, Verkhratsky A. 2016. Astrocytes: a central element in neurological diseases. Acta Neuropathol 131: 323–345. 10.1007/s00401-015-1513-1 [DOI] [PubMed] [Google Scholar]
- Pekny M, Wilhelmsson U, Tatlisumak T, Pekna M. 2019. Astrocyte activation and reactive gliosis—a new target in stroke? Neurosci Lett 689: 45–55. 10.1016/j.neulet.2018.07.021 [DOI] [PubMed] [Google Scholar]
- Perlson E, Hanz S, Ben-Yaakov K, Segal-Ruder Y, Seger R, Fainzilber M. 2005. Vimentin-dependent spatial translocation of an activated MAP kinase in injured nerve. Neuron 45: 715–726. 10.1016/j.neuron.2005.01.023 [DOI] [PubMed] [Google Scholar]
- Peuhu E, Virtakoivu R, Mai A, Wärri A, Ivaska J. 2017. Epithelial vimentin plays a functional role in mammary gland development. Development 144: 4103–4113. 10.1242/dev.154229 [DOI] [PubMed] [Google Scholar]
- Potokar M, Kreft M, Li L, Daniel Andersson J, Pangršič T, Chowdhury HH, Pekny M, Zorec R. 2007. Cytoskeleton and vesicle mobility in astrocytes. Traffic 8: 12–20. 10.1111/j.1600-0854.2006.00509.x [DOI] [PubMed] [Google Scholar]
- Potokar M, Stenovec M, Gabrijel M, Li L, Kreft M, Grilc S, Pekny M, Zorec R. 2010. Intermediate filaments attenuate stimulation-dependent mobility of endosomes/lysosomes in astrocytes. Glia 58: 1208–1219. 10.1002/glia.21000 [DOI] [PubMed] [Google Scholar]
- Prigge AD, Ma R, Coates BM, Singer BD, Ridge KM. 2020. Age-dependent differences in T-cell responses to influenza a virus. Am J Respir Cell Mol Biol 63: 415–423. 10.1165/rcmb.2020-0169TR [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin Z, Kreplak L, Buehler MJ. 2009. Hierarchical structure controls nanomechanical properties of vimentin intermediate filaments. PLoS One 4: e7294. 10.1371/journal.pone.0007294 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quax W, Egberts WV, Hendriks W, Quax-Jeuken Y, Bloemendal H. 1983. The structure of the vimentin gene. Cell 35: 215–223. 10.1016/0092-8674(83)90224-6 [DOI] [PubMed] [Google Scholar]
- Rathje LS, Nordgren N, Pettersson T, Rönnlund D, Widengren J, Aspenström P, Gad AK. 2014. Oncogenes induce a vimentin filament collapse mediated by HDAC6 that is linked to cell stiffness. Proc Natl Acad Sci 111: 1515–1520. 10.1073/pnas.1300238111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reyfman PA, Gottardi CJ. 2019. Idiopathic pulmonary fibrosis and lung cancer: finding similarities within differences. Am J Respir Cell Mol Biol 61: 667–668. 10.1165/rcmb.2019-0172ED [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richardson AM, Havel LS, Koyen AE, Konen JM, Shupe J, Wiles WG, Martin WD, Grossniklaus HE, Sica G, Gilbert-Ross M, et al. 2018. Vimentin is required for lung adenocarcinoma metastasis via heterotypic tumor cell-cancer-associated fibroblast interactions during collective invasion. Clin Cancer Res 24: 420–432. 10.1158/1078-0432.CCR-17-1776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rogel MR, Soni PN, Troken JR, Sitikov A, Trejo HE, Ridge KM. 2011. Vimentin is sufficient and required for wound repair and remodeling in alveolar epithelial cells. FASEB J 25: 3873–3883. 10.1096/fj.10-170795 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roy S, Kapoor A, Zhu F, Mukhopadhyay R, Ghosh AK, Lee H, Mazzone J, Posner GH, Arav-Boger R. 2020. Artemisinins target the intermediate filament protein vimentin for human cytomegalovirus inhibition. J Biol Chem 295: 15013–15028. 10.1074/jbc.RA120.014116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Runembert I, Queffeulou G, Federici P, Vrtovsnik F, Colucci-Guyon E, Babinet C, Briand P, Trugnan G, Friedlander G, Terzi F. 2002. Vimentin affects localization and activity of sodium-glucose cotransporter SGLT1 in membrane rafts. J Cell Sci 115: 713–724. 10.1242/jcs.115.4.713 [DOI] [PubMed] [Google Scholar]
