Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Vis Exp ; (123)2017 05 14.
Article in English | MEDLINE | ID: mdl-28570523

ABSTRACT

Reactive oxygen species (ROS) regulate essential cellular processes including gene expression, migration, differentiation and proliferation. However, excessive ROS levels induce a state of oxidative stress, which is accompanied by irreversible oxidative damage to DNA, lipids and proteins. Thus, quantification of ROS provides a direct proxy for cellular health condition. Since mitochondria are among the major cellular sources and targets of ROS, joint analysis of mitochondrial function and ROS production in the same cells is crucial for better understanding the interconnection in pathophysiological conditions. Therefore, a high-content microscopy-based strategy was developed for simultaneous quantification of intracellular ROS levels, mitochondrial membrane potential (ΔΨm) and mitochondrial morphology. It is based on automated widefield fluorescence microscopy and image analysis of living adherent cells, grown in multi-well plates, and stained with the cell-permeable fluorescent reporter molecules CM-H2DCFDA (ROS) and TMRM (ΔΨm and mitochondrial morphology). In contrast with fluorimetry or flow-cytometry, this strategy allows quantification of subcellular parameters at the level of the individual cell with high spatiotemporal resolution, both before and after experimental stimulation. Importantly, the image-based nature of the method allows extracting morphological parameters in addition to signal intensities. The combined feature set is used for explorative and statistical multivariate data analysis to detect differences between subpopulations, cell types and/or treatments. Here, a detailed description of the assay is provided, along with an example experiment that proves its potential for unambiguous discrimination between cellular states after chemical perturbation.


Subject(s)
Microscopy, Fluorescence/methods , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/physiology , Fluoresceins , Fluorescent Dyes , Humans , Membrane Potential, Mitochondrial , Mitochondria/physiology , Oxidation-Reduction , Oxidative Stress , Rhodamines
2.
Cell Adh Migr ; 11(1): 98-109, 2017 01 02.
Article in English | MEDLINE | ID: mdl-27111836

ABSTRACT

The mechanical properties of living cells reflect their propensity to migrate and respond to external forces. Both cellular and nuclear stiffnesses are strongly influenced by the rigidity of the extracellular matrix (ECM) through reorganization of the cyto- and nucleoskeletal protein connections. Changes in this architectural continuum affect cell mechanics and underlie many pathological conditions. In this context, an accurate and combined quantification of the mechanical properties of both cells and nuclei can contribute to a better understanding of cellular (dys-)function. To address this challenge, we have established a robust method for probing cellular and nuclear deformation during spreading and detachment from micropatterned substrates. We show that (de-)adhesion kinetics of endothelial cells are modulated by substrate stiffness and rely on the actomyosin network. We combined this approach with measurements of cell stiffness by magnetic tweezers to show that relaxation dynamics can be considered as a reliable parameter of cellular pre-stress in adherent cells. During the adhesion stage, large cellular and nuclear deformations occur over a long time span (>60 min). Conversely, nuclear deformation and condensed chromatin are relaxed in a few seconds after detachment. Finally, our results show that accumulation of farnesylated prelamin leads to modifications of the nuclear viscoelastic properties, as reflected by increased nuclear relaxation times. Our method offers an original and non-intrusive way of simultaneously gauging cellular and nuclear mechanics, which can be extended to high-throughput screens of pathological conditions and potential countermeasures.


Subject(s)
Cell Nucleus/metabolism , Cytoskeleton/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Microtechnology/methods , Stress, Mechanical , Actomyosin/metabolism , Biomechanical Phenomena , Cell Adhesion , Cell Movement , Cell Nucleus Shape , Cell Shape , Extracellular Matrix/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kinetics , Lamin Type A/metabolism , Time Factors
3.
Cell Adh Migr ; 11(5-6): 447-463, 2017 Sep 03.
Article in English | MEDLINE | ID: mdl-27791462

ABSTRACT

The nuclear lamina mechanically integrates the nucleus with the cytoskeleton and extracellular environment and regulates gene expression. These functions are exerted through direct and indirect interactions with the lamina's major constituent proteins, the A-type lamins, which are encoded by the LMNA gene. Using quantitative stable isotope labeling-based shotgun proteomics we have analyzed the proteome of human dermal fibroblasts in which we have depleted A-type lamins by means of a sustained siRNA-mediated LMNA knockdown. Gene ontology analysis revealed that the largest fraction of differentially produced proteins was involved in actin cytoskeleton organization, in particular proteins involved in focal adhesion dynamics, such as actin-related protein 2 and 3 (ACTR2/3), subunits of the ARP2/3 complex, and fascin actin-bundling protein 1 (FSCN1). Functional validation using quantitative immunofluorescence showed a significant reduction in the size of focal adhesion points in A-type lamin depleted cells, which correlated with a reduction in early cell adhesion capacity and an increased cell motility. At the same time, loss of A-type lamins led to more pronounced stress fibers and higher traction forces. This phenotype could not be mimicked or reversed by experimental modulation of the STAT3-IL6 pathway, but it was partly recapitulated by chemical inhibition of the ARP2/3 complex. Thus, our data suggest that the loss of A-type lamins perturbs the balance between focal adhesions and cytoskeletal tension. This imbalance may contribute to mechanosensing defects observed in certain laminopathies.


Subject(s)
Cytoskeleton/metabolism , Focal Adhesions/metabolism , Lamin Type A/metabolism , Actin-Related Protein 2/genetics , Actin-Related Protein 2/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 3/genetics , Actin-Related Protein 3/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion/genetics , Cell Adhesion/physiology , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/metabolism , Fibroblasts , Humans , Interleukin-6/metabolism , Lamin Type A/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Proteome/metabolism , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Wound Healing/physiology
4.
Adv Anat Embryol Cell Biol ; 219: 149-77, 2016.
Article in English | MEDLINE | ID: mdl-27207366

ABSTRACT

Oxidative stress arises from an imbalance between the production of reactive oxygen species (ROS) and their removal by cellular antioxidant systems. Especially under pathological conditions, mitochondria constitute a relevant source of cellular ROS. These organelles harbor the electron transport chain, bringing electrons in close vicinity to molecular oxygen. Although a full understanding is still lacking, intracellular ROS generation and mitochondrial function are also linked to changes in mitochondrial morphology. To study the intricate relationships between the different factors that govern cellular redox balance in living cells, we have developed a high-content microscopy-based strategy for simultaneous quantification of intracellular ROS levels and mitochondrial morphofunction. Here, we summarize the principles of intracellular ROS generation and removal, and we explain the major considerations for performing quantitative microscopy analyses of ROS and mitochondrial morphofunction in living cells. Next, we describe our workflow, and finally, we illustrate that a multiparametric readout enables the unambiguous classification of chemically perturbed cells as well as laminopathy patient cells.


Subject(s)
Image Processing, Computer-Assisted/statistics & numerical data , Microscopy, Fluorescence/methods , Mitochondria/ultrastructure , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Humans , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence/instrumentation , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/antagonists & inhibitors , Workflow
5.
Nucleus ; 6(3): 236-46, 2015.
Article in English | MEDLINE | ID: mdl-25996284

ABSTRACT

The cell nucleus is structurally and functionally organized by lