Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 432
Filtrar
1.
Methods Mol Biol ; 2761: 257-266, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38427242

RESUMO

Cytoskeletal dysregulation forms an important aspect of many neurodegenerative diseases such as Alzheimer's disease. Cytoskeletal functions require the dynamic activity of the cytoskeletal proteins-actin, tubulin, and the associated proteins. One of such important phenomena is that of actin remodeling, which helps the cell to migrate, navigate, and interact with extracellular materials. Podosomes are complex actin-rich cytoskeletal structures, abundant in proteins that interact and degrade the extracellular matrix, enabling cells to displace and migrate. The formation of podosomes requires extensive actin networks and remodeling. Here we present a novel immunofluorescence-based approach to study actin remodeling in neurons through the medium of podosomes.


Assuntos
Actinas , Podossomos , Actinas/metabolismo , Podossomos/metabolismo , Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Citoesqueleto de Actina/metabolismo
2.
Curr Biol ; 34(6): R244-R246, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38531317

RESUMO

During cancer progression, tumor cells need to disseminate by remodeling the extracellular tumor matrix. A recent study sheds light on the intricate cooperation between caveolae and invadosomes that facilitates the spread of cancer cells.


Assuntos
Podossomos , Humanos , Podossomos/patologia , Cavéolas , Matriz Extracelular , Invasividade Neoplásica/patologia , Crime
3.
Biochem Biophys Res Commun ; 704: 149636, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38402724

RESUMO

Osteoclasts are hematopoietic cells attached to the bones containing type I collagen-deposited hydroxyapatite during bone resorption. Two major elements determine the stiffness of bones: regular calcified bone (bone that is resorbable by osteoclasts) and un-calcified osteoid bone (bone that is un-resorbable by osteoclasts). The osteolytic cytokine RANKL promotes osteoclast differentiation; however, the roles of the physical interactions of osteoclasts with calcified and un-calcified bone at the sealing zones and the subsequent cellular signaling remain unclear. In this study, we investigated podosomes, actin-rich adhesion structures (actin-ring) in the sealing zone that participates in sensing hard stiffness with collagen in the physical environment during osteoclast differentiation. RANKL-induced osteoclast differentiation induction was promoted when Raw264.7 cells were cultured on collagen-coated plastic dishes but not on non-coated plastic dishes, which was associated with the increased expression of podosome-related genes and Src. In contrast, when cells were cultured on collagen gel, expression of podosome-related genes and Src were not upregulated. The induction of podosome-related genes and Src requires hard stiffness with RGD-containing substratum and integrin-mediated F-actin polymerization. These results indicate that osteoclasts sense both the RGD sequence and stiffness of calcified collagen through their podosome components regulating osteoclast differentiation via the c-Src pathway.


Assuntos
Reabsorção Óssea , Podossomos , Humanos , Osteoclastos/metabolismo , Podossomos/metabolismo , Actinas/metabolismo , Diferenciação Celular/fisiologia , Reabsorção Óssea/metabolismo , Proteína Tirosina Quinase CSK/metabolismo , Colágeno/metabolismo , Oligopeptídeos/metabolismo
4.
Bull Math Biol ; 86(3): 30, 2024 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-38347328

RESUMO

One of the most crucial and lethal characteristics of solid tumors is represented by the increased ability of cancer cells to migrate and invade other organs during the so-called metastatic spread. This is allowed thanks to the production of matrix metalloproteinases (MMPs), enzymes capable of degrading a type of collagen abundant in the basal membrane separating the epithelial tissue from the connective one. In this work, we employ a synergistic experimental and mathematical modelling approach to explore the invasion process of tumor cells. A mathematical model composed of reaction-diffusion equations describing the evolution of the tumor cells density on a gelatin substrate, MMPs enzymes concentration and the degradation of the gelatin is proposed. This is completed with a calibration strategy. We perform a sensitivity analysis and explore a parameter estimation technique both on synthetic and experimental data in order to find the optimal parameters that describe the in vitro experiments. A comparison between numerical and experimental solutions ends the work.


