Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters











Publication year range
1.
Exp Cell Res ; 405(1): 112666, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34052237

ABSTRACT

Vimentin protein is one of the main cytoskeleton and plays an important role in cell motility and metastasis. Nowadays, vimentin is widely studied as an epithelial-mesenchymal transition (EMT) marker of cancer cells while its involvement in cancer proliferation is poorly understood. In this study, we investigated the participation of vimentin in regulating cancer proliferation by silencing VIM gene in four cancer cell lines. Our results demonstrated that vimentin loss significantly induced cancer cell proliferation both in vitro and in vivo, which has not been reported so far. Mechanistically, knockdown of vimentin expression activated AKT phosphorylation and its downstream ß-catenin signaling. Nuclear translocation and transcriptional activity of ß-catenin was enhanced after silencing vimentin expression. Furthermore, vimentin loss could prevent Rictor from autophagy-dependent degradation via reducing AMPK-mediated autophagy signaling. AICAR, an AMPK activator, down-regulated Rictor and p-AKT levels while vimentin knockdown could rescue the effects. In vivo, it was also found that Ki67 expression and p-AKT/ß-catenin signaling pathway were obviously up-regulated in the tumor tissues in which vimentin was silenced compared to control groups. Taken together, these data showed the novel function of vimentin in regulating cancer proliferation via Rictor/AKT/ß-catenin signaling pathway, which suggested that it need more careful consideration before inhibiting metastatic cancers through targeting vimentin.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Vimentin/deficiency , beta Catenin/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Tumor Cells, Cultured , Vimentin/genetics , Vimentin/metabolism , Xenograft Model Antitumor Assays , beta Catenin/genetics
2.
Small ; 15(50): e1903180, 2019 12.
Article in English | MEDLINE | ID: mdl-31721440

ABSTRACT

The migration of cells through constricting spaces or along fibrous tracks in tissues is important for many biological processes and depends on the mechanical properties of a cytoskeleton made up of three different filaments: F-actin, microtubules, and intermediate filaments. The signaling pathways and cytoskeletal structures that control cell motility on 2D are often very different from those that control motility in 3D. Previous studies have shown that intermediate filaments can promote actin-driven protrusions at the cell edge, but have little effect on overall motility of cells on flat surfaces. They are however important for cells to maintain resistance to repeated compressive stresses that are expected to occur in vivo. Using mouse embryonic fibroblasts derived from wild-type and vimentin-null mice, it is found that loss of vimentin increases motility in 3D microchannels even though on flat surfaces it has the opposite effect. Atomic force microscopy and traction force microscopy experiments reveal that vimentin enhances perinuclear cell stiffness while maintaining the same level of acto-myosin contractility in cells. A minimal model in which a perinuclear vimentin cage constricts along with the nucleus during motility through confining spaces, providing mechanical resistance against large strains that could damage the structural integrity of cells, is proposed.


Subject(s)
Cell Movement , Vimentin/deficiency , Animals , Biomechanical Phenomena , Capillaries/drug effects , Collagen/pharmacology , Cytoskeleton/metabolism , Hydrogels/pharmacology , Mice , Myosin Type II/metabolism , NIH 3T3 Cells , Vimentin/metabolism
3.
Biophys J ; 116(3): 518-529, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30685055

ABSTRACT

In development, wound healing, and pathology, cell biomechanical properties are increasingly recognized as being of central importance. To measure these properties, experimental probes of various types have been developed, but how each probe reflects the properties of heterogeneous cell regions has remained obscure. To better understand differences attributable to the probe technology, as well as to define the relative sensitivity of each probe to different cellular structures, here we took a comprehensive approach. We studied two cell types-Schlemm's canal endothelial cells and mouse embryonic fibroblasts (MEFs)-using four different probe technologies: 1) atomic force microscopy (AFM) with sharp tip, 2) AFM with round tip, 3) optical magnetic twisting cytometry (OMTC), and 4) traction microscopy (TM). Perturbation of Schlemm's canal cells with dexamethasone treatment, α-actinin overexpression, or RhoA overexpression caused increases in traction reported by TM and stiffness reported by sharp-tip AFM as compared to corresponding controls. By contrast, under these same experimental conditions, stiffness reported by round-tip AFM and by OMTC indicated little change. Knockout (KO) of vimentin in MEFs caused a diminution of traction reported by TM, as well as stiffness reported by sharp-tip and round-tip AFM. However, stiffness reported by OMTC in vimentin-KO MEFs was greater than in wild type. Finite-element analysis demonstrated that this paradoxical OMTC result in vimentin-KO MEFs could be attributed to reduced cell thickness. Our results also suggest that vimentin contributes not only to intracellular network stiffness but also cortex stiffness. Taken together, this evidence suggests that AFM sharp tip and TM emphasize properties of the actin-rich shell of the cell, whereas round-tip AFM and OMTC emphasize those of the noncortical intracellular network.


Subject(s)
Cytoskeleton/metabolism , Mechanical Phenomena , Animals , Biomechanical Phenomena , Endothelial Cells/cytology , Fibroblasts/cytology , Gene Knockout Techniques , Humans , Mice , Vimentin/deficiency , Vimentin/genetics
4.
J Biomech Eng ; 140(6)2018 06 01.
Article in English | MEDLINE | ID: mdl-29450503

ABSTRACT

Contractile stress generation by adherent cells is largely determined by the interplay of forces within their cytoskeleton. It is known that actin stress fibers, connected to focal adhesions, provide contractile stress generation, while microtubules and intermediate filaments provide cells compressive stiffness. Recent studies have shown the importance of the interplay between the stress fibers and the intermediate filament vimentin. Therefore, the effect of the interplay between the stress fibers and vimentin on stress generation was quantified in this study. We hypothesized that net stress generation comprises the stress fiber contraction combined with the vimentin resistance. We expected an increased net stress in vimentin knockout (VimKO) mouse embryonic fibroblasts (MEFs) compared to their wild-type (vimentin wild-type (VimWT)) counterparts, due to the decreased resistance against stress fiber contractility. To test this, the net stress generation by VimKO and VimWT MEFs was determined using the thin film method combined with sample-specific finite element modeling. Additionally, focal adhesion and stress fiber organization were examined via immunofluorescent staining. Net stress generation of VimKO MEFs was three-fold higher compared to VimWT MEFs. No differences in focal adhesion size or stress fiber organization and orientation were found between the two cell types. This suggests that the increased net stress generation in VimKO MEFs was caused by the absence of the resistance that vimentin provides against stress fiber contraction. Taken together, these data suggest that vimentin resists the stress fiber contractility, as hypothesized, thus indicating the importance of vimentin in regulating cellular stress generation by adherent cells.


Subject(s)
Fibroblasts/cytology , Stress, Mechanical , Vimentin/metabolism , Actins/metabolism , Animals , Anisotropy , Biomechanical Phenomena , Fibroblasts/metabolism , Finite Element Analysis , Focal Adhesions/metabolism , Gene Knockout Techniques , Mice , Microtubules/metabolism , Phenotype , Vimentin/deficiency , Vimentin/genetics
5.
Mol Neurobiol ; 55(7): 5478-5489, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28956310

ABSTRACT

Vimentin is an intermediate filament (also known as nanofilament) protein expressed in several cell types of the central nervous system, including astrocytes and neural stem/progenitor cells. Mutation of the vimentin serine sites that are phosphorylated during mitosis (VIM SA/SA ) leads to cytokinetic failures in fibroblasts and lens epithelial cells, resulting in chromosomal instability and increased expression of cell senescence markers. In this study, we investigated morphology, proliferative capacity, and motility of VIM SA/SA astrocytes, and their effect on the differentiation of neural stem/progenitor cells. VIM SA/SA astrocytes expressed less vimentin and more GFAP but showed a well-developed intermediate filament network, exhibited normal cell morphology, proliferation, and motility in an in vitro wound closing assay. Interestingly, we found a two- to fourfold increased neuronal differentiation of VIM SA/SA neurosphere cells, both in a standard 2D and in Bioactive3D cell culture systems, and determined that this effect was neurosphere cell autonomous and not dependent on cocultured astrocytes. Using BrdU in vivo labeling to assess neural stem/progenitor cell proliferation and differentiation in the hippocampus of adult mice, one of the two major adult neurogenic regions, we found a modest increase (by 8%) in the fraction of newly born and surviving neurons. Thus, mutation of the serine sites phosphorylated in vimentin during mitosis alters intermediate filament protein expression but has no effect on astrocyte morphology or proliferation, and leads to increased neuronal differentiation of neural progenitor cells.


