Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Elife ; 132024 May 07.
Article in English | MEDLINE | ID: mdl-38712822

ABSTRACT

Pancreatic ductal adenocarcinoma carries a dismal prognosis, with high rates of metastasis and few treatment options. Hyperactivation of KRAS in almost all tumours drives RAC1 activation, conferring enhanced migratory and proliferative capacity as well as macropinocytosis. Macropinocytosis is well understood as a nutrient scavenging mechanism, but little is known about its functions in trafficking of signaling receptors. We find that CYRI-B is highly expressed in pancreatic tumours in a mouse model of KRAS and p53-driven pancreatic cancer. Deletion of Cyrib (the gene encoding CYRI-B protein) accelerates tumourigenesis, leading to enhanced ERK and JNK-induced proliferation in precancerous lesions, indicating a potential role as a buffer of RAC1 hyperactivation in early stages. However, as disease progresses, loss of CYRI-B inhibits metastasis. CYRI-B depleted tumour cells show reduced chemotactic responses to lysophosphatidic acid, a major driver of tumour spread, due to impaired macropinocytic uptake of the lysophosphatidic acid receptor-1. Overall, we implicate CYRI-B as a mediator of growth and signaling in pancreatic cancer, providing new insights into pathways controlling metastasis.

2.
J Pathol ; 262(4): 454-466, 2024 04.
Article in English | MEDLINE | ID: mdl-38229581

ABSTRACT

MAP4K4 is a serine/threonine kinase of the STE20 family involved in the regulation of actin cytoskeleton dynamics and cell motility. It has been proposed as a target of angiogenesis and inhibitors show potential in cardioprotection. MAP4K4 also mediates cell invasion in vitro, is overexpressed in various types of cancer, and is associated with poor patient prognosis. Recently, MAP4K4 has been shown to be overexpressed in pancreatic cancer, but its role in tumour initiation, progression, and metastasis is unknown. Here, using the KrasG12D Trp53R172H Pdx1-Cre (KPC) mouse model of pancreatic ductal adenocarcinoma (PDAC), we show that deletion of Map4k4 drives tumour initiation and progression. Moreover, we report that the acceleration of tumour onset is also associated with an overactivation of ERK and AKT, two major downstream effectors of KRAS, in vitro and in vivo. In contrast to the accelerated tumour onset caused by loss of MAP4K4, we observed a reduction in metastatic burden with both the KPC model and in an intraperitoneal transplant assay indicating a major role of MAP4K4 in metastatic seeding. In summary, our study sheds light on the dichotomous role of MAP4K4 in the initiation of PDAC onset, progression, and metastatic dissemination. It also identifies MAP4K4 as a possible druggable target against pancreatic cancer spread, but with the caveat that targeting MAP4K4 might accelerate early tumorigenesis. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Humans , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , MAP Kinase Signaling System , Cell Line, Tumor , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Serine , Intracellular Signaling Peptides and Proteins/metabolism
3.
J Cell Sci ; 135(24)2022 12 15.
Article in English | MEDLINE | ID: mdl-36546396

ABSTRACT

Pancreatic cancer is a deadly and highly metastatic disease, although how metastatic lesions establish is not fully understood. A key feature of pancreatic tumours is extensive fibrosis and deposition of extracellular matrix (ECM). While pancreatic cancer cells are programmed by stimuli derived from a stiff ECM, metastasis requires loss of attachment and adaptation to a softer microenvironment at distant sites. Growing evidence suggests that stiff ECM influences pancreatic cancer cell behaviour. Here, we argue that this influence is reversible and that pancreatic cancer cells can be reprogrammed upon sensing soft substrates. Using engineered polyacrylamide hydrogels with tuneable mechanical properties, we show that collagen VI is specifically upregulated in pancreatic cancer cells on soft substrates, due to a lack of integrin engagement. Furthermore, the expression of collagen VI is inversely correlated with mechanosensing and activity of YAP (also known as YAP1), which might be due to a direct or indirect effect on transcription of genes encoding collagen VI. Collagen VI supports migration in vitro and metastasis formation in vivo. Metastatic nodules formed by pancreatic cancer cells lacking Col6a1 display stromal cell-derived collagen VI deposition, suggesting that collagen VI derived from either cancer cells or the stroma is an essential component of the metastatic niche. This article has an associated First Person interview with Vasileios Papalazarou, joint first author of the paper.


Subject(s)
Collagen , Pancreatic Neoplasms , Humans , Collagen/metabolism , Extracellular Matrix/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Integrins/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
4.
Nat Metab ; 2(1): 62-80, 2020 01.
Article in English | MEDLINE | ID: mdl-32694686

ABSTRACT

Pancreatic ductal adenocarcinoma is particularly metastatic, with dismal survival rates and few treatment options. Stiff fibrotic stroma is a hallmark of pancreatic tumours, but how stromal mechanosensing affects metastasis is still unclear. Here, we show that mechanical changes in the pancreatic cancer cell environment affect not only adhesion and migration, but also ATP/ADP and ATP/AMP ratios. Unbiased metabolomic analysis reveals that the creatine-phosphagen ATP-recycling system is a major mechanosensitive target. This system depends on arginine flux through the urea cycle, which is reflected by the increased incorporation of carbon and nitrogen from L-arginine into creatine and phosphocreatine on stiff matrix. We identify that CKB is a mechanosensitive transcriptional target of YAP, and thus it increases phosphocreatine production. We further demonstrate that the creatine-phosphagen system has a role in invasive migration, chemotaxis and liver metastasis of cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Phosphocreatine/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Arginine Kinase/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/pathology , Cells, Cultured , Creatine Kinase/metabolism , Extracellular Matrix/pathology , Humans , Metabolome , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology
5.
Dev Cell ; 51(4): 431-445.e7, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31668663

ABSTRACT

Pancreatic ductal adenocarcinoma is one of the most invasive and metastatic cancers and has a dismal 5-year survival rate. We show that N-WASP drives pancreatic cancer metastasis, with roles in both chemotaxis and matrix remodeling. lysophosphatidic acid, a signaling lipid abundant in blood and ascites fluid, is both a mitogen and chemoattractant for cancer cells. Pancreatic cancer cells break lysophosphatidic acid down as they respond to it, setting up a self-generated gradient driving tumor egress. N-WASP-depleted cells do not recognize lysophosphatidic acid gradients, leading to altered RhoA activation, decreased contractility and traction forces, and reduced metastasis. We describe a signaling loop whereby N-WASP and the endocytic adapter SNX18 promote lysophosphatidic acid-induced RhoA-mediated contractility and force generation by controlling lysophosphatidic acid receptor recycling and preventing degradation. This chemotactic loop drives collagen remodeling, tumor invasion, and metastasis and could be an important target against pancreatic cancer spread.


Subject(s)
Lysophospholipids/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptors, Lysophosphatidic Acid/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement/physiology , Chemotaxis , Female , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Transport , Rats , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/isolation & purification , Signal Transduction , Sorting Nexins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , rhoA GTP-Binding Protein/metabolism
6.
Sci Signal ; 12(567)2019 02 05.
Article in English | MEDLINE | ID: mdl-30723174

ABSTRACT

Intratumoral hypoxia causes the formation of dysfunctional blood vessels, which contribute to tumor metastasis and reduce the efficacy of therapeutic treatments. Blood vessels are embedded in the tumor stroma of which cancer-associated fibroblasts (CAFs) constitute a prominent cellular component. We found that hypoxic human mammary CAFs promoted angiogenesis in CAF-endothelial cell cocultures in vitro. Mass spectrometry-based proteomic analysis of the CAF secretome unraveled that hypoxic CAFs contributed to blood vessel abnormalities by altering their secretion of various pro- and anti-angiogenic factors. Hypoxia induced pronounced remodeling of the CAF proteome, including proteins that have not been previously related to this process. Among those, the uncharacterized protein NCBP2-AS2 that we renamed HIAR (hypoxia-induced angiogenesis regulator) was the protein most increased in abundance in hypoxic CAFs. Silencing of HIAR abrogated the pro-angiogenic and pro-migratory function of hypoxic CAFs by decreasing secretion of the pro-angiogenic factor VEGFA and consequently reducing VEGF/VEGFR downstream signaling in the endothelial cells. Our study has identified a regulator of angiogenesis and provides a map of hypoxia-induced molecular alterations in mammary CAFs.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hypoxia , Neovascularization, Pathologic/genetics , Proteome/metabolism , Proteomics/methods , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics
7.
J Cell Biol ; 207(4): 517-33, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25422375

ABSTRACT

Accumulation of type I collagen fibrils in tumors is associated with an increased risk of metastasis. Invadosomes are F-actin structures able to degrade the extracellular matrix. We previously found that collagen I fibrils induced the formation of peculiar linear invadosomes in an unexpected integrin-independent manner. Here, we show that Discoidin Domain Receptor 1 (DDR1), a collagen receptor overexpressed in cancer, colocalizes with linear invadosomes in tumor cells and is required for their formation and matrix degradation ability. Unexpectedly, DDR1 kinase activity is not required for invadosome formation or activity, nor is Src tyrosine kinase. We show that the RhoGTPase Cdc42 is activated on collagen in a DDR1-dependent manner. Cdc42 and its specific guanine nucleotide-exchange factor (GEF), Tuba, localize to linear invadosomes, and both are required for linear invadosome formation. Finally, DDR1 depletion blocked cell invasion in a collagen gel. Altogether, our data uncover an important role for DDR1, acting through Tuba and Cdc42, in proteolysis-based cell invasion in a collagen-rich environment.


Subject(s)
Collagen Type I/metabolism , Cytoskeletal Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , cdc42 GTP-Binding Protein/metabolism , Actin Cytoskeleton , Actins/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cell Line, Tumor , Collagenases/metabolism , Dipeptides/pharmacology , Discoidin Domain Receptor 1 , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinase Inhibitors/pharmacology , Neoplasm Invasiveness/genetics , RNA Interference , RNA, Small Interfering , Receptor Protein-Tyrosine Kinases/genetics
8.
Cell Adh Migr ; 8(3): 280-92, 2014.
Article in English | MEDLINE | ID: mdl-24840388

ABSTRACT

Invadosomes are actin-based structures involved in extracellular-matrix degradation. Invadosomes, either known as podosomes or invadopodia, are found in an increasing number of cell types. Moreover, their overall organization and molecular composition may vary from one cell type to the other. Some are constitutive such as podosomes in hematopoietic cells whereas others are inducible. However, they share the same feature, their ability to interact and to degrade the extracellular matrix. Based on the literature and our own experiments, the aim of this study was to establish a minimal molecular definition of active invadosomes. We first highlighted that Cdc42 is the key RhoGTPase involved in invadosome formation in all described models. Using different cellular models, such as NIH-3T3, HeLa, and endothelial cells, we demonstrated that overexpression of an active form of Cdc42 is sufficient to form invadosome actin cores. Therefore, active Cdc42 must be considered not only as an inducer of filopodia, but also as an inducer of invadosomes. Depending on the expression level of Tks5, these Cdc42-dependent actin cores were endowed or not with a proteolytic activity. In fact, Tks5 overexpression rescued this activity in Tks5 low expressing cells. We thus described the adaptor protein Tks5 as a major actor of the invadosome degradation function. Surprisingly, we found that Src kinases are not always required for invadosome formation and function. These data suggest that even if Src family members are the principal kinases involved in the majority of invadosomes, it cannot be considered as a common element for all invadosome structures. We thus define a minimal and universal molecular signature of invadosome that includes Cdc42 activity and Tks5 presence in order to drive the actin machinery and the proteolytic activity of these invasive structures.


Subject(s)
Actin Cytoskeleton/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cell Movement/physiology , Extracellular Matrix/metabolism , Phosphoproteins/metabolism , cdc42 GTP-Binding Protein/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Humans , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Phosphate-Binding Proteins , Phosphoproteins/genetics
9.
Gastroenterology ; 146(5): 1386-96.e1-17, 2014 May.
Article in English | MEDLINE | ID: mdl-24462734