- Runembert I, Couette S, Federici P, Colucci-Guyon E, Babinet C, Briand P, Friedlander G, Terzi F. 2004. Recovery of Na-glucose cotransport activity after renal ischemia is impaired in mice lacking vimentin. Am J Physiol Renal Physiol 287: F960–F968. 10.1152/ajprenal.00064.2004 [DOI] [PubMed] [Google Scholar]
- Salvador J, Hernandez GE, Ma F, Abrahamson CW, Pelliegrini M, Goldman RD, Ridge KM, Iruela-Arispe ML. 2021. Transcriptional evaluation of the ductus arteriosus at the single-cell level uncovers a requirement for vimentin for complete closure. bioRxiv 10.1101/2021.10.30.466605 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schiffers PM, Henrion D, Boulanger CM, Colucci-Guyon E, Langa-Vuves F, van Essen H, Fazzi GE, Lévy BI, De Mey JG. 2000. Altered flow-induced arterial remodeling in vimentin-deficient mice. Arterioscler Thromb Vasc Biol 20: 611–616. 10.1161/01.ATV.20.3.611 [DOI] [PubMed] [Google Scholar]
- Schoumacher M, Goldman RD, Louvard D, Vignjevic DM. 2010. Actin, microtubules, and vimentin intermediate filaments cooperate for elongation of invadopodia. J Cell Biol 189: 541–556. 10.1083/jcb.200909113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma P, Alsharif S, Fallatah A, Chung BM. 2019. Intermediate filaments as effectors of cancer development and metastasis: a focus on keratins, vimentin, and nestin. Cells 8: 497. 10.3390/cells8050497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen WJ, Patel S, Natu V, Hong R, Wang J, Azhar S, Kraemer FB. 2003. Interaction of hormone-sensitive lipase with steroidogenic acute regulatory protein: facilitation of cholesterol transfer in adrenal. J Biol Chem 278: 43870–43876. 10.1074/jbc.M303934200 [DOI] [PubMed] [Google Scholar]
- Shen WJ, Patel S, Eriksson JE, Kraemer FB. 2010. Vimentin is a functional partner of hormone sensitive lipase and facilitates lipolysis. J Proteome Res 9: 1786–1794. 10.1021/pr900909t [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen WJ, Zaidi SK, Patel S, Cortez Y, Ueno M, Azhar R, Azhar S, Kraemer FB. 2012. Ablation of vimentin results in defective steroidogenesis. Endocrinology 153: 3249–3257. 10.1210/en.2012-1048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen Y, Wu H, Lu PJ, Wang D, Shayegan M, Li H, Shi W, Wang Z, Cai L-H, Xia J, et al. 2021. Effects of vimentin intermediate filaments on the structure and dynamics of in vitro multicomponent interpenetrating cytoskeletal networks. Phys Rev Lett 127: 108101. 10.1103/PhysRevLett.127.108101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sihlbom C, Wilhelmsson U, Li L, Nilsson CL, Pekny M. 2007. 14-3-3 expression in denervated hippocampus after entorhinal cortex lesion assessed by culture-derived isotope tags in quantitative proteomics. J Proteome Res 6: 3491–3500. 10.1021/pr070108e [DOI] [PubMed] [Google Scholar]
- Starger JM, Goldman RD. 1977. Isolation and preliminary characterization of 10-nm filaments from baby hamster kidney (BHK-21) cells. Proc Natl Acad Sci 74: 2422–2426. 10.1073/pnas.74.6.2422 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sumimoto H. 2008. Structure, regulation and evolution of Nox-family NADPH oxidases that produce reactive oxygen species. FEBS J 275: 3249–3277. 10.1111/j.1742-4658.2008.06488.x [DOI] [PubMed] [Google Scholar]