Assuntos
Podossomos , Humanos , Podossomos/metabolismo , Podossomos/patologia , Gelatina/metabolismo , Matriz Extracelular/patologia , Modelos Biológicos , Conceitos Matemáticos , Metaloproteinases da Matriz/metabolismo , Invasividade Neoplásica/patologia
5.
J Cell Biol ; 223(3)2024 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-38353696

RESUMO

The microtubule-associated protein MAP1B has been implicated in axonal growth and brain development. We found that MAP1B is highly expressed in the most aggressive and deadliest breast cancer subtype, triple-negative breast cancer (TNBC), but not in other subtypes. Expression of MAP1B was found to be highly correlated with poor prognosis. Depletion of MAP1B in TNBC cells impairs cell migration and invasion concomitant with a defect in tumorigenesis. We found that MAP1B interacts with key components for invadopodia formation, cortactin, and Tks5, the latter of which is a PtdIns(3,4)P2-binding and scaffold protein that localizes to invadopodia. We also found that Tks5 associates with microtubules and supports the association between MAP1B and α-tubulin. In accordance with their interaction, depletion of MAP1B leads to Tks5 destabilization, leading to its degradation via the autophagic pathway. Collectively, these findings suggest that MAP1B is a convergence point of the cytoskeleton to promote malignancy in TNBC and thereby a potential diagnostic and therapeutic target for TNBC.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular , Cortactina , Proteínas Associadas aos Microtúbulos , Neoplasias de Mama Triplo Negativas , Humanos , Carcinogênese/genética , Transformação Celular Neoplásica , Cortactina/genética , Proteínas Associadas aos Microtúbulos/genética , Neoplasias de Mama Triplo Negativas/genética , Células MDA-MB-231 , Proteínas Adaptadoras de Transporte Vesicular/genética , Microtúbulos/metabolismo , Citoesqueleto/metabolismo , Feminino , Animais , Camundongos , Camundongos Endogâmicos BALB C , Podossomos/metabolismo , Tubulina (Proteína)/metabolismo
6.
Cancer Sci ; 115(3): 836-846, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38273817

RESUMO

Matrix stiffness potently promotes the malignant phenotype in various biological contexts. Therefore, identification of gene expression to participate in mechanical force signals transduced into downstream biochemical signaling will contribute substantially to the advances in nasopharyngeal carcinoma (NPC) treatment. In the present study, we detected that cortactin (CTTN) played an indispensable role in matrix stiffness-induced cell migration, invasion, and invadopodia formation. Advances in cancer research have highlighted that dysregulated alternative splicing contributes to cancer progression as an oncogenic driver. However, whether WT-CTTN or splice variants (SV1-CTTN or SV2-CTTN) regulate matrix stiffness-induced malignant phenotype is largely unknown. We proved that alteration of WT-CTTN expression modulated matrix stiffness-induced cell migration, invasion, and invadopodia formation. Considering that splicing factors might drive cancer progression through positive feedback loops, we analyzed and showed how the splicing factor PTBP2 and TIA1 modulated the production of WT-CTTN. Moreover, we determined that high stiffness activated PTBP2 expression. Taken together, our findings showed that the PTBP2-WT-CTTN level increases upon stiffening and then promotes cell migration, invasion, and invadopodia formation in NPC.


Assuntos
Neoplasias Nasofaríngeas , Podossomos , Humanos , Cortactina/genética , Cortactina/metabolismo , Carcinoma Nasofaríngeo/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Nasofaríngeas/genética , Invasividade Neoplásica
7.
Cancer Sci ; 115(2): 369-384, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38050654

RESUMO

In gastric cancer (GC), the liver is a common organ for distant metastasis, and patients with gastric cancer with liver metastasis (GCLM) generally have poor prognosis. The mechanism of GCLM is unclear. Invadopodia are special membrane protrusions formed by tumor cells that can degrade the basement membrane and ECM. Herein, we investigated the role of invadopodia in GCLM. We found that the levels of invadopodia-associated proteins were significantly higher in liver metastasis than in the primary tumors of patients with GCLM. Furthermore, GC cells could activate hepatic stellate cells (HSCs) within the tumor microenvironment of liver metastases through the secretion of platelet-derived growth factor subunit B (PDGFB). Activated HSCs secreted hepatocyte growth factor (HGF), which activated the MET proto-oncogene, MET receptor of GC cells, thereby promoting invadopodia formation through the PI3K/AKT pathway and subsequently enhancing the invasion and metastasis of GC cells. Therefore, cross-talk between GC cells and HSCs by PDGFB/platelet derived growth factor receptor beta (PDGFRß) and the HGF/MET axis might represent potential therapeutic targets to treat GCLM.


Assuntos
Neoplasias Hepáticas , Podossomos , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Proteínas Proto-Oncogênicas c-sis/metabolismo , Células Estreladas do Fígado/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias Hepáticas/patologia , Transdução de Sinais , Microambiente Tumoral
8.
Mol Oncol ; 18(3): 620-640, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38098337

RESUMO

The small GTPase Rac1 (Ras-related C3 botulinum toxin substrate 1) has been implicated in cancer progression and in the poor prognosis of various types of tumors. Rac1 SUMOylation occurs during epithelial-mesenchymal transition (EMT), and it is required for tumor cell migration and invasion. Here we identify POTEE (POTE Ankyrin domain family member E) as a novel Rac1-SUMO1 effector involved in breast cancer malignancy that controls invadopodium formation through the activation of Rac1-SUMO1. POTEE activates Rac1 in the invadopodium by recruiting TRIO-GEF (triple functional domain protein), and it induces tumor cell proliferation and metastasis in vitro and in vivo. We found that the co-localization of POTEE with Rac1 is correlated with more aggressive breast cancer subtypes. Given its role in tumor dissemination, the leading cause of cancer-related deaths, POTEE could represent a potential therapeutic target for these types of cancer.


Assuntos
Neoplasias da Mama , Podossomos , Humanos , Feminino , Transdução de Sinais , Podossomos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Movimento Celular , Linhagem Celular Tumoral
9.
Cancer Lett ; 582: 216597, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38145655

RESUMO

Growing evidence has suggested that increased matrix stiffness can significantly strengthen the malignant characteristics of hepatocellular carcinoma (HCC) cells. However, whether and how increased matrix stiffness regulates the formation of invadopodia in HCC cells remain largely unknown. In the study, we developed different experimental systems in vitro and in vivo to explore the effects of matrix stiffness on the formation of invadopodia and its relevant molecular mechanism. Our results demonstrated that increased matrix stiffness remarkably augmented the migration and invasion abilities of HCC cells, upregulated the expressions of invadopodia-associated genes and enhanced the number of invadopodia. Two regulatory pathways contribute to matrix stiffness-driven invadopodia formation together in HCC cells, including direct triggering invadopodia formation through activating integrin ß1 or Piezo1/ FAK/Src/Arg/cortactin pathway, and indirect stimulating invadopodia formation through improving EGF production to activate EGFR/Src/Arg/cortactin pathway. Src was identified as the common hub molecule of two synergistic regulatory pathways. Simultaneously, activation of integrin ß1/RhoA/ROCK1/MLC2 and Piezo1/Ca2+/MLCK/MLC2 pathways mediate matrix stiffness-reinforced cell migration. This study uncovers a new mechanism by which mechanosensory pathway and biochemical signal pathway synergistically regulate the formation of invadopodia in HCC cells.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Podossomos , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Cortactina/metabolismo , Podossomos/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Integrina beta1/metabolismo , Matriz Extracelular/metabolismo , Linhagem Celular Tumoral , Invasividade Neoplásica , Quinases Associadas a rho/metabolismo
10.
Nat Cell Biol ; 25(12): 1787-1803, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37903910

