Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(17): e2220565120, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37071684

ABSTRACT

DNA-based biomaterials have been proposed for tissue engineering approaches due to their predictable assembly into complex morphologies and ease of functionalization. For bone tissue regeneration, the ability to bind Ca2+ and promote hydroxyapatite (HAP) growth along the DNA backbone combined with their degradation and release of extracellular phosphate, a known promoter of osteogenic differentiation, make DNA-based biomaterials unlike other currently used materials. However, their use as biodegradable scaffolds for bone repair remains scarce. Here, we describe the design and synthesis of DNA hydrogels, gels composed of DNA that swell in water, their interactions in vitro with the osteogenic cell lines MC3T3-E1 and mouse calvarial osteoblast, and their promotion of new bone formation in rat calvarial wounds. We found that DNA hydrogels can be readily synthesized at room temperature, and they promote HAP growth in vitro, as characterized by Fourier transform infrared spectroscopy, X-ray diffraction, scanning electron microscopy, atomic force microscopy, and transmission electron microscopy. Osteogenic cells remain viable when seeded on DNA hydrogels in vitro, as characterized by fluorescence microscopy. In vivo, DNA hydrogels promote the formation of new bone in rat calvarial critical size defects, as characterized by micro-computed tomography and histology. This study uses DNA hydrogels as a potential therapeutic biomaterial for regenerating lost bone.


Subject(s)
Hydrogels , Osteogenesis , Mice , Rats , Animals , Hydrogels/chemistry , X-Ray Microtomography , Bone Regeneration , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Durapatite/pharmacology , Durapatite/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry
2.
FASEB J ; 38(11): e23731, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38855909

ABSTRACT

Ca2+ permeation through TRPV4 in fibroblasts is associated with pathological matrix degradation. In human gingival fibroblasts, IL-1ß binding to its signaling receptor (IL-1R1) induces activation of extracellular regulated kinase (ERK) and MMP1 expression, processes that require Ca2+ flux across the plasma membrane. It is not known how IL-1R1, which does not conduct Ca2+, generates Ca2+ signals in response to IL-1. We examined whether TRPV4 mediates the Ca2+ fluxes required for ERK signaling in IL-1 stimulated gingival fibroblasts. TRPV4 was immunostained in fibroblasts of human gingival connective tissue and in focal adhesions of cultured mouse gingival fibroblasts. Human gingival fibroblasts treated with IL-1ß showed no change of TRPV4 expression but there was increased MMP1 expression. In mouse, gingival fibroblasts expressing TRPV4, IL-1 strongly increased [Ca2+]i. Pre-incubation of cells with IL-1 Receptor Antagonist blocked Ca2+ entry induced by IL-1 or the TRPV4 agonist GSK101. Knockout of TRPV4 or expression of a non-Ca2+-conducting TRPV4 pore-mutant or pre-incubation with the TRPV4 inhibitor RN1734, blocked IL-1-induced Ca2+ transients and expression of the mouse interstitial collagenase, MMP13. Treatment of mouse gingival fibroblasts with GSK101 phenocopied Ca2+ and ERK responses induced by IL-1; these responses were absent in TRPV4-null cells or cells expressing a non-conducting TRPV4 pore-mutant. Immunostained IL-1R1 localized with TRPV4 in adhesions within cell extensions. While TRPV4 immunoprecipitates analyzed by mass spectrometry showed no association with IL-1R1, TRPV4 associated with Src-related proteins and Src co-immunoprecipitated with TRPV4. Src inhibition reduced IL-1-induced Ca2+ responses. The functional linkage of TRPV4 with IL-1R1 expands its repertoire of innate immune signaling processes by mediating IL-1-driven Ca2+ responses that drive matrix remodeling in fibroblasts. Thus, inhibiting TRPV4 activity may provide a new pharmacological approach for blunting matrix degradation in inflammatory diseases.