Subject(s)
Cell Differentiation , Neural Stem Cells/cytology , Neurons/cytology , Vimentin/deficiency , Vimentin/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Proliferation , Cell Survival , Dentate Gyrus/cytology , Intermediate Filaments/metabolism , Mice, Inbred C57BL , Neurogenesis , Phosphorylation , Spheroids, Cellular/cytology , Wound Healing
6.
Development ; 144(22): 4103-4113, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28947532

ABSTRACT

In the mammary gland, vimentin intermediate filaments are expressed in stromal cells and in basal epithelial cell populations, including gland-reconstituting mammary stem cells, with largely undefined functions. Here, we have studied how vimentin deficiency affects mouse mammary gland development. We find that, in adult vimentin knockout mice (Vim-/- ), mammary ductal outgrowth is delayed. The adult Vim-/- glands display dilated ducts and a reduced basal-to-luminal mouse mammary epithelial cell (MMEC) ratio indicative of altered progenitor cell activity. Accordingly, isolated Vim-/- MMECs form fewer mammospheres and basal-like organoids in vitro than their wild-type counterparts. Importantly, reduced basal MMEC number translates into defects in Vim-/- mammary gland regeneration in vivo Global gene expression profiling of basal MMECs reveals that lack of vimentin alters multiple pathways, including adhesion, cancer and Wnt signalling. Furthermore, vimentin contributes to stem-like cell properties in MDA-MB-231 breast cancer cells, wherein vimentin depletion reduces tumoursphere formation and attenuates expression of breast cancer stem cell-associated surface markers. Together, our findings identify vimentin as a positive regulator of stemness in the developing mouse mammary gland and in breast cancer cells.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Vimentin/metabolism , Animals , Cell Count , Cell Line, Tumor , Cell Proliferation , Epithelial Cells/cytology , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Silencing , Humans , Mammary Glands, Animal/cytology , Mice, Knockout , Organoids/metabolism , Regeneration , Spheroids, Cellular/pathology , Stem Cells/cytology , Stem Cells/metabolism , Stromal Cells/metabolism , Vimentin/deficiency
7.
Sci Rep ; 7(1): 11628, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912461

ABSTRACT

Intermediate filaments are involved in stress-related cell mechanical properties and in plasticity via the regulation of focal adhesions (FAs) and the actomyosin network. We investigated whether vimentin regulates endothelial cells (ECs) and vascular smooth muscle cells (SMCs) and thereby influences vasomotor tone and arterial stiffness. Vimentin knockout mice (Vim-/-) exhibited increased expression of laminin, fibronectin, perlecan, collagen IV and VE-cadherin as well as von Willebrand factor deposition in the subendothelial basement membrane. Smooth muscle (SM) myosin heavy chain, α-SM actin and smoothelin were decreased in Vim-/- mice. Electron microscopy revealed a denser endothelial basement membrane and increased SM cell-matrix interactions. Integrin αv, talin and vinculin present in FAs were increased in Vim-/- mice. Phosphorylated FA kinase and its targets Src and ERK1/2 were elevated in Vim-/- mice. Knockout of vimentin, but not of synemin, resulted in increased carotid stiffness and contractility and endothelial dysfunction, independently of blood pressure and the collagen/elastin ratio. The increase in arterial stiffness in Vim-/- mice likely involves vasomotor tone and endothelial basement membrane organization changes. At the tissue level, the results show the implication of FAs both in ECs and vascular SMCs in the role of vimentin in arterial stiffening.


Subject(s)
Basement Membrane/metabolism , Carotid Artery Diseases/etiology , Carotid Artery Diseases/metabolism , Gene Expression Regulation , Intermediate Filaments/genetics , Intermediate Filaments/metabolism , Vascular Stiffness/genetics , Vimentin/deficiency , Animals , Biomarkers , Blood Pressure , Carotid Artery Diseases/physiopathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Endothelium/metabolism , Fluorescent Antibody Technique , Mechanical Phenomena , Mice , Mice, Knockout , Microscopy, Confocal , Vasodilation/genetics
8.
Proc Natl Acad Sci U S A ; 114(23): E4574-E4581, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28533359

ABSTRACT

Notch signaling is a key regulator of angiogenesis, in which sprouting is regulated by an equilibrium between inhibitory Dll4-Notch signaling and promoting Jagged-Notch signaling. Whereas Fringe proteins modify Notch receptors and strengthen their activation by Dll4 ligands, other mechanisms balancing Jagged and Dll4 signaling are yet to be described. The intermediate filament protein vimentin, which has been previously shown to affect vascular integrity and regenerative signaling, is here shown to regulate ligand-specific Notch signaling. Vimentin interacts with Jagged, impedes basal recycling endocytosis of ligands, but is required for efficient receptor ligand transendocytosis and Notch activation upon receptor binding. Analyses of Notch signal activation by using chimeric ligands with swapped intracellular domains (ICDs), demonstrated that the Jagged ICD binds to vimentin and contributes to signaling strength. Vimentin also suppresses expression of Fringe proteins, whereas depletion of vimentin enhances Fringe levels to promote Dll4 signaling. In line with these data, the vasculature in vimentin knockout (VimKO) embryos and placental tissue is underdeveloped with reduced branching. Disrupted angiogenesis in aortic rings from VimKO mice and in endothelial 3D sprouting assays can be rescued by reactivating Notch signaling by recombinant Jagged ligands. Taken together, we reveal a function of vimentin and demonstrate that vimentin regulates Notch ligand signaling activities during angiogenesis.