RESUMO

Invadosomes and caveolae are mechanosensitive structures that are implicated in metastasis. Here, we describe a unique juxtaposition of caveola clusters and matrix degradative invadosomes at contact sites between the plasma membrane of cancer cells and constricting fibrils both in 2D and 3D type I collagen matrix environments. Preferential association between caveolae and straight segments of the fibrils, and between invadosomes and bent segments of the fibrils, was observed along with matrix remodelling. Caveola recruitment precedes and is required for invadosome formation and activity. Reciprocally, invadosome disruption results in the accumulation of fibril-associated caveolae. Moreover, caveolae and the collagen receptor ß1 integrin co-localize at contact sites with the fibrils, and integrins control caveola recruitment to fibrils. In turn, caveolae mediate the clearance of ß1 integrin and collagen uptake in an invadosome-dependent and collagen-cleavage-dependent mechanism. Our data reveal a reciprocal interplay between caveolae and invadosomes that coordinates adhesion to and proteolytic remodelling of confining fibrils to support tumour cell dissemination.


Assuntos
Podossomos , Humanos , Matriz Extracelular/metabolismo , Cavéolas/metabolismo , Integrina beta1/metabolismo , Colágeno Tipo I/metabolismo , Invasividade Neoplásica
11.
Cell Rep ; 42(10): 113302, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37862167

RESUMO

During metastasis, tumor cells invade through the basement membrane and intravasate into blood vessels and then extravasate into distant organs to establish metastases. Here, we report a critical role of a transmembrane serine protease fibroblast activation protein (FAP) in tumor metastasis. Expression of FAP and TWIST1, a metastasis driver, is significantly correlated in several types of human carcinomas, and FAP is required for TWIST1-induced breast cancer metastasis to the lung. Mechanistically, FAP is localized at invadopodia and required for invadopodia-mediated extracellular matrix degradation independent of its proteolytic activity. Live cell imaging shows that association of invadopodia precursors with FAP at the cell membrane promotes the stabilization and growth of invadopodia precursors into mature invadopodia. Together, our study identified FAP as a functional target of TWIST1 in driving tumor metastasis via promoting invadopodia-mediated matrix degradation and uncovered a proteolytic activity-independent role of FAP in stabilizing invadopodia precursors for maturation.


Assuntos
Neoplasias da Mama , Podossomos , Humanos , Feminino , Podossomos/metabolismo , Linhagem Celular Tumoral , Peptídeo Hidrolases/metabolismo , Invasividade Neoplásica/patologia , Neoplasias da Mama/patologia , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Fibroblastos/metabolismo , Matriz Extracelular/metabolismo
12.
Nat Commun ; 14(1): 5882, 2023 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-37735172

RESUMO

The activation and accumulation of lung fibroblasts resulting in aberrant deposition of extracellular matrix components, is a pathogenic hallmark of Idiopathic Pulmonary Fibrosis, a lethal and incurable disease. In this report, increased expression of TKS5, a scaffold protein essential for the formation of podosomes, was detected in the lung tissue of Idiopathic Pulmonary Fibrosis patients and bleomycin-treated mice. Τhe profibrotic milieu is found to induce TKS5 expression and the formation of prominent podosome rosettes in lung fibroblasts, that are retained ex vivo, culminating in increased extracellular matrix invasion. Tks5+/- mice are found resistant to bleomycin-induced pulmonary fibrosis, largely attributed to diminished podosome formation in fibroblasts and decreased extracellular matrix invasion. As computationally predicted, inhibition of src kinase is shown to potently attenuate podosome formation in lung fibroblasts and extracellular matrix invasion, and bleomycin-induced pulmonary fibrosis, suggesting pharmacological targeting of podosomes as a very promising therapeutic option in pulmonary fibrosis.