Subject(s)
Calcium Signaling , Fibroblasts , Gingiva , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Animals , Humans , Mice , Fibroblasts/metabolism , Gingiva/metabolism , Gingiva/cytology , Calcium/metabolism , MAP Kinase Signaling System , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-1/metabolism , Interleukin-1/pharmacology , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology
3.
FASEB J ; 37(6): e22946, 2023 06.
Article in English | MEDLINE | ID: mdl-37219464

ABSTRACT

Transient Receptor Potential Vanilloid-type 4 (TRPV4) is a mechanosensitive, Ca2+ -permeable plasma membrane channel that associates with focal adhesions, influences collagen remodeling, and is associated with fibrotic processes through undefined mechanisms. While TRPV4 is known to be activated by mechanical forces transmitted through collagen adhesion receptors containing the ß1 integrin, it is not understood whether TRPV4 affects matrix remodeling by altering ß1 integrin expression and function. We tested the hypothesis that TRPV4 regulates collagen remodeling through its impact on the ß1 integrin in cell-matrix adhesions. In cultured fibroblasts derived from mouse gingival connective tissues, which exhibit very rapid collagen turnover, we found that higher TRPV4 expression is associated with reduced ß1 integrin abundance and adhesion to collagen, reduced focal adhesion size and total adhesion area, and reduced alignment and compaction of extracellular fibrillar collagen. The reduction of ß1 integrin expression mediated by TRPV4 is associated with the upregulation of miRNAs that target ß1 integrin mRNA. Our data suggest a novel mechanism by which TRPV4 modulates collagen remodeling through post-transcriptional downregulation of ß1 integrin expression and function.


Subject(s)
Integrin beta1 , TRPV Cation Channels , Animals , Mice , Cell-Matrix Junctions , Collagen , Focal Adhesions
4.
FASEB J ; 37(8): e23097, 2023 08.
Article in English | MEDLINE | ID: mdl-37440280

ABSTRACT

Colorectal cancer (CRC) is a high prevalence adenocarcinoma with progressive increases in metastasis-related mortality, but the mechanisms governing the extracellular matrix (ECM) degradation important for metastasis in CRC are not well-defined. We investigated a functional relationship between vimentin (Vim) and myosin 10 (Myo10), and whether this relationship is associated with cancer progression. We tested the hypothesis that Vim regulates the aggregation of Myo10 at the tips of cell extensions, which increases membrane-type 1 matrix metalloproteinase (MT1-MMP)-associated local collagen proteolysis and ECM degradation. Analysis of CRC samples revealed colocalization of Vim with Myo10 and MT1-MMP in cell extensions adjacent to sites of collagen degradation, suggesting an association with local cell invasion. We analyzed cultured CRC cells and fibroblasts and found that Vim accelerates aggregation of Myo10 at cell tips, which increases the cell extension rate. Vim stabilizes the interaction of Myo10 with MT1-MMP, which in turn increases collagenolysis. Vim depletion reduced the aggregation of Myo10 at the cell extension tips and MT1-MMP-dependent collagenolysis. We propose that Vim interacts with Myo10, which in turn associates with MT1-MMP to facilitate the transport of these molecules to the termini of cell extensions and there enhance cancer invasion of soft connective tissues.


Subject(s)
Colorectal Neoplasms , Matrix Metalloproteinase 14 , Humans , Matrix Metalloproteinase 14/metabolism , Vimentin/metabolism , Collagen , Myosins
5.
Int J Mol Sci ; 25(7)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38612378

ABSTRACT

Disturbed remodeling of the extracellular matrix (ECM) is frequently observed in several high-prevalence pathologies that include fibrotic diseases of organs such as the heart, lung, periodontium, liver, and the stiffening of the ECM surrounding invasive cancers. In many of these lesions, matrix remodeling mediated by fibroblasts is dysregulated, in part by alterations to the regulatory and effector systems that synthesize and degrade collagen, and by alterations to the functions of the integrin-based adhesions that normally mediate mechanical remodeling of collagen fibrils. Cell-matrix adhesions containing collagen-binding integrins are enriched with regulatory and effector systems that initiate localized remodeling of pericellular collagen fibrils to maintain ECM homeostasis. A large cadre of regulatory molecules is enriched in cell-matrix adhesions that affect ECM remodeling through synthesis, degradation, and contraction of collagen fibrils. One of these regulatory molecules is Transient Receptor Potential Vanilloid-type 4 (TRPV4), a mechanically sensitive, Ca2+-permeable plasma membrane channel that regulates collagen remodeling. The gating of Ca2+ across the plasma membrane by TRPV4 and the consequent generation of intracellular Ca2+ signals affect several processes that determine the structural and mechanical properties of collagen-rich ECM. These processes include the synthesis of new collagen fibrils, tractional remodeling by contractile forces, and collagenolysis. While the specific mechanisms by which TRPV4 contributes to matrix remodeling are not well-defined, it is known that TRPV4 is activated by mechanical forces transmitted through collagen adhesion receptors. Here, we consider how TRPV4 expression and function contribute to physiological and pathological collagen remodeling and are associated with collagen adhesions. Over the long-term, an improved understanding of how TRPV4 regulates collagen remodeling could pave the way for new approaches to manage fibrotic lesions.