- Surolia R, Li FJ, Wang Z, Li H, Dsouza K, Thomas V, Mirov S, Pérez-Sala D, Athar M, Thannickal VJ, et al. 2019. Vimentin intermediate filament assembly regulates fibroblast invasion in fibrogenic lung injury. JCI Insight 4: e123253. 10.1172/jci.insight.123253 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sutoh Yoneyama M, Hatakeyama S, Habuchi T, Inoue T, Nakamura T, Funyu T, Wiche G, Ohyama C, Tsuboi S. 2014. Vimentin intermediate filament and plectin provide a scaffold for invadopodia, facilitating cancer cell invasion and extravasation for metastasis. Eur J Cell Biol 93: 157–169. 10.1016/j.ejcb.2014.03.002 [DOI] [PubMed] [Google Scholar]
- Tanaka H, Goto H, Inoko A, Makihara H, Enomoto A, Horimoto K, Matsuyama M, Kurita K, Izawa I, Inagaki M. 2015. Cytokinetic failure-induced tetraploidy develops into aneuploidy, triggering skin aging in phosphovimentin-deficient mice. J Biol Chem 290: 12984–12998. 10.1074/jbc.M114.633891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor AC. 1966. Microtubules in the microspikes and cortical cytoplasm of isolated cells. J Cell Biol 28: 155–168. 10.1083/jcb.28.2.155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terzi F, Henrion D, Colucci-Guyon E, Federici P, Babinet C, Levy BI, Briand P, Friedlander G. 1997. Reduction of renal mass is lethal in mice lacking vimentin. Role of endothelin-nitric oxide imbalance. J Clin Invest 100: 1520–1528. 10.1172/JCI119675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Triolo D, Dina G, Taveggia C, Vaccari I, Porrello E, Rivellini C, Domi T, La Marca R, Cerri F, Bolino A, et al. 2012. Vimentin regulates peripheral nerve myelination. Development 139: 1359–1367. 10.1242/dev.072371 [DOI] [PubMed] [Google Scholar]
- Vahabikashi A, Park CY, Perkumas K, Zhang Z, Deurloo EK, Wu H, Weitz DA, Stamer WD, Goldman RD, Fredberg JJ, et al. 2019. Probe sensitivity to cortical versus intracellular cytoskeletal network stiffness. Biophys J 116: 518–529. 10.1016/j.bpj.2018.12.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vaittinen S, Lukka R, Sahlgren C, Rantanen J, Hurme T, Lendahl U, Eriksson JE, Kalimo H. 1999. Specific and innervation-regulated expression of the intermediate filament protein nestin at neuromuscular and myotendinous junctions in skeletal muscle. Am J Pathol 154: 591–600. 10.1016/S0002-9440(10)65304-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Engeland NCA, Suarez Rodriguez F, Rivero-Müller A, Ristori T, Duran CL, Stassen O, Antfolk D, Driessen RCH, Ruohonen S, Ruohonen ST, et al. 2019. Vimentin regulates Notch signaling strength and arterial remodeling in response to hemodynamic stress. Sci Rep 9: 12415. 10.1038/s41598-019-48218-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vardjan N, Gabrijel M, Potokar M, Švajger U, Kreft M, Jeras M, de Pablo Y, Faiz M, Pekny M, Zorec R. 2012. IFN-γ-induced increase in the mobility of MHC class II compartments in astrocytes depends on intermediate filaments. J Neuroinflammation 9: 144. 10.1186/1742-2094-9-144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Verardo MR, Lewis GP, Takeda M, Linberg KA, Byun J, Luna G, Wilhelmsson U, Pekny M, Chen DF, Fisher SK. 2008. Abnormal reactivity of müller cells after retinal detachment in mice deficient in GFAP and vimentin. Invest Ophthalmol Vis Sci 49: 3659–3665. 10.1167/iovs.07-1474 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Virtakoivu R, Mai A, Mattila E, De Franceschi N, Imanishi SY, Corthals G, Kaukonen R, Saari M, Cheng F, Torvaldson E, et al. 2015. Vimentin–ERK signaling uncouples slug gene regulatory function. Cancer Res 75: 2349–2362. 10.1158/0008-5472.CAN-14-2842 [DOI] [PubMed] [Google Scholar]