Subject(s)
Neovascularization, Physiologic , Receptors, Notch/metabolism , Vimentin/metabolism , Animals , Aorta/metabolism , Chick Embryo , Endocytosis , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Ligands , Mice , Mice, 129 Strain , Mice, Knockout , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transcriptional Activation , Vimentin/deficiency , Vimentin/genetics
9.
Proc Natl Acad Sci U S A ; 113(30): E4320-7, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27466403

ABSTRACT

Vimentin has been shown to be involved in wound healing, but its functional contribution to this process is poorly understood. Here we describe a previously unrecognized function of vimentin in coordinating fibroblast proliferation and keratinocyte differentiation during wound healing. Loss of vimentin led to a severe deficiency in fibroblast growth, which in turn inhibited the activation of two major initiators of epithelial-mesenchymal transition (EMT), TGF-ß1 signaling and the Zinc finger transcriptional repressor protein Slug, in vimentin-deficient (VIM(-/-)) wounds. Correspondingly, VIM(-/-) wounds exhibited loss of EMT-like keratinocyte activation, limited keratinization, and slow reepithelialization. Furthermore, the fibroblast deficiency abolished collagen accumulation in the VIM(-/-) wounds. Vimentin reconstitution in VIM(-/-) fibroblasts restored both their proliferation and TGF-ß1 production. Similarly, restoring paracrine TGF-ß-Slug-EMT signaling reactivated the transdifferentiation of keratinocytes, reviving their migratory properties, a critical feature for efficient healing. Our results demonstrate that vimentin orchestrates the healing by controlling fibroblast proliferation, TGF-ß1-Slug signaling, collagen accumulation, and EMT processing, all of which in turn govern the required keratinocyte activation.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , Snail Family Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Vimentin/genetics , Wound Healing/genetics , Animals , Animals, Newborn , Cells, Cultured , Epithelial-Mesenchymal Transition/genetics , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling/methods , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , Signal Transduction/genetics , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Vimentin/deficiency
10.
Oncotarget ; 7(10): 11299-309, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26849235

ABSTRACT

Bone dissemination and bone disease occur in approximately 80% of patients with multiple myeloma (MM) and are a major cause of patient mortality. We previously demonstrated that MM cell-derived heparanase (HPSE) is a major driver of MM dissemination to and progression in new bone sites. However the mechanism(s) by which HPSE promotes MM progression remains unclear. In the present study, we investigated the involvement of mesenchymal features in HPSE-promoted MM progression in bone. Using a combination of molecular, biochemical, cellular, and in vivo approaches, we demonstrated that (1) HPSE enhanced the expression of mesenchymal markers in both MM and vascular endothelial cells; (2) HPSE expression in patient myeloma cells positively correlated with the expression of the mesenchymal markers vimentin and fibronectin. Additional mechanistic studies revealed that the enhanced mesenchymal-like phenotype induced by HPSE in MM cells is due, at least in part, to the stimulation of the ERK signaling pathway. Finally, knockdown of vimentin in HPSE expressing MM cells resulted in significantly attenuated MM cell dissemination and tumor growth in vivo. Collectively, these data demonstrate that the mesenchymal features induced by HPSE in MM cells contribute to enhanced tumor cell motility and bone-dissemination.


Subject(s)
Cell Movement/physiology , Glucuronidase/metabolism , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/pathology , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Growth Processes/physiology , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Glucuronidase/antagonists & inhibitors , Heparin/analogs & derivatives , Heparin/pharmacology , Heterografts , Humans , MAP Kinase Signaling System , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Multiple Myeloma/metabolism , Phenotype , Signal Transduction , Tumor Microenvironment , Vimentin/deficiency , Vimentin/genetics , Vimentin/metabolism
11.
J Neurochem ; 135(2): 234-48, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26118771