Assuntos
Fibrose Pulmonar Idiopática , Podossomos , Animais , Humanos , Camundongos , Proteínas Adaptadoras de Transporte Vesicular , Bleomicina , Matriz Extracelular , Fibroblastos , Fibrose Pulmonar Idiopática/induzido quimicamente , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo
13.
Sci Signal ; 16(803): eadh4210, 2023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37725664

RESUMO

Alternative splicing regulates gene expression and functional diversity and is often dysregulated in human cancers. Here, we discovered that the long noncoding RNA (lncRNA) MIR99AHG regulated alternative splicing to alter the activity of a chromatin remodeler and promote metastatic behaviors in colorectal cancer (CRC). MIR99AHG was abundant in invasive CRC cells and metastatic tumors from patients and promoted motility and invasion in cultured CRC cells. MIR99AHG bound to and stabilized the RNA splicing factor PTBP1, and this complex increased cassette exon inclusion in the mRNA encoding the chromatin remodeling gene SMARCA1. Specifically, MIR99AHG altered the nature of PTBP1 binding to the splice sites on intron 12 of SMARCA1 pre-mRNA, thereby triggering a splicing switch from skipping to including exon 13 to produce the long isoform, SMARCA1-L. SMARCA1, but not SMARCA1-L, suppressed invadopodia formation, cell migration, and invasion. Analysis of CRC samples revealed that the abundance of MIR99AHG transcript positively correlated with that of SMARCA1-L mRNA and PTBP1 protein and with poor prognosis in patients with CRC. Furthermore, TGF-ß1 secretion from cancer-associated fibroblasts increased MIR99AHG expression in CRC cells. Our findings identify an lncRNA that is induced by cues from the tumor microenvironment and that interacts with PTBP1 to regulate alternative splicing, potentially providing a therapeutic target and predictive biomarker for metastatic CRC.


Assuntos
Neoplasias Colorretais , Podossomos , RNA Longo não Codificante , Humanos , Processamento Alternativo , Cromatina , Neoplasias Colorretais/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Splicing de RNA , RNA Longo não Codificante/genética , Microambiente Tumoral
14.
Eur J Cell Biol ; 102(4): 151356, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37625234

RESUMO

Podosomes have been known for several decades as micron-sized, F-actin-rich structures that play a pivotal role in cell migration and invasion, as they are able to mediate both cell-matrix attachment as well as extracellular matrix degradation. Particularly in monocytic cells, podosomes have been shown to fulfill a variety of additional functions such as sensing of substrate rigidity and topography, or cell-cell fusion. Increasing evidence now points to the involvement of podosome-like structures also during phagocytosis by immune cells such as macrophages, dendritic cells, and neutrophils. Here, we compare the different cell models and experimental set ups where "phagocytic podosomes" have been described. We also discuss the composition and architecture of these structures, their potential involvement in mechanosensing and particle disruption, as well as the pros and cons for addressing them as bona fide podosomes.


Assuntos
Podossomos , Podossomos/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Macrófagos/metabolismo , Fagocitose
15.
Exp Cell Res ; 431(1): 113743, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37591452

RESUMO

A critical challenge in the treatment of glioblastoma (GBM) is its highly invasive nature which promotes cell migration throughout the brain and hinders surgical resection and effective drug delivery. GBM cells demonstrate augmented invasive capabilities following exposure to the current gold standard treatment of radiotherapy (RT) and concomitant and adjuvant temozolomide (TMZ), resulting in rapid disease recurrence. Elucidating the mechanisms employed by post-treatment invasive GBM cells is critical to the development of more effective therapies. In this study, we utilized a Nanostring® Cancer Progression gene expression panel to identify candidate genes that may be involved in enhanced GBM cell invasion after treatment with clinically relevant doses of RT/TMZ. Our findings identified thrombospondin-1 (THBS1) as a pro-invasive gene that is upregulated in these cells. Immunofluorescence staining revealed that THBS1 localised within functional matrix-degrading invadopodia that formed on the surface of GBM cells. Furthermore, overexpression of THBS1 resulted in enhanced GBM cell migration and secretion of MMP-2, which was reduced with silencing of THBS1. The preliminary data demonstrates that THBS1 is associated with invadopodia in GBM cells and is likely involved in the invadopodia-mediated invasive process in GBM cells exposed to RT/TMZ treatment. Therapeutic inhibition of THBS1-mediated invadopodia activity, which facilitates GBM cell invasion, should be further investigated as a treatment for GBM.