Subject(s)
Extracellular Matrix , TRPV Cation Channels , Cell Membrane , Cell-Matrix Junctions , Collagen , Integrins , TRPV Cation Channels/genetics , Humans
6.
J Cell Sci ; 134(6)2021 03 29.
Article in English | MEDLINE | ID: mdl-33558312

ABSTRACT

Vimentin is a structural protein that is required for mesenchymal cell migration and directly interacts with actin, ß1 integrin and paxillin. We examined how these interactions enable vimentin to regulate cell migration on collagen. In fibroblasts, depletion of vimentin increased talin-dependent activation of ß1 integrin by more than 2-fold. Loss of vimentin was associated with reduction of ß1 integrin clustering by 50% and inhibition of paxillin recruitment to focal adhesions by more than 60%, which was restored by vimentin expression. This reduction of paxillin was associated with 65% lower Cdc42 activation, a 60% reduction of cell extension formation and a greater than 35% decrease in cell migration on collagen. The activation of PAK1, a downstream effector of Cdc42, was required for vimentin phosphorylation and filament maturation. We propose that vimentin tunes cell migration through collagen by acting as an adaptor protein for focal adhesion proteins, thereby regulating ß1 integrin activation, resulting in well-organized, mature integrin clusters.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Collagen , Integrin beta1 , Cell Adhesion , Cell Movement , Cluster Analysis , Integrin beta1/genetics , Integrin beta1/metabolism , Paxillin/genetics , Paxillin/metabolism , Vimentin/genetics , Vimentin/metabolism
7.
Oral Dis ; 29(7): 2845-2853, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36458549

ABSTRACT

OBJECTIVES: Current methods for periodontal regeneration do not promote collagen fiber insertions into new bone and cementum. We used a pig wound model to screen different functionalized collagen membranes in promoting periodontal reattachment to root surfaces. METHODS: Treatment groups included (1) control with no membranes, (2) collagen-coated membranes, (3) membranes with insulin-like growth factor-1 (IGF-1), (4) membranes with amelotin, or (5) membranes attached with calcium phosphate cement (CPC), or with CPC combined with IGF-1. Flap procedures were performed on mandibular and maxillary premolars of each pig. RESULTS: Histomorphometric, micro-CT, and clinical measurements obtained at 4 and 12 weeks after surgery showed cementum formation on denuded roots and reformation of alveolar bone, indicating that the pig model can model healing responses in periodontal regeneration. Calcium phosphate cement simplified procedures by eliminating the need for sutures and improved regeneration of alveolar bone (p < 0.05) compared with other treatments. There was a reduction (p < 0.05) of PD only for the IGF group. Large observed variances between treatment groups indicated that a priori power analyses should be conducted to optimize statistical analysis. CONCLUSIONS: Pigs can model discrete elements of periodontal healing using collagen-based, functionalized membranes. Screening indicates that membrane anchorage with calcium phosphate cements improve regeneration of alveolar bone.