ABSTRACT

Astrocytes have multiple roles in the CNS including control of adult neurogenesis. We recently showed that astrocyte inhibition of neurogenesis through Notch signaling depends on the intermediate filament proteins glial fibrillary acidic protein (GFAP) and vimentin. Here, we used real-time quantitative PCR to analyze gene expression in individual mouse astrocytes in primary cultures and in GFAP(POS) or Aldh1L1(POS) astrocytes freshly isolated from uninjured, contralesional and lesioned hippocampus 4 days after entorhinal cortex lesion. To determine the Notch signaling competence of individual astrocytes, we measured the mRNA levels of Notch ligands and Notch1 receptor. We found that whereas most cultured and freshly isolated astrocytes were competent to receive Notch signals, only a minority of astrocytes were competent to send Notch signals. Injury increased the fraction of astrocyte subpopulation unable to send and receive Notch signals, thus resembling primary astrocytes in vitro. Astrocytes deficient of GFAP and vimentin showed decreased Notch signal sending competence and altered expression of Notch signaling pathway-related genes Dlk2, Notch1, and Sox2. Furthermore, we identified astrocyte subpopulations based on their mRNA and protein expression of nestin and HB-EGF. This study improves our understanding of astrocyte heterogeneity, and points to astrocyte cytoplasmic intermediate filaments as targets for neural cell replacement strategies.


Subject(s)
Astrocytes/physiology , Glial Fibrillary Acidic Protein/deficiency , Glial Fibrillary Acidic Protein/genetics , Receptors, Notch/genetics , Receptors, Notch/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Vimentin/deficiency , Vimentin/genetics , Animals , Epidermal Growth Factor/genetics , Hippocampus/cytology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Notch1 , SOXB1 Transcription Factors
12.
Oncotarget ; 6(18): 15966-83, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-25965826

ABSTRACT

Modulations of cytoskeletal organization and focal adhesion turnover correlate to tumorigenesis and epithelial-mesenchymal transition (EMT), the latter process accompanied by the loss of epithelial markers and the gain of mesenchymal markers (e.g., vimentin). Clinical microarray results demonstrated that increased levels of vimentin mRNA after chemotherapy correlated to a poor prognosis of breast cancer patients. We hypothesized that vimentin mediated the reorganization of cytoskeletons to maintain the mechanical integrity in EMT cancer cells. By using knockdown strategy, the results showed reduced cell proliferation, impaired wound healing, loss of directional migration, and increased large membrane extension in MDA-MB 231 cells. Vimentin depletion also induced reorganization of cytoskeletons and reduced focal adhesions, which resulted in impaired mechanical strength because of reduced cell stiffness and contractile force. In addition, overexpressing vimentin in MCF7 cells increased cell stiffness, elevated cell motility and directional migration, reoriented microtubule polarity, and increased EMT phenotypes due to the increased ß1-integrin and the loss of junction protein E-cadherin. The EMT-related transcription factor slug was also mediated by vimentin. The current study demonstrated that vimentin serves as a regulator to maintain intracellular mechanical homeostasis by mediating cytoskeleton architecture and the balance of cell force generation in EMT cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cytoskeleton/metabolism , Focal Adhesions/physiology , Vimentin/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cytoskeleton/pathology , Epithelial-Mesenchymal Transition/physiology , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells , Microtubules/metabolism , Microtubules/pathology , Vimentin/deficiency , Vimentin/genetics
13.
Glia ; 63(6): 1036-56, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25731615

ABSTRACT

Reactive astrocytes with an increased expression of intermediate filament (IF) proteins Glial Fibrillary Acidic Protein (GFAP) and Vimentin (VIM) surround amyloid plaques in Alzheimer's disease (AD). The functional consequences of this upregulation are unclear. To identify molecular pathways coupled to IF regulation in reactive astrocytes, and to study the interaction with microglia, we examined WT and APPswe/PS1dE9 (AD) mice lacking either GFAP, or both VIM and GFAP, and determined the transcriptome of cortical astrocytes and microglia from 15- to 18-month-old mice. Genes involved in lysosomal degradation (including several cathepsins) and in inflammatory response (including Cxcl5, Tlr6, Tnf, Il1b) exhibited a higher AD-induced increase when GFAP, or VIM and GFAP, were absent. The expression of Aqp4 and Gja1 displayed the same pattern. The downregulation of neuronal support genes in astrocytes from AD mice was absent in GFAP/VIM null mice. In contrast, the absence of IFs did not affect the transcriptional alterations induced by AD in microglia, nor was the cortical plaque load altered. Visualizing astrocyte morphology in GFAP-eGFP mice showed no clear structural differences in GFAP/VIM null mice, but did show diminished interaction of astrocyte processes with plaques. Microglial proliferation increased similarly in all AD groups. In conclusion, absence of GFAP, or both GFAP and VIM, alters AD-induced changes in gene expression profile of astrocytes, showing a compensation of the decrease of neuronal support genes and a trend for a slightly higher inflammatory expression profile. However, this has no consequences for the development of plaque load, microglial proliferation, or microglial activation.