- Wang Z, Divanyan A, Jourd'heuil FL, Goldman RD, Ridge KM, Jourd'heuil D, Lopez-Soler RI. 2018. Vimentin expression is required for the development of EMT-related renal fibrosis following unilateral ureteral obstruction in mice. Am J Physiol Renal Physiol 315: F769–F780. 10.1152/ajprenal.00340.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Widestrand A, Faijerson J, Wilhelmsson U, Smith PL, Li L, Sihlbom C, Eriksson PS, Pekny M. 2007. Increased neurogenesis and astrogenesis from neural progenitor cells grafted in the hippocampus of GFAP−/− Vim−/− mice. Stem Cells 25: 2619–2627. 10.1634/stemcells.2007-0122 [DOI] [PubMed] [Google Scholar]
- Wilhelmsson U, Li L, Pekna M, Berthold CH, Blom S, Eliasson C, Renner O, Bushong E, Ellisman M, Morgan TE, et al. 2004. Absence of glial fibrillary acidic protein and vimentin prevents hypertrophy of astrocytic processes and improves post-traumatic regeneration. J Neurosci 24: 5016–5021. 10.1523/JNEUROSCI.0820-04.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilhelmsson U, Faiz M, de Pablo Y, Sjöqvist M, Andersson D, Widestrand A, Potokar M, Stenovec M, Smith PL, Shinjyo N, et al. 2012. Astrocytes negatively regulate neurogenesis through the Jagged1-mediated notch pathway. Stem Cells 30: 2320–2329. 10.1002/stem.1196 [DOI] [PubMed] [Google Scholar]
- Wilhelmsson U, Lebkuechner I, Leke R, Marasek P, Yang X, Antfolk D, Chen M, Mohseni P, Lasič E, Bobnar ST, et al. 2019a. Nestin regulates neurogenesis in mice through notch signaling from astrocytes to neural stem cells. Cereb Cortex 29: 4050–4066. 10.1093/cercor/bhy284 [DOI] [PubMed] [Google Scholar]
- Wilhelmsson U, Pozo-Rodrigalvarez A, Kalm M, de Pablo Y, Widestrand A, Pekna M, Pekny M. 2019b. The role of GFAP and vimentin in learning and memory. Biol Chem 400: 1147–1156. 10.1515/hsz-2019-0199 [DOI] [PubMed] [Google Scholar]
- Wilhelmsson U, Stillemark-Billton P, Borén J, Pekny M. 2019c. Vimentin is required for normal accumulation of body fat. Biol Chem 400: 1157–1162. 10.1515/hsz-2019-0170 [DOI] [PubMed] [Google Scholar]
- Wisniewski H, Shelanski ML, Terry RD. 1968. Effects of mitotic spindle inhibitors on neurotubules and neurofilaments in anterior horn cells. J Cell Biol 38: 224–229. 10.1083/jcb.38.1.224 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu H, Shen Y, Sivagurunathan S, Weber MS, Adam SA, Shin JH, Fredberg JJ, Medalia O, Goldman R, Weitz DA. 2022. Vimentin intermediate filaments and filamentous actin form unexpected interpenetrating networks that redefine the cell cortex. Proc Natl Acad Sci 119: e2115217119. 10.1073/pnas.2115217119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wunderlich KA, Tanimoto N, Grosche A, Zrenner E, Pekny M, Reichenbach A, Seeliger MW, Pannicke T, Perez MT. 2015. Retinal functional alterations in mice lacking intermediate filament proteins glial fibrillary acidic protein and vimentin. FASEB J 29: 4815–4828. 10.1096/fj.15-272963 [DOI] [PubMed] [Google Scholar]
- Yu MB, Guerra J, Firek A, Langridge WHR. 2018. Extracellular vimentin modulates human dendritic cell activation. Mol Immunol 104: 37–46. 10.1016/j.molimm.2018.09.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zackroff RV, Goldman RD. 1979. In vitro assembly of intermediate filaments from baby hamster kidney (BHK-21) cells. Proc Natl Acad Sci 76: 6226–6230. 10.1073/pnas.76.12.6226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu QS, Rosenblatt K, Huang KL, Lahat G, Brobey R, Bolshakov S, Nguyen T, Ding Z, Belousov R, Bill K, et al. 2011. Vimentin is a novel AKT1 target mediating motility and invasion. Oncogene 30: 457–470. 10.1038/onc.2010.421 [DOI] [PMC free article] [PubMed] [Google Scholar]