Assuntos
Glioblastoma , Podossomos , Humanos , Glioblastoma/genética , Glioblastoma/terapia , Recidiva Local de Neoplasia , Temozolomida/farmacologia , Encéfalo
16.
Cell Struct Funct ; 48(2): 161-174, 2023 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-37482421

RESUMO

Invadopodia are protrusive structures that mediate the extracellular matrix (ECM) degradation required for tumor invasion and metastasis. Rho small GTPases regulate invadopodia formation, but the molecular mechanisms of how Rho small GTPase activities are regulated at the invadopodia remain unclear. Here we have identified FilGAP, a GTPase-activating protein (GAP) for Rac1, as a negative regulator of invadopodia formation in tumor cells. Depletion of FilGAP in breast cancer cells increased ECM degradation and conversely, overexpression of FilGAP decreased it. FilGAP depletion promoted the formation of invadopodia with ECM degradation. In addition, FilGAP depletion and Rac1 overexpression increased the emergence of invadopodia induced by epidermal growth factor, whereas FilGAP overexpression suppressed it. Overexpression of GAP-deficient FilGAP mutant enhanced invadopodia emergence as well as FilGAP depletion. The pleckstrin-homology (PH) domain of FilGAP binds phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2], which is distributed on membranes of the invadopodia. FilGAP localized to invadopodia in breast cancer cells on the ECM, but FilGAP mutant lacking PI(3,4)P2-binding showed low localization. Similarly, the decrease of PI(3,4)P2 production reduced the FilGAP localization. Our results suggest that FilGAP localizes to invadopodia through its PH domain binding to PI(3,4)P2 and down-regulates invadopodia formation by inactivating Rac1, inhibiting ECM degradation in invasive tumor cells.Key words: invadopodia, breast carcinoma, Rac1, FilGAP, PI(3,4)P2.


Assuntos
Neoplasias da Mama , Podossomos , Humanos , Feminino , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Podossomos/metabolismo , Podossomos/patologia , Proteínas rho de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia
17.
Cell Rep ; 42(8): 112893, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37516960

RESUMO

Invadopodia are extracellular matrix (ECM) degrading structures, which promote cancer cell invasion. The nucleus is increasingly viewed as a mechanosensory organelle that determines migratory strategies. However, how the nucleus crosstalks with invadopodia is little known. Here, we report that the oncogenic septin 9 isoform 1 (SEPT9_i1) is a component of breast cancer invadopodia. SEPT9_i1 depletion diminishes invadopodium formation and the clustering of the invadopodium precursor components TKS5 and cortactin. This phenotype is characterized by deformed nuclei and nuclear envelopes with folds and grooves. We show that SEPT9_i1 localizes to the nuclear envelope and juxtanuclear invadopodia. Moreover, exogenous lamin A rescues nuclear morphology and juxtanuclear TKS5 clusters. Importantly, SEPT9_i1 is required for the amplification of juxtanuclear invadopodia, which is induced by the epidermal growth factor. We posit that nuclei of low deformability favor the formation of juxtanuclear invadopodia in a SEPT9_i1-dependent manner, which functions as a tunable mechanism for overcoming ECM impenetrability.


Assuntos
Neoplasias da Mama , Podossomos , Humanos , Feminino , Septinas/metabolismo , Podossomos/metabolismo , Isoformas de Proteínas/metabolismo , Neoplasias da Mama/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Linhagem Celular Tumoral , Invasividade Neoplásica
18.
Methods Mol Biol ; 2692: 79-90, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37365462

RESUMO

Macrophages are motile, morphologically plastic cells that undergo substantial cytoskeletal remodeling to facilitate their roles in innate and adaptive immunity. Macrophages are adept at producing a variety of specialized actin-driven structures and processes including the formation of podosomes and the ability to engulf particles through phagocytosis and sample large amounts of extracellular fluid via micropinocytosis. Here, we describe techniques for immunostaining proteins and transfecting macrophages with plasmids for use with either fixed or live cell imaging. Furthermore, we discuss the use of spinning-disk super-resolution using optical reassignment to generate sub-diffraction limited structures using this type of confocal microscope.


Assuntos
Citoesqueleto de Actina , Podossomos , Citoesqueleto de Actina/metabolismo , Microscopia Confocal/métodos , Macrófagos/metabolismo , Actinas/metabolismo , Podossomos/metabolismo
19.
J Cell Sci ; 136(10)2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37226883

RESUMO

Rac (herein referring to the Rac family) and Cdc42 are Rho GTPases that regulate the formation of lamellipoda and filopodia, and are therefore crucial in processes such as cell migration. Relocation-based biosensors for Rac and Cdc42 have not been characterized well in terms of their specificity or affinity. In this study, we identify relocation sensor candidates for both Rac and Cdc42. We compared their (1) ability to bind the constitutively active Rho GTPases, (2) specificity for Rac and Cdc42, and (3) relocation efficiency in cell-based assays. Subsequently, the relocation efficiency was improved by a multi-domain approach. For Rac1, we found a sensor candidate with low relocation efficiency. For Cdc42, we found several sensors with sufficient relocation efficiency and specificity. These optimized sensors enable the wider application of Rho GTPase relocation sensors, which was showcased by the detection of local endogenous Cdc42 activity at assembling invadopodia. Moreover, we tested several fluorescent proteins and HaloTag for their influence on the recruitment efficiency of the Rho location sensor, to find optimal conditions for a multiplexing experiment. This characterization and optimization of relocation sensors will broaden their application and acceptance.


Assuntos
Podossomos , Proteínas rho de Ligação ao GTP , Movimento Celular , Pseudópodes
20.
Int J Cancer ; 153(6): 1287-1299, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37212571

RESUMO

In a previous study, our research group observed that estrogen promotes the metastasis of non-small cell lung cancer (NSCLC) through the estrogen receptor ß (ERß). Invadopodia are key structures involved in tumor metastasis. However, it is unclear whether ERß is involved in the promotion of NSCLC metastasis through invadopodia. In our study, we used scanning electron microscopy to observe the formation of invadopodia following the overexpression of ERß and treatment with E2. In vitro experiments using multiple NSCLC cell lines demonstrated that ERß can increase the formation of invadopodia and cell invasion. Mechanistic studies revealed that ERß can upregulate the expression of ICAM1 by directly binding to estrogen-responsive elements (EREs) located on the ICAM1 promoter, which in turn can enhance the phosphorylation of Src/cortactin. We also confirmed these findings in vivo using an orthotopic lung transplantation mouse model, which validated the results obtained from the in vitro experiments. Finally, we examined the expressions of ERß and ICAM1 using immunohistochemistry in both NSCLC tissue and paired metastatic lymph nodes. The results confirmed that ERß promotes the formation of invadopodia in NSCLC cells through the ICAM1/p-Src/p-Cortactin signaling pathway.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Podossomos , Animais , Camundongos , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Cortactina/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Neoplasias Pulmonares/patologia , Invasividade Neoplásica/patologia , Podossomos/metabolismo , Podossomos/patologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...