Subject(s)
Alzheimer Disease/metabolism , Astrocytes/metabolism , Glial Fibrillary Acidic Protein/deficiency , Microglia/metabolism , Vimentin/deficiency , Aged , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Cell Proliferation/physiology , Chemokine CXCL5/metabolism , Disease Models, Animal , Gene Expression/physiology , Glial Fibrillary Acidic Protein/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , Vimentin/genetics
14.
PLoS One ; 8(11): e79395, 2013.
Article in English | MEDLINE | ID: mdl-24223940

ABSTRACT

Peripheral axotomy of motoneurons triggers Wallerian degeneration of injured axons distal to the lesion, followed by axon regeneration. Centrally, axotomy induces loss of synapses (synaptic stripping) from the surface of lesioned motoneurons in the spinal cord. At the lesion site, reactive Schwann cells provide trophic support and guidance for outgrowing axons. The mechanisms of synaptic stripping remain elusive, but reactive astrocytes and microglia appear to be important in this process. We studied axonal regeneration and synaptic stripping of motoneurons after a sciatic nerve lesion in mice lacking the intermediate filament (nanofilament) proteins glial fibrillary acidic protein (GFAP) and vimentin, which are upregulated in reactive astrocytes and Schwann cells. Seven days after sciatic nerve transection, ultrastructural analysis of synaptic density on the somata of injured motoneurons revealed more remaining boutons covering injured somata in GFAP(-/-)Vim(-/-) mice. After sciatic nerve crush in GFAP(-/-)Vim(-/-) mice, the fraction of reinnervated motor endplates on muscle fibers of the gastrocnemius muscle was reduced 13 days after the injury, and axonal regeneration and functional recovery were delayed but complete. Thus, the absence of GFAP and vimentin in glial cells does not seem to affect the outcome after peripheral motoneuron injury but may have an important effect on the response dynamics.


Subject(s)
Axons/pathology , Glial Fibrillary Acidic Protein/deficiency , Nerve Regeneration , Sciatic Neuropathy/physiopathology , Vimentin/deficiency , Animals , Axotomy , Female , Glial Fibrillary Acidic Protein/metabolism , Mice , Motor Neurons/pathology , Muscles/innervation , Myelin Sheath/physiology , Recovery of Function , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Synapses/pathology , Up-Regulation , Vimentin/metabolism
15.
PLoS One ; 8(7): e70699, 2013.
Article in English | MEDLINE | ID: mdl-23936241

ABSTRACT

Neurotrauma or focal brain ischemia are known to trigger molecular and structural responses in the uninjured hemisphere. These responses may have implications for tissue repair processes as well as for the recovery of function. To determine whether the plasticity response in the uninjured hemisphere occurs even after a subtle trauma, we subjected mice to a partial unilateral deafferentation of the hippocampus induced by stereotactically performed entorhinal cortex lesion (ECL). The expression of selected genes was assessed by quantitative real-time PCR in the hippocampal tissue at the injured side and the contralesional side at day 4 and 14 after injury. We observed that expression of genes coding for synaptotagmin 1, ezrin, thrombospondin 4, and C1q proteins, that have all been implicated in the synapse formation, re-arrangement and plasticity, were upregulated both in the injured and the contralesional hippocampus, implying a plasticity response in the uninjured hemisphere. Several of the genes, the expression of which was altered in response to ECL, are known to be expressed in astrocytes. To test whether astrocyte activation plays a role in the observed plasticity response to ECL, we took advantage of mice deficient in two intermediate filament (nanofilament) proteins glial fibrillary acidic protein (GFAP) and vimentin (GFAP(-/-)Vim(-/-) ) and exhibiting attenuated astrocyte activation and reactive gliosis. The absence of GFAP and vimentin reduced the ECL-induced upregulation of thrombospondin 4, indicating that this response to ECL depends on astrocyte activation and reactive gliosis. We conclude that even a very limited focal neurotrauma triggers a distinct response at the contralesional side, which at least to some extent depends on astrocyte activation.


Subject(s)
Afferent Pathways/metabolism , Cerebrum/metabolism , Craniocerebral Trauma/metabolism , Entorhinal Cortex/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Neuronal Plasticity , Afferent Pathways/injuries , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cerebrum/injuries , Craniocerebral Trauma/physiopathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Entorhinal Cortex/injuries , Gene Expression Profiling , Glial Fibrillary Acidic Protein , Hippocampus/injuries , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Stereotaxic Techniques , Synaptotagmin I/genetics , Synaptotagmin I/metabolism , Thrombospondins/genetics , Thrombospondins/metabolism , Vimentin/deficiency , Vimentin/genetics
16.
Mol Cell Endocrinol ; 371(1-2): 15-9, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23089211

ABSTRACT

Intracellular lipid droplets (LDs) are dynamic organelles that contain a number of associated proteins including perilipin (Plin) and vimentin. Cholesteryl ester (CE)-rich LDs normally accumulate in steroidogenic cells and their mobilization is the preferred initial source of cholesterol for steroidogenesis. Plin1a, 1b and 5 were found to preferentially associate with triacylglycerol-rich LDs and Plin1c and Plin4 to associate with CE-rich LDs, but the biological significance of this remains unanswered. Vimentin null mice were found to have decreased ACTH-stimulated corticosterone levels, and decreased progesterone levels in females, but normal hCG-stimulated testosterone levels in males. Smaller LDs were seen in null cells. Lipoprotein cholesterol delivery to adrenals and ovary was normal, as was the expression of steroidogenic genes; however, the movement of cholesterol to mitochondria was reduced in vimentin null mice. These results suggest that vimentin is important in the maintenance of CE-rich LDs and in the movement of cholesterol for steroidogenesis.


Subject(s)
Adrenal Glands/metabolism , Cholesterol Esters/chemistry , Mitochondria/metabolism , Steroids/biosynthesis , Vimentin/deficiency , Adrenocorticotropic Hormone/metabolism , Animals , Carrier Proteins/metabolism , Corticosterone/blood , Humans , Mice , Perilipin-1 , Perilipin-4 , Perilipin-5 , Phosphoproteins/metabolism , Progesterone/blood , Proteins/metabolism , Testosterone/blood , Vimentin/genetics
17.
Stem Cells ; 30(10): 2320-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22887872

ABSTRACT

Adult neurogenesis is regulated by a number of cellular players within the neurogenic niche. Astrocytes participate actively in brain development, regulation of the mature central nervous system (CNS), and brain plasticity. They are important regulators of the local environment in adult neurogenic niches through the secretion of diffusible morphogenic factors, such as Wnts. Astrocytes control the neurogenic niche also through membrane-associated factors, however, the identity of these factors and the mechanisms involved are largely unknown. In this study, we sought to determine the mechanisms underlying our earlier finding of increased neuronal differentiation of neural progenitor cells when cocultured with astrocytes lacking glial fibrillary acidic protein (GFAP) and vimentin (GFAP(-/-) Vim(-/-) ). We used primary astrocyte and neurosphere cocultures to demonstrate that astrocytes inhibit neuronal differentiation through a cell-cell contact. GFAP(-/-) Vim(-/-) astrocytes showed reduced endocytosis of Notch ligand Jagged1, reduced Notch signaling, and increased neuronal differentiation of neurosphere cultures. This effect of GFAP(-/-) Vim(-/-) astrocytes was abrogated in the presence of immobilized Jagged1 in a manner dependent on the activity of γ-secretase. Finally, we used GFAP(-/-) Vim(-/-) mice to show that in the absence of GFAP and vimentin, hippocampal neurogenesis under basal conditions as well as after injury is increased. We conclude that astrocytes negatively regulate neurogenesis through the Notch pathway, and endocytosis of Notch ligand Jagged1 in astrocytes and Notch signaling from astrocytes to neural stem/progenitor cells depends on the intermediate filament proteins GFAP and vimentin.


Subject(s)
Astrocytes/metabolism , Calcium-Binding Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Receptors, Notch/genetics , Vimentin/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Astrocytes/cytology , Calcium-Binding Proteins/metabolism , Cell Communication/genetics , Cell Differentiation , Coculture Techniques , Endocytosis , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Primary Cell Culture , Receptors, Notch/metabolism , Serrate-Jagged Proteins , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Vimentin/deficiency , Wnt Proteins/genetics , Wnt Proteins/metabolism
18.
J Neurosci ; 31(43): 15575-85, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22031903