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is often lethal because it is highly invasive and metastasizes rapidly. The actin-bundling protein fascin has been identified as a biomarker of invasive and advanced PDAC and regulates cell migration and invasion in vitro. We investigated fascin expression and its role in PDAC progression in mice. METHODS: We used KRas(G12D) p53(R172H) Pdx1-Cre (KPC) mice to investigate the effects of fascin deficiency on development of pancreatic intraepithelial neoplasia (PanIn), PDAC, and metastasis. We measured levels of fascin in PDAC cell lines and 122 human resected PDAC samples, along with normal ductal and acinar tissues; we associated levels with patient outcomes. RESULTS: Pancreatic ducts and acini from control mice and early-stage PanINs from KPC mice were negative for fascin, but approximately 6% of PanIN3 and 100% of PDAC expressed fascin. Fascin-deficient KRas(G12D) p53(R172H) Pdx1-Cre mice had longer survival times, delayed onset of PDAC, and a lower PDAC tumor burdens than KPC mice; loss of fascin did not affect invasion of PDAC into bowel or peritoneum in mice. Levels of slug and fascin correlated in PDAC cells; slug was found to regulate transcription of Fascin along with the epithelial-mesenchymal transition. In PDAC cell lines and cells from mice, fascin concentrated in filopodia and was required for their assembly and turnover. Fascin promoted intercalation of filopodia into mesothelial cell layers and cell invasion. Nearly all human PDAC samples expressed fascin, and higher fascin histoscores correlated with poor outcomes, vascular invasion, and time to recurrence. CONCLUSIONS: The actin-bundling protein fascin is regulated by slug and involved in late-stage PanIN and PDAC formation in mice. Fascin appears to promote formation of filopodia and invasive activities of PDAC cells. Its levels in human PDAC correlate with outcomes and time to recurrence, indicating it might be a marker or therapeutic target for pancreatic cancer.


Subject(s)
Carcinoma in Situ/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carrier Proteins/metabolism , Microfilament Proteins/metabolism , Pancreatic Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Carrier Proteins/genetics , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition , Humans , Mice , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Neoplasm Staging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Pseudopodia/metabolism , RNA Interference , Snail Family Transcription Factors , Survival Analysis , Time Factors , Transcription Factors/genetics , Transfection
10.
Biol Cell ; 105(1): 46-57, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23106484

ABSTRACT

BACKGROUND INFORMATION: Podosomes are actin-based structures involved in cell adhesion, migration, invasion and extracellular matrix degradation. They have been described in large vessel endothelial cells, but nothing is known concerning microvascular endothelial cells. Here, we focussed on liver sinusoidal endothelial cells (LSECs), fenestrated microvascular cells that play major roles in liver physiology. Liver fibrosis induces a dedifferentiation of LSECs leading notably to a loss of fenestrae. Because liver fibrosis is associated with increased matrix stiffness, and because substrate stiffness is known to regulate the actin cytoskeleton, we investigated the impact of matrix rigidity on podosome structures in LSECs. RESULTS: Using primary LSECs, we demonstrated that microvascular endothelial cells are able to form constitutive podosomes. Podosome presence in LSECs was independent of cytokines such as transforming growth factor-ß or vascular endothelial growth factor, but could be modulated by matrix stiffness. As expected, LSECs lost their differentiated phenotype during cell culture, which was paralleled by a loss of podosomes. LSECs however retained the capacity to form active podosomes following detachment/reseeding or actin-destabilising drug treatments. Finally, constitutive podosomes were also found in primary microvascular endothelial cells from other organs. CONCLUSIONS: Our results show that microvascular endothelial cells are able to form podosomes without specific stimulation. Our data suggest that the major determinant of podosome induction in these cells is substrate rigidity.


Subject(s)
Actin Cytoskeleton/metabolism , Endothelial Cells/cytology , Extracellular Matrix/metabolism , Liver/metabolism , Microvessels/metabolism , Signal Transduction/physiology , Cell Adhesion/physiology , Humans , Liver/blood supply , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
11.
J Cell Sci ; 125(Pt 18): 4278-87, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22718352

ABSTRACT

The endoplasmic reticulum (ER) is an organelle specialized for the folding and assembly of secretory and transmembrane proteins. ER homeostasis is often perturbed in tumor cells because of dramatic changes in the microenvironment of solid tumors, thereby leading to the activation of an adaptive mechanism named the unfolded protein response (UPR). The activation of the UPR sensor IRE1α has been described to play an important role in tumor progression. However, the molecular events associated with this phenotype remain poorly characterized. In the present study, we examined the effects of IRE1α signaling on the adaptation of glioma cells to their microenvironment. We show that the characteristics of U87 cell migration are modified under conditions where IRE1α activity is impaired (DN_IRE1). This is linked to increased stress fiber formation and enhanced RhoA activity. Gene expression profiling also revealed that loss of functional IRE1α signaling mostly resulted in the upregulation of genes encoding extracellular matrix proteins. Among these genes, Sparc, whose mRNA is a direct target of IRE1α endoribonuclease activity, was in part responsible for the phenotypic changes associated with IRE1α inactivation. Hence, our data demonstrate that IRE1α is a key regulator of SPARC expression in vitro in a glioma model. Our results also further support the crucial contribution of IRE1α to tumor growth, infiltration and invasion and extend the paradigm of secretome control in tumor microenvironment conditioning.


Subject(s)
Autocrine Communication , Brain Neoplasms/pathology , Cell Movement , Endoribonucleases/metabolism , Glioma/pathology , Osteonectin/metabolism , Protein Serine-Threonine Kinases/metabolism , Actin Cytoskeleton/metabolism , Autocrine Communication/genetics , Brain Neoplasms/genetics , Cell Adhesion/genetics , Cell Movement/genetics , Cell Proliferation , Down-Regulation/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , Models, Biological , Osteonectin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Spheroids, Cellular/pathology , Tumor Cells, Cultured , rhoA GTP-Binding Protein/metabolism
12.
Mol Biol Cell ; 23(2): 297-309, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22114353

ABSTRACT

Invadosomes are F-actin structures capable of degrading the matrix through the activation of matrix metalloproteases. As fibrillar type I collagen promotes pro-matrix metalloproteinase 2 activation by membrane type 1 matrix metalloproteinase, we aimed at investigating the functional relationships between collagen I organization and invadosome induction. We found that fibrillar collagen I induced linear F-actin structures, distributed along the fibrils, on endothelial cells, macrophages, fibroblasts, and tumor cells. These structures share features with conventional invadosomes, as they express cortactin and N-WASP and accumulate the scaffold protein Tks5, which proved essential for their formation. On the basis of their ability to degrade extracellular matrix elements and their original architecture, we named these structures "linear invadosomes." Interestingly, podosomes or invadopodia were replaced by linear invadosomes upon contact of the cells with fibrillar collagen I. However, linear invadosomes clearly differ from classical invadosomes, as they do not contain paxillin, vinculin, and ß1/ß3 integrins. Using knockout mouse embryonic fibroblasts and RGD peptide, we demonstrate that linear invadosome formation and activity are independent of ß1 and ß3 integrins. Finally, linear invadosomes also formed in a three-dimensional collagen matrix. This study demonstrates that fibrillar collagen I is the physiological inducer of a novel class of invadosomes.


Subject(s)
Collagen Type I/metabolism , Collagen Type I/ultrastructure , Extracellular Matrix/metabolism , Actins/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cattle , Cell Line , Cricetinae , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Integrin beta1/metabolism , Integrin beta3/metabolism , Mice , Mice, Knockout , Oligopeptides/pharmacology , Swine , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism
13.
Eur J Cell Biol ; 90(2-3): 100-7, 2011.
Article in English | MEDLINE | ID: mdl-20605056

ABSTRACT

Podosomes and invadopodia are highly dynamic, actin-rich adhesion structures and represent the two founding members of the invadosome family. Podosomes form spontaneously in cells of the myelomonocytic lineage but a plethora of other cells are endowed with this capacity, under appropriate stimulation, such as a soluble factor, matrix receptor, or cell stress. Related structures called invadopodia are detected in some cancer cells or appear on cells upon oncogenic transformation. In contrast to other cell adhesion devices, invadosomes harbour metalloproteases which degrade components of the extracellular matrix. Because of this distinctive feature, invadosomes have been systematically linked with invasion processes. However, it now appears that these intriguing structures are endowed with other functions and are therefore expected to contribute to a wider range of biological processes. The invadosome field has been progressing for thirty years, expanding exponentially during the last decade, where tremendous advances have been made regarding the molecular mechanism underlying their formation, dynamics and function. Invadosomes are involved in human diseases but the causative link remains to be established. To this end, 3D analysis of invadosomes is now being actively developed in ex vivo and in vivo models to demonstrate their occurrence and establish their role in pathological and physiological processes.


Subject(s)
Cell Membrane Structures/physiology , Cell Surface Extensions/physiology , Cytoskeleton/physiology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...