Subject(s)
Alveolar Bone Loss , Insulin-Like Growth Factor I , Animals , Swine , Bone Regeneration , Collagen , Dental Cementum , Calcium Phosphates/pharmacology , Guided Tissue Regeneration, Periodontal/methods , Periodontal Ligament , Alveolar Bone Loss/drug therapy
8.
J Cell Physiol ; 237(5): 2451-2468, 2022 05.
Article in English | MEDLINE | ID: mdl-35150133

ABSTRACT

Tissue fibrosis manifests as excessive deposition of compacted, highly aligned collagen fibrils, which interfere with organ structure and function. Cells in collagen-rich lesions often exhibit marked overexpression of discoidin domain receptor 1 (DDR1), which is linked to increased collagen compaction through the association of DDR1 with the Ca2+ -dependent nonmuscle myosin IIA (NMIIA). We examined the functional relationship between DDR1 and the transient receptor potential vanilloid type 4 (TRPV4) channel, a Ca2+ -permeable ion channel that is implicated in collagen compaction. Fibroblasts expressing high levels of DDR1 were used to model cells in lesions with collagen compaction. In these cells, the expression of the ß1 integrin was deleted to simplify studies of DDR1 function. Compared with DDR1 wild-type cells, high DDR1 expression was associated with increased Ca2+ influx through TRPV4, enrichment of TRPV4 in collagen adhesions, and enhanced contractile activity mediated by NMIIA. At cell adhesion sites to collagen, DDR1 associated with TRPV4, which enhanced DDR1-mediated collagen alignment and compaction. We conclude that DDR1 regulates Ca2+ influx through the TRPV4 channel to promote critical, DDR1-mediated processes that are important in lesions with collagen compaction and alignment.


Subject(s)
Calcium , Discoidin Domain Receptor 1 , Calcium/metabolism , Calcium, Dietary , Cell-Matrix Junctions/metabolism , Collagen/metabolism , Discoidin Domain Receptor 1/genetics , Myosins/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
9.
Physiology (Bethesda) ; 36(6): 382-391, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34704856

ABSTRACT

Cells spread on surfaces and within three-dimensional (3-D) matrixes as they grow, divide, and move. Both chemical and physical signals orchestrate spreading during normal development, wound healing, and pathological states such as fibrosis and tumor growth. Diverse molecular mechanisms drive different forms of cell spreading. This article discusses mechanisms by which cells spread in 2-D and 3-D and illustrates new directions in studies of this aspect of cell function.


Subject(s)
Physics , Cell Adhesion , Cell Movement
10.
J Cell Sci ; 133(24)2020 12 23.
Article in English | MEDLINE | ID: mdl-33571108

ABSTRACT

Multinucleated giant cells (MGCs) are prominent in foreign body granulomas, infectious and inflammatory processes, and auto-immune, neoplastic and genetic disorders, but the molecular determinants that specify the formation and function of these cells are not defined. Here, using tandem mass tag-mass spectrometry, we identified a differentially upregulated protein, C-type lectin domain family 10 member (herein denoted CD301, also known as CLEC10A), that was strongly upregulated in mouse RAW264.7 macrophages and primary murine macrophages undergoing interleukin (IL-4)-induced MGC formation. CD301+ MGCs were identified in biopsy specimens of human inflammatory lesions. Function-inhibiting CD301 antibodies or CRISPR/Cas9 deletion of the two mouse CD301 genes (Mgl1 and Mgl2) inhibited IL-4-induced binding of N-acetylgalactosamine-coated beads by 4-fold and reduced MGC formation by 2.3-fold (P<0.05). IL-4-driven fusion and MGC formation were restored by re-expression of CD301 in the knockout cells. We conclude that in monocytes, IL-4 increases CD301 expression, which mediates intercellular adhesion and fusion processes that are required for the formation of MGCs.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Asialoglycoproteins , Cell Fusion , Giant Cells , Interleukin-4 , Lectins, C-Type , Membrane Proteins , Monocytes , Animals , Antibodies , Interleukin-4/genetics , Macrophages , Mice
11.
FASEB J ; 35(10): e21932, 2021 10.
Article in English | MEDLINE | ID: mdl-34549830

ABSTRACT

Myocardial fibrosis is a characteristic of various cardiomyopathies, and myocardial fibroblasts play a central role in this process. Gelsolin (GSN) is an actin severing and capping protein that regulates actin assembly and may be involved in fibroblast activation. While the role of GSN in mechanical stress-mediated cardiac fibrosis has been explored, its role in myocardial fibrosis in the absence of mechanical stress is not defined. In this study, we investigated the role of GSN in myocardial fibrosis induced by Angiotensin II (Ang II), a profibrotic hormone that is elevated in cardiovascular disease. We utilized mice lacking GSN (Gsn-/- ) and cultured primary adult cardiac fibroblasts (cFB). In vivo, Ang II infusion in mice resulted in significantly less severe myocardial fibrosis in Gsn-/- compared with Gsn+/+ mice, along with diminished activation of the TGFß1-Smad2/3 pathway, and reduced expression of cardiac extracellular matrix proteins (collagen, fibronectin, periostin). Moreover, Gsn-deficient hearts exhibited suppressed activity of the AMPK pathway and its downstream effectors, mTOR and P70S6Kinase, which could contribute to the suppressed TGFß1 activity. In vitro, the Ang II-induced activation of cFBs was reduced in Gsn-deficient fibroblasts evident from decreased expression of αSMA and periostin, diminished actin filament turnover; which also exhibited reduced activity of the AMPK-mTOR pathway, and P70S6K phosphorylation. AMPK inhibition compensated for the loss of GSN, restored the levels of G-actin in Gsn-/- cFBs and promoted activation to myofibroblasts by increasing αSMA and periostin levels. This study reveals a novel role for GSN in mediating myocardial fibrosis by regulating the AMPK-mTOR-P70S6K pathway in cFB activation independent from mechanical stress-induced factors.


Subject(s)
Angiotensin II/pharmacology , Fibroblasts/drug effects , Fibrosis/pathology , Gelsolin/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/metabolism , Actins/metabolism , Animals , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/metabolism , Gelsolin/deficiency , Gelsolin/genetics , Homeostasis , Male , Mice , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/drug effects , Myofibroblasts/pathology , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/metabolism
12.
J Biol Chem ; 295(10): 3134-3147, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32005666

ABSTRACT

The actin cytoskeleton is a dynamic array of filaments that undergoes rapid remodeling to drive many cellular processes. An essential feature of filament remodeling is the spatio-temporal regulation of actin filament nucleation. One family of actin filament nucleators, the Diaphanous-related formins, is activated by the binding of small G-proteins such as RhoA. However, RhoA only partially activates formins, suggesting that additional factors are required to fully activate the formin. Here we identify one such factor, IQ motif containing GTPase activating protein-1 (IQGAP1), which enhances RhoA-mediated activation of the Diaphanous-related formin (DIAPH1) and targets DIAPH1 to the plasma membrane. We find that the inhibitory intramolecular interaction within DIAPH1 is disrupted by the sequential binding of RhoA and IQGAP1. Binding of RhoA and IQGAP1 robustly stimulates DIAPH1-mediated actin filament nucleation in vitro In contrast, the actin capping protein Flightless-I, in conjunction with RhoA, only weakly stimulates DIAPH1 activity. IQGAP1, but not Flightless-I, is required to recruit DIAPH1 to the plasma membrane where actin filaments are generated. These results indicate that IQGAP1 enhances RhoA-mediated activation of DIAPH1 in vivo Collectively these data support a model where the combined action of RhoA and an enhancer ensures the spatio-temporal regulation of actin nucleation to stimulate robust and localized actin filament production in vivo.


Subject(s)
Actins/metabolism , Formins/metabolism , ras GTPase-Activating Proteins/metabolism , Actin Cytoskeleton/metabolism , Cell Line, Tumor , Formins/antagonists & inhibitors , Formins/genetics , Humans , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism , ras GTPase-Activating Proteins/antagonists & inhibitors , ras GTPase-Activating Proteins/genetics , rhoA GTP-Binding Protein/metabolism
13.
J Cell Physiol ; 236(2): 1270-1280, 2021 02.
Article in English | MEDLINE | ID: mdl-32643295