ABSTRACT

Infantile neuronal ceroid lipofuscinosis (INCL) is an inherited neurodegenerative disorder affecting the CNS during infancy. INCL is caused by mutations in the CLN1 gene that lead to a deficiency in the lysosomal hydrolase, palmitoyl protein thioesterase 1 (PPT1). A murine model of INCL, the PPT1-deficient (PPT1(-/-)) mouse, is an accurate phenocopy of the human disease. The first pathological change observed in the PPT1(-/-) brain is regional areas of glial fibrillary acidic protein (GFAP) upregulation, which predicts future areas of neurodegeneration. We hypothesized that preventing GFAP and vimentin upregulation in reactive astrocytes will alter the CNS disease. To test this hypothesis, we generated mice simultaneously carrying null mutations in the GFAP, Vimentin, and PPT1 genes (GFAP(-/-)Vimentin(-/-)PPT1(-/-)). Although the clinical and pathological features of the GFAP(-/-)Vimentin(-/-)PPT1(-/-) mice are similar to INCL, the disease appears earlier and progresses more rapidly. One mechanism underlying this accelerated phenotype is a profound neuroinflammatory response within the CNS. Thus, our data identify a protective role for intermediate filament upregulation during astrocyte activation in INCL, a model of chronic neurodegeneration.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/physiopathology , Up-Regulation/genetics , Analysis of Variance , Animals , Blood-Testis Barrier/physiopathology , Brain/metabolism , Brain/pathology , Capillary Permeability/genetics , Cerebral Cortex/pathology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Glial Fibrillary Acidic Protein/deficiency , Humans , Infarction, Middle Cerebral Artery/complications , Intermediate Filaments/genetics , Intermediate Filaments/metabolism , Longevity/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Ceroid-Lipofuscinoses/genetics , Organ Size/genetics , RNA, Messenger/metabolism , Silver Staining , Thiolester Hydrolases/deficiency , Vimentin/deficiency
19.
Am J Physiol Renal Physiol ; 300(5): F1244-54, 2011 May.
Article in English | MEDLINE | ID: mdl-21270094

ABSTRACT

Vitronectin (Vtn) is a glycoprotein found in normal serum and pathological extracellular matrix. Given its known interactions with plasminogen activator inhibitor-1 (PAI-1) and Vtn cellular receptors, especially αvß3 integrin and the urokinase receptor (uPAR), this study was designed to investigate its role in renal fibrogenesis in the mouse model of unilateral ureteral obstruction (UUO). Kidney Vtn mRNA levels were increased ×1.8-5.1 and Vtn protein levels ×1.9-3 on days 7, 14, and 21 after UUO compared with sham kidney levels. Groups of age-matched C57BL/6 wild-type (Vtn+/+) and Vtn-/- mice (n = 10-11/group) were killed 7, 14, or 21 days after UUO. Absence of Vtn resulted in the following significant differences, but only on day 14: fewer αSMA+ interstitial myofibroblasts (×0.53), lower procollagen III mRNA levels (×0.41), lower PAI-1 protein (×0.23), higher uPA activity (×1.1), and lower αv protein (×0.32). The number of CD68+ macrophages did not differ between the genotypes. Despite these transient differences on day 14, the absence of Vtn had no effect on fibrosis severity based on both picrosirius red-positive interstitial area and total kidney collagen measured by the hydroxyproline assay. These findings suggest that despite significant interstitial Vtn deposition in the UUO model of chronic kidney disease, its fibrogenic role is either nonessential or redundant. These data are remarkable given Vtn's strong affinity for the potent fibrogenic molecule PAI-1.


Subject(s)
Kidney Diseases/etiology , Kidney/metabolism , Myofibroblasts/metabolism , Ureteral Obstruction/complications , Vimentin/metabolism , Animals , Chronic Disease , Disease Models, Animal , Fibrosis , Genotype , Integrin alphaVbeta3/metabolism , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/pathology , Phenotype , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activators/metabolism , RNA, Messenger/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Time Factors , Up-Regulation , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology , Vimentin/deficiency , Vimentin/genetics
20.
Blood ; 114(2): 478-84, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19420356

ABSTRACT

Pathologische Anatomie Leiden-endothelium antibody has been used for more than 20 years as a marker for vascular endothelium. Despite its widespread use, the target of this antibody was only recently identified as plasmalemma vesicle-associated protein-1 (PV-1). However, no function has been identified for this molecule. Here we report that activation of human umbilical vein endothelial cells with tumor necrosis factor-alpha resulted in a remarkable redistribution of PV-1 toward the peripheral areas of the cells. Furthermore, in vitro endpoint transmigration experiments showed that transcellularly migrating lymphocytes are surrounded by rings containing PV-1 and caveolin-1. Moreover, PV-1 associates physically with vimentin. In addition, administration of anti-PV-1 antibody during capillary flow assays resulted in a significant inhibition of lymphocyte transmigration through the endothelial cell layer, whereas rolling and adhesion were unaffected. In vivo blockage of PV-1 by an antibody in acute peritonitis and air pouch model resulted in a significant decrease in the number of migrating leukocytes. Here we thus define leukocyte transendothelial migration as the first known function for PV-1.


Subject(s)
Carrier Proteins/metabolism , Chemotaxis, Leukocyte , Endothelial Cells/cytology , Leukocytes/cytology , Leukocytes/metabolism , Membrane Proteins/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Endothelium/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Vimentin/deficiency , Vimentin/genetics , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL