Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Protoc ; 15(8): 2413-2442, 2020 08.
Article in English | MEDLINE | ID: mdl-32690957

ABSTRACT

Cancer invasion and metastasis are challenging to study in vivo since they occur deep inside the body over extended time periods. Organotypic 3D culture of fresh tumor tissue enables convenient real-time imaging, genetic and microenvironmental manipulation and molecular analysis. Here, we provide detailed protocols to isolate and culture heterogenous organoids from murine and human primary and metastatic site tumors. The time required to isolate organoids can vary based on the tissue and organ type but typically takes <7 h. We describe a suite of assays that model specific aspects of metastasis, including proliferation, survival, invasion, dissemination and colony formation. We also specify comprehensive protocols for downstream applications of organotypic cultures that will allow users to (i) test the role of specific genes in regulating various cellular processes, (ii) distinguish the contributions of several microenvironmental factors and (iii) test the effects of novel therapeutics.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Tissue Culture Techniques/methods , Animals , Humans , Mice , Neoplasm Metastasis
2.
Bioinformatics ; 36(10): 3292-3294, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32091578

ABSTRACT

SUMMARY: Organoid model systems recapitulate key features of mammalian tissues and enable high throughput experiments. However, the impact of these experiments may be limited by manual, non-standardized, static or qualitative phenotypic analysis. OrgDyn is an open-source and modular pipeline to quantify organoid shape dynamics using a combination of feature- and model-based approaches on time series of 2D organoid contour images. Our pipeline consists of (i) geometrical and signal processing feature extraction, (ii) dimensionality reduction to differentiate dynamical paths, (iii) time series clustering to identify coherent groups of organoids and (iv) dynamical modeling using point distribution models to explain temporal shape variation. OrgDyn can characterize, cluster and model differences among unique dynamical paths that define diverse final shapes, thus enabling quantitative analysis of the molecular basis of tissue development and disease. AVAILABILITY AND IMPLEMENTATION: https://github.com/zakih/organoidDynamics (BSD 3-Clause License). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Organoids , Software , Animals , Cluster Analysis
3.
Cancer Res ; 80(2): 204-218, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31676574

ABSTRACT

Dissemination is an essential early step in metastasis but its molecular basis remains incompletely understood. To define the essential targetable effectors of this process, we developed a 3D mammary epithelial culture model, in which dissemination is induced by overexpression of the transcription factor Twist1. Transcriptomic analysis and ChIP-PCR together demonstrated that protein kinase D1 (Prkd1) is a direct transcriptional target of Twist1 and is not expressed in the normal mammary epithelium. Pharmacologic and genetic inhibition of Prkd1 in the Twist1-induced dissemination model demonstrated that Prkd1 was required for cells to initiate extracellular matrix (ECM)-directed protrusions, release from the epithelium, and migrate through the ECM. Antibody-based protein profiling revealed that Prkd1 induced broad phosphorylation changes, including an inactivating phosphorylation of ß-catenin and two microtubule depolymerizing phosphorylations of Tau, potentially explaining the release of cell-cell contacts and persistent activation of Prkd1. In patients with breast cancer, TWIST1 and PRKD1 expression correlated with metastatic recurrence, particularly in basal breast cancer. Prkd1 knockdown was sufficient to block dissemination of both murine and human mammary tumor organoids. Finally, Prkd1 knockdown in vivo blocked primary tumor invasion and distant metastasis in a mouse model of basal breast cancer. Collectively, these data identify Prkd1 as a novel and targetable signaling node downstream of Twist1 that is required for epithelial invasion and dissemination. SIGNIFICANCE: Twist1 is a known regulator of metastatic cell behaviors but not directly targetable. This study provides a molecular explanation for how Twist1-induced dissemination works and demonstrates that it can be targeted. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/2/204/F1.large.jpg.


Subject(s)
Breast Neoplasms/genetics , Mammary Neoplasms, Experimental/genetics , Nuclear Proteins/metabolism , Protein Kinase C/genetics , Twist-Related Protein 1/metabolism , Animals , Breast/cytology , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Datasets as Topic , Epithelial Cells/cytology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Epithelium/pathology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Microtubules/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Primary Cell Culture , RNA-Seq , Signal Transduction/genetics
4.
J Vis Exp ; (147)2019 05 10.
Article in English | MEDLINE | ID: mdl-31132065

ABSTRACT

Intestinal organoid cultures provide a unique opportunity to investigate intestinal stem cell and crypt biology in vitro, although efficient approaches to manipulate gene expression in organoids have made limited progress in this arena. While CRISPR/Cas9 technology allows for precise genome editing of cells for organoid generation, this strategy requires extensive selection and screening by sequence analysis, which is both time-consuming and costly. Here, we provide a detailed protocol for efficient viral transduction of intestinal organoids. This approach is rapid and highly efficient, thus decreasing the time and expense inherent in CRISPR/Cas9 technology. We also present a protocol to generate frozen sections from intact organoid cultures for further analysis with immunohistochemical or immunofluorescent staining, which can be used to confirm gene expression or silencing. After successful transduction of viral vectors for gene expression or silencing is achieved, intestinal stem cell and crypt function can be rapidly assessed. Although most organoid studies employ in vitro assays, organoids can also be delivered to mice for in vivo functional analyses. Moreover, our approaches are advantageous for predicting therapeutic responses to drugs because currently available therapies generally function by modulating gene expression or protein function rather than altering the genome.


Subject(s)
Frozen Sections , Genetic Engineering/methods , Genetic Vectors/metabolism , Intestines/physiology , Magnetite Nanoparticles/chemistry , Organoids/metabolism , Transduction, Genetic , Animals , DNA/genetics , Gene Editing/methods , HEK293 Cells , Humans , Magnetic Fields , Mice
5.
Eur J Cell Biol ; 97(8): 533-545, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30287085

ABSTRACT

Actin subunits assemble into actin filaments whose dynamics and three-dimensional architectures are further regulated by a variety of cellular factors to establish the functional actin cytoskeleton. The C-glucosidic ellagitannin vescalagin and its simpler analogue vescalin, affect both the dynamics and the ultrastructure of the actin cytoskeleton by directly binding to F-actin. Herein, we show that in vitro, the two compounds induce the formation of distinct F-actin networks characterized by different superstructures and dynamics. In living mature osteoclasts, highly specialized bone-degrading cells that constantly remodel their cytoskeleton, vescalagin and vescalin alter actin dynamics at podosomes and compromise the integrity of the podosome belt that forms the bone-degrading apparatus. Both compounds target the bone-resorbing activity at concentrations that preserve osteoclastic maturation and survival and with no detectable impact on the behaviour of bone-forming osteoblastic cells. This anti-osteoclastic activity of vescalagin and vescalin reveals the potential of targeting actin dynamics as a new therapeutic opportunity and, in this case, as a plausible approach for the local treatment of osteoporosis.


Subject(s)
Actins/metabolism , Glucosides/pharmacology , Hydrolyzable Tannins/pharmacology , Osteoclasts/cytology , Osteoclasts/metabolism , Actin Cytoskeleton/metabolism , Animals , Bone Resorption/pathology , Cell Adhesion/drug effects , Cell Differentiation , Cell Survival/drug effects , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Glucosides/chemistry , Hydrolyzable Tannins/chemistry , Mice, Inbred C57BL , Osteoclasts/drug effects , Podosomes/metabolism , Polymerization
6.
J Clin Invest ; 127(11): 4001-4017, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28945203

ABSTRACT

Despite its central position in oncogenic intracellular signaling networks, the role of mTORC1 in epithelial development has not been studied extensively in vivo. Here, we have used the epidermis as a model system to elucidate the cellular effects and signaling feedback sequelae of mTORC1 loss of function in epithelial tissue. In mice with conditional epidermal loss of the mTORC1 components Rheb or Rptor, mTORC1 loss of function unexpectedly resulted in a profound skin barrier defect with epidermal abrasions, blistering, and early postnatal lethality, due to a thinned epidermis with decreased desmosomal protein expression and incomplete biochemical differentiation. In mice with mTORC1 loss of function, we found that Rho kinase (ROCK) signaling was constitutively activated, resulting in increased cytoskeletal tension and impaired cell-cell adhesion. Inhibition or silencing of ROCK1 was sufficient to rescue keratinocyte adhesion and biochemical differentiation in these mice. mTORC1 loss of function also resulted in marked feedback upregulation of upstream TGF-ß signaling, triggering ROCK activity and its downstream effects on desmosomal gene expression. These findings elucidate a role for mTORC1 in the regulation of epithelial barrier formation, cytoskeletal tension, and cell adhesion, underscoring the complexity of signaling feedback following mTORC1 inhibition.


Subject(s)
Keratinocytes/physiology , Mechanistic Target of Rapamycin Complex 1/genetics , Signal Transduction , Animals , Cell Adhesion , Cell Differentiation , Cells, Cultured , Desmosomes/physiology , Enzyme Activation , Epidermal Cells , Female , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , rho-Associated Kinases/metabolism
7.
J Cell Sci ; 130(19): 3213-3221, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28821574

ABSTRACT

The mammary epithelium elaborates through hormonally regulated changes in proliferation, migration and differentiation. Non-muscle myosin II (NMII) functions at the interface between contractility, adhesion and signal transduction. It is therefore a plausible regulator of mammary morphogenesis. We tested the genetic requirement for NMIIA and NMIIB in mammary morphogenesis through deletion of the three NMII heavy chain-encoding genes (NMHCIIA, NMHCIIB and NMHCIIC; also known as MYH9, MYH10 and MYH14, respectively) that confer specificity to the complex. Surprisingly, mosaic loss, but not ubiquitous loss, of NMHCIIA and NMHCIIB induced high levels of proliferation in 3D culture. This phenotype was observed even when cells were cultured in basal medium, which does not support tissue level growth of wild-type epithelium. Mosaic loss of NMIIA and NMIIB combined with FGF signaling to induce hyperplasia. Mosaic analysis revealed that the cells that were null for both NMIIA and NMIIB, as well as wild-type cells, proliferated, indicating that the regulation of proliferation is both cell autonomous and non-autonomous within epithelial tissues. This phenotype appears to be mediated by cell-cell contact, as co-culture did not induce proliferation. Mosaic loss of NMIIA and NMIIB also induced excess proliferation in vivo Our data therefore reveal a role for NMIIA and NMIIB as negative regulators of proliferation in the mammary epithelium.


Subject(s)
Cell Proliferation , Mammary Glands, Animal/metabolism , Myosin Heavy Chains/metabolism , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIB/metabolism , Animals , Cell Culture Techniques , Cells, Cultured , Female , Mammary Glands, Animal/cytology , Mice , Mice, Knockout , Myosin Heavy Chains/genetics , Myosin Type II/genetics , Myosin Type II/metabolism , Nonmuscle Myosin Type IIA/genetics , Nonmuscle Myosin Type IIB/genetics
8.
Nat Commun ; 8: 15008, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28452345

ABSTRACT

High-mobility group A1 (Hmga1) chromatin remodelling proteins are enriched in intestinal stem cells (ISCs), although their function in this setting was unknown. Prior studies showed that Hmga1 drives hyperproliferation, aberrant crypt formation and polyposis in transgenic mice. Here we demonstrate that Hmga1 amplifies Wnt/ß-catenin signalling to enhance self-renewal and expand the ISC compartment. Hmga1 upregulates genes encoding both Wnt agonist receptors and downstream Wnt effectors. Hmga1 also helps to 'build' an ISC niche by expanding the Paneth cell compartment and directly inducing Sox9, which is required for Paneth cell differentiation. In human intestine, HMGA1 and SOX9 are positively correlated, and both become upregulated in colorectal cancer. Our results define a unique role for Hmga1 in intestinal homeostasis by maintaining the stem cell pool and fostering terminal differentiation to establish an epithelial stem cell niche. This work also suggests that deregulated Hmga1 perturbs this equilibrium during intestinal carcinogenesis.


Subject(s)
HMGA1a Protein/metabolism , Intestinal Mucosa/metabolism , Paneth Cells/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , HMGA1a Protein/genetics , Humans , Intestinal Mucosa/cytology , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Paneth Cells/cytology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Stem Cell Niche , Stem Cells/cytology , Time-Lapse Imaging
9.
Biol Open ; 5(9): 1216-28, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27402962

ABSTRACT

Dissemination is the process by which cells detach and migrate away from a multicellular tissue. The epithelial-to-mesenchymal transition (EMT) conceptualizes dissemination in a stepwise fashion, with downregulation of E-cadherin leading to loss of intercellular junctions, induction of motility, and then escape from the epithelium. This gain of migratory activity is proposed to be mutually exclusive with proliferation. We previously developed a dissemination assay based on inducible expression of the transcription factor Twist1 and here utilize it to characterize the timing and dynamics of intercellular adhesion, proliferation and migration during dissemination. Surprisingly, Twist1(+) epithelium displayed extensive intercellular junctions, and Twist1(-) luminal epithelial cells could still adhere to disseminating Twist1(+) cells. Although proteolysis and proliferation were both observed throughout dissemination, neither was absolutely required. Finally, Twist1(+) cells exhibited a hybrid migration mode; their morphology and nuclear deformation were characteristic of amoeboid cells, whereas their dynamic protrusive activity, pericellular proteolysis and migration speeds were more typical of mesenchymal cells. Our data reveal that epithelial cells can disseminate while retaining competence to adhere and proliferate.

10.
Calcif Tissue Int ; 97(1): 69-79, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25987164

ABSTRACT

Vitamin D and FGF23 play a major role in calcium/phosphate balance. Vitamin D may control bone resorption but the potential role of FGF23 has never been evaluated. The objective of this study was therefore to compare the effects of vitamin D and FGF23 on osteoclast differentiation and activity in human monocyte-derived osteoclasts. Human monocytes, purified from blood of healthy donors, were incubated with M-CSF and RANKL to obtain mature multinucleated osteoclasts (MNC). Experiments were carried out to assess the effects of FGF23 as compared to native vitamin D (25-D) and active vitamin D (1,25-D) on osteoclast differentiation and on bone-resorbing osteoclast activity. Additional experiments with the pan fibroblast growth factor receptor inhibitor (FGFR-i) were performed. Phosphorylation Akt and Erk pathways were analyzed by Western blot analyses. Both 1,25-D and FGF23, to a lesser extent, significantly inhibited osteoclastogenesis at early stages; when adding FGFR-i, osteoclast formation was restored. Biochemical experiments showed an activation of the Akt and Erk pathways under FGF23 treatment. In contrast, in terms of activity, 1,25-D had no effect on resorption, whereas FGF23 slightly but significantly increased bone resorption; 25-D had no effects on either differentiation or on activity. These data show that 1,25-D inhibits osteoclastogenesis without regulating osteoclast-mediated bone resorption activity; FGF23 has biphasic effects on osteoclast physiology, inhibiting osteoclast formation while stimulating slightly osteoclast activity. These results may be of importance and taken into account in chronic kidney disease when therapies modulating FGF23 are available.


Subject(s)
Cell Differentiation/drug effects , Fibroblast Growth Factors/pharmacology , Osteoclasts/drug effects , Vitamin D/pharmacology , Bone Resorption/drug therapy , Cells, Cultured , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Humans , Macrophage Colony-Stimulating Factor/metabolism , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/metabolism , Vitamin D/metabolism
11.
Cell Adh Migr ; 8(3): 191-204, 2014.
Article in English | MEDLINE | ID: mdl-24714644

ABSTRACT

Osteoclasts are the cells responsible for physiological bone resorption. A specific organization of their most prominent cytoskeletal structures, podosomes, is crucial for the degradation of mineralized bone matrix. Each podosome is constituted of an F-actin-enriched central core surrounded by a loose F-actin network, called the podosome cloud. In addition to intrinsic actin dynamics, podosomes are defined by their adhesion to the extracellular matrix, mainly via core-linking CD44 and cloud-linking integrins. These properties allow podosomes to collectively evolve into different patterns implicated in migration and bone resorption. Indeed, to resorb bone, osteoclasts polarize, actively secrete protons, and proteases into the resorption pit where these molecules are confined by a podosome-containing sealing zone. Here, we review recent advancements on podosome structure and regulatory pathways in osteoclasts. We also discuss the distinct functions of different podosome patterns during the lifespan of a single osteoclast.


Subject(s)
Bone Resorption/metabolism , Osteoclasts/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Adhesion/physiology , Cell Differentiation/physiology , Humans , Osteoclasts/cytology
12.
Mol Biol Cell ; 25(3): 380-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24284899

ABSTRACT

The function of osteoclasts (OCs), multinucleated giant cells (MGCs) of the monocytic lineage, is bone resorption. To resorb bone, OCs form podosomes. These are actin-rich adhesive structures that pattern into rings that drive OC migration and into "sealing-zones" (SZs) that confine the resorption lacuna. Although changes in actin dynamics during podosome patterning have been documented, the mechanisms that regulate these changes are largely unknown. From human monocytic precursors, we differentiated MGCs that express OC degradation enzymes but are unable to resorb the mineral matrix. We demonstrated that, despite exhibiting bona fide podosomes, these cells presented dysfunctional SZs. We then performed two-step differential transcriptomic profiling of bone-resorbing OCs versus nonresorbing MGCs to generate a list of genes implicated in bone resorption. From this list of candidate genes, we investigated the role of Rho/Rnd3. Using primary RhoE-deficient OCs, we demonstrated that RhoE is indispensable for OC migration and bone resorption by maintaining fast actin turnover in podosomes. We further showed that RhoE activates podosome component cofilin by inhibiting its Rock-mediated phosphorylation. We conclude that the RhoE-Rock-cofilin pathway, by promoting podosome dynamics and patterning, is central for OC migration, SZ formation, and, ultimately, bone resorption.


Subject(s)
Actins/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , rho GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Amides/pharmacology , Animals , Bone Resorption/genetics , Cattle , Cell Differentiation/genetics , Cell Movement , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Giant Cells/metabolism , Humans , Mice , Mice, Transgenic , Phosphorylation , Pyridines/pharmacology , Transcriptome , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
13.
J Clin Invest ; 122(5): 1791-802, 2012 May.
Article in English | MEDLINE | ID: mdl-22505457

ABSTRACT

Autoimmunity is complicated by bone loss. In human rheumatoid arthritis (RA), the most severe inflammatory joint disease, autoantibodies against citrullinated proteins are among the strongest risk factors for bone destruction. We therefore hypothesized that these autoantibodies directly influence bone metabolism. Here, we found a strong and specific association between autoantibodies against citrullinated proteins and serum markers for osteoclast-mediated bone resorption in RA patients. Moreover, human osteoclasts expressed enzymes eliciting protein citrullination, and specific N-terminal citrullination of vimentin was induced during osteoclast differentiation. Affinity-purified human autoantibodies against mutated citrullinated vimentin (MCV) not only bound to osteoclast surfaces, but also led to robust induction of osteoclastogenesis and bone-resorptive activity. Adoptive transfer of purified human MCV autoantibodies into mice induced osteopenia and increased osteoclastogenesis. This effect was based on the inducible release of TNF-α from osteoclast precursors and the subsequent increase of osteoclast precursor cell numbers with enhanced expression of activation and growth factor receptors. Our data thus suggest that autoantibody formation in response to citrullinated vimentin directly induces bone loss, providing a link between the adaptive immune system and bone.


Subject(s)
Autoantibodies/metabolism , Bone Resorption/immunology , Citrulline/immunology , Osteoclasts/physiology , Vimentin/immunology , Animals , Antibody Specificity , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Autoantibodies/blood , Autoantibodies/isolation & purification , Biomarkers/blood , Bone and Bones/immunology , Bone and Bones/pathology , Case-Control Studies , Cell Differentiation , Cells, Cultured , Collagen Type I/blood , Humans , Hydrolases/metabolism , Mice , Mice, Transgenic , Osteoclasts/enzymology , Osteoclasts/metabolism , Protein Processing, Post-Translational , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Tumor Necrosis Factor-alpha/blood
14.
Mol Biol Cell ; 22(17): 3120-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21737683

ABSTRACT

Podosomes are dynamic, actin-containing adhesion structures that collectively self-organize as rings. In this study, we first show by observing osteoclasts plated on bead-seeded soft substrates that podosome assemblies, such as rings, are involved in tension forces. During the expansion of a podosome ring, substrate displacement is oriented outward, suggesting that podosomal structures push the substrate away. To further elucidate the function of forces generated by podosomes, we analyze osteoclast migration. Determining the centers of mass of the whole cell (G) and of actin (P), we demonstrate that osteoclasts migrate by "jumps" and that the trajectories of G and P are strongly correlated. The velocity of the center of mass as a function of time reveals that osteoclasts rapidly catch up with podosomal structures in a periodic pattern. We conclude that actin dynamics inside the cell are not only correlated with cell migration, but drive it.


Subject(s)
Cell Adhesion , Cell Movement , Cell-Matrix Junctions/metabolism , Osteoclasts/physiology , Actins/metabolism , Algorithms , Animals , Biomechanical Phenomena , Cell Line , Green Fluorescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Osteoclasts/metabolism , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis , Stress, Mechanical , Time-Lapse Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...