ABSTRACT

Many adult connective tissues undergo continuous remodeling to maintain matrix homeostasis. Physiological remodeling involves the degradation of collagen fibers by the intracellular cathepsin-dependent phagocytic pathway. We considered that a multidomain, small GTPase activating protein, IQGAP1, which is involved in the generation of cell extensions, is required for collagen phagocytosis, possibly arising from its interactions with cdc42 and the actin-binding protein Flightless I (FliI). We examined the role of IQGAP1 in collagen phagocytosis by human gingival fibroblasts (HGFs) and by IQGAP1+/+ and IQGAP1-/- mouse embryonic fibroblasts. IQGAP1 was strongly expressed by HGFs, localized to vinculin-stained cell adhesions and sites where cell extensions are initiated, and colocalized with FliI. Immunoprecipitation showed that IQGAP1 associated with FliI. HGFs showed 10-fold increases of collagen binding, 6-fold higher internalization, and 3-fold higher ß1 integrin activation between 30 and 180 min after incubation with collagen. Compared with IQGAP1+/+ fibroblasts, deletion of IQGAP1 reduced collagen binding (1.4-fold), collagen internalization (3-fold), ß1 integrin activation (2-fold), and collagen degradation (1.8-fold). We conclude that IQGAP1 affects collagen remodeling through its regulation of phagocytic degradation pathways, which may involve the interaction of IQGAP1 with FliI.


Subject(s)
Collagen/genetics , Microfilament Proteins/genetics , Phagocytosis/genetics , Trans-Activators/genetics , cdc42 GTP-Binding Protein/genetics , ras GTPase-Activating Proteins/genetics , Animals , Cell Adhesion/genetics , Collagen/metabolism , Fibroblasts/metabolism , Gingiva/metabolism , Gingiva/pathology , Humans , Integrin beta1 , Mice , Monomeric GTP-Binding Proteins/genetics , Protein Binding/genetics , Signal Transduction/genetics
14.
Wound Repair Regen ; 29(4): 602-612, 2021 07.
Article in English | MEDLINE | ID: mdl-33887795

ABSTRACT

The intermediate filament protein vimentin is a widely used phenotypic marker for identifying cells of the mesenchymal linkage such as fibroblasts and myofibroblasts, but the full repertoire of vimentin's functional attributes has not been fully explored. Here we consider how vimentin, in addition to its contributions to mechanical stabilization of cell structure, also helps to control the assembly of cell adhesions and migration through collagen matrices. While the assembly and function of matrix adhesions are critical for the differentiation of myofibroblasts and many other types of adherent cells, a potential mechanism that explains how vimentin affects the recruitment and abundance of centrally important proteins in cell adhesions has been elusive. Here we review recent data indicating that vimentin plays a central regulatory role in the assembly of focal adhesions which form in response to the attachment to collagen. We show that in particular, vimentin is a key organizer of the ß1 integrin adhesive machinery, which affects cell migration through collagen. This review provides a comprehensive picture of the surprisingly broad array of processes and molecules with which vimentin interacts to affect cell function in the context of fibroblast and myofibroblast adhesion and migration on collagen.


Subject(s)
Myofibroblasts , Wound Healing , Cell Adhesion , Collagen , Fibroblasts , Focal Adhesions , Vimentin
15.
J Cell Physiol ; 235(3): 3096-3111, 2020 03.
Article in English | MEDLINE | ID: mdl-31556111

ABSTRACT

Focal adhesion kinase (FAK) is critical for collagen expression but its regulation of collagen remodeling is not defined. We examined the role of FAK in the degradation and reorganization of fibrillar collagen. Compared with wild-type (WT) mouse embryonic fibroblasts, FAK null (FAK-/- ) fibroblasts generated twofold (p < .0001) higher levels of ¾ collagen I fragment and expressed up to fivefold more membrane-type matrix metalloproteinase (MMP). When plated on stiff collagen substrates, compared with WT, FAK-/- cells were smaller (threefold reduced cell surface area; p < .0001) and produced fivefold fewer cell extensions (p < .0001) that were 40% shorter (p < .001). When cultured on soft collagen gels (stiffness of ~100 Pa) for 6-48 hr, cell spreading and cell extension formation were reduced by greater than twofold (p < .05 and p < .0001, respectively) while collagen compaction and alignment were reduced by approximately 30% (p < .0001) in FAK-/- cells. Similar results were found after treatment with PF573228, a FAK inhibitor. Reconstitution of FAK-/- cells with FAK mutants showed that compared with WT, cell extension formation was reduced twofold (p < .0001) in the absence of the kinase domain and sixfold (p < .0001) with a Y397F mutant. Enhanced collagen degradation was exhibited by the mutants (~threefold increase; p < .0001 of ¾ collagen fragments without kinase domain or Y397F mutant; p < .01). Compared with FAK+/+ cells, matrices produced by FAK-/- cells generated higher levels of ß1 integrin activation (p < 0.05), extracellular-signal-regulated kinase (ERK) phosphorylation, and production of ¾ collagen I fragment by human gingival fibroblasts. Collectively these data indicate that (a) the kinase activity of FAK enhances collagen remodeling by tractional forces but inhibits collagen degradation by MMPs; (b) FAK influences the biological activity of fibroblast-secreted extracellular matrices, which in turn impacts ß1 integrin and ERK signaling, and collagen degradation.


Subject(s)
Collagen/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Matrix Metalloproteinases/metabolism , Animals , Cell Adhesion/physiology , Cells, Cultured , Collagen Type I/metabolism , Fibroblasts/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Mice , Signal Transduction/genetics , Signal Transduction/physiology
16.
Am J Physiol Lung Cell Mol Physiol ; 319(2): L294-L311, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32491951

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive, often fatal, fibrosing lung disease for which treatment remains suboptimal. Fibrogenic cytokines, including transforming growth factor-ß (TGF-ß), are central to its pathogenesis. Protein tyrosine phosphatase-α (PTPα) has emerged as a key regulator of fibrogenic signaling in fibroblasts. We have reported that mice globally deficient in PTPα (Ptpra-/-) were protected from experimental pulmonary fibrosis, in part via alterations in TGF-ß signaling. The goal of this study was to determine the lung cell types and mechanisms by which PTPα controls fibrogenic pathways and whether these pathways are relevant to human disease. Immunohistochemical analysis of lungs from patients with IPF revealed that PTPα was highly expressed by mesenchymal cells in fibroblastic foci and by airway and alveolar epithelial cells. To determine whether PTPα promotes profibrotic signaling pathways in lung fibroblasts and/or epithelial cells, we generated mice with conditional (floxed) Ptpra alleles (Ptpraf/f). These mice were crossed with Dermo1-Cre or with Sftpc-CreERT2 mice to delete Ptpra in mesenchymal cells and alveolar type II cells, respectively. Dermo1-Cre/Ptpraf/f mice were protected from bleomycin-induced pulmonary fibrosis, whereas Sftpc-CreERT2/Ptpraf/f mice developed pulmonary fibrosis equivalent to controls. Both canonical and noncanonical TGF-ß signaling and downstream TGF-ß-induced fibrogenic responses were attenuated in isolated Ptpra-/- compared with wild-type fibroblasts. Furthermore, TGF-ß-induced tyrosine phosphorylation of TGF-ß type II receptor and of PTPα were attenuated in Ptpra-/- compared with wild-type fibroblasts. The phenotype of cells genetically deficient in PTPα was recapitulated with the use of a Src inhibitor. These findings suggest that PTPα amplifies profibrotic TGF-ß-dependent pathway signaling in lung fibroblasts.


Subject(s)
Fibroblasts/metabolism , Lung/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Bleomycin/pharmacology , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Lung/drug effects , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/physiology , Signal Transduction/drug effects
17.
J Cell Sci ; 131(11)2018 06 05.
Article in English | MEDLINE | ID: mdl-29724913

ABSTRACT

Adseverin is an actin-binding protein involved in osteoclastogenesis, but its role in inflammation-induced bone loss is not well-defined. Here, we examined whether IL1ß and TNFα regulate adseverin expression to control osteoclastogenesis in mouse primary monocytes and RAW264.7 cells. Adseverin was colocalized with subcortical actin filaments and was enriched in the fusopods of fusing cells. In precursor cells, adseverin overexpression boosted the formation of RANKL-induced multinucleated cells. Both IL1ß and TNFα enhanced RANKL-dependent TRAcP activity by 1.6-fold and multinucleated cell formation (cells with ≥3 nuclei) by 2.6- and 3.3-fold, respectively. However, IL1ß and TNFα did not enhance osteoclast formation in adseverin-knockdown cells. RANKL-dependent adseverin expression in bone marrow cells was increased by both IL1ß (5.4-fold) and TNFα (3.3-fold). Luciferase assays demonstrated that this expression involved transcriptional regulation of the adseverin promoter. Activation of the promoter was restricted to a 1118 bp sequence containing an NF-κB binding site, upstream of the transcription start site. TNFα also promoted RANKL-induced osteoclast precursor cell migration. We conclude that IL1ß and TNFα enhance RANKL-dependent expression of adseverin, which contributes to fusion processes in osteoclastogenesis.


Subject(s)
Gelsolin/genetics , Interleukin-1beta/metabolism , Osteogenesis/physiology , RANK Ligand/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Fusion , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Monocytes , Primary Cell Culture , Promoter Regions, Genetic , RAW 264.7 Cells
18.
Am J Pathol ; 189(6): 1145-1158, 2019 06.
Article in English | MEDLINE | ID: mdl-30926333

ABSTRACT

Monocyte lineage cells play important roles in health and disease. Their differentiation into macrophages is crucial for a broad array of immunologic processes that regulate inflammation, neoplasia, and infection. In certain pathologic conditions, such as foreign body reactions and peripheral inflammatory lesions, monocytes fuse to form large, multinucleated giant cells (MGCs). Currently, our knowledge of the fusion mechanisms of monocytes and the regulation of MGC formation and function in discrete pathologies is limited. Herein, we consider the types and function of MGCs in disease and assess the mechanisms by which monocyte fusion contributes to the formation of MGCs. An improved understanding of the cellular origins and metabolic functions of MGCs will facilitate their identification and ultimately the treatment of diseases and disorders that involve MGCs.


Subject(s)
Giant Cells/pathology , Bone Cysts, Aneurysmal/pathology , Bone Neoplasms/pathology , Cell Adhesion Molecules/physiology , Cell Differentiation/physiology , Cell Fusion , Cherubism/pathology , Giant Cell Arteritis/pathology , Giant Cell Tumor of Bone/pathology , Granuloma, Foreign-Body/pathology , Humans , Macrophage Activation/physiology , Monocytes/physiology , Osteosarcoma/pathology , Tuberculosis/pathology
19.
J Periodontal Res ; 55(6): 959-968, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32725852

ABSTRACT

Tony Melcher, a highly influential and forward-thinking scientist and teacher, focussed on the origins, behaviour and regulation of cells in periodontal tissues. His recent death in April 2020, has motivated us to highlight his multi-level contributions to research in biology and the dental sciences. Tony was particularly adept at recognizing the inherent instructive power of the periodontium, most notably as a model system for studying the inter-relationships between the structure, development and functions of connective tissues. Further, his mentoring of dozens of students who subsequently went on to develop their own careers in research, and his leadership in promoting collaborations in dental sciences world-wide, engendered important advances in the importance and utility of research relating to oral tissues. Here, we reflect upon his development of a large, multi-disciplinary research enterprise, the MRC Group in Periodontal Physiology at the University of Toronto and brief commentaries of those who worked with him there. We examine his early career development and then go on to consider some of his most highly cited publications and their impact on subsequent research trends.


Subject(s)
Periodontal Ligament , Periodontium , Regeneration , Biology/history , Connective Tissue , History, 20th Century , History, 21st Century , Humans
20.
Exp Cell Res ; 379(1): 119-128, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30910400

ABSTRACT

Activated fibroblasts promote physiological wound repair following tissue injury. However, dysregulation of fibroblast activation contributes to the development of fibrosis by enhanced production and contraction of collagen-rich extracellular matrix. At the peak of their activities, fibroblasts undergo phenotypic conversion into highly contractile myofibroblasts by developing muscle-like features, including formation of contractile actin-myosin bundles. The phenotype and function of fibroblasts and myofibroblasts are mechanically regulated by matrix stiffness using a feedback control system that is integrated with the progress of tissue remodelling. The actomyosin contraction machinery and cell-matrix adhesion receptors are critical elements that are needed for mechanosensing by fibroblasts and the translation of mechanical signals into biological responses. Here, we focus on mechanical and chemical regulation of collagen contraction by fibroblasts and the involvement of these factors in their phenotypic conversion to myofibroblasts.


Subject(s)
Collagen/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Animals , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Phenotype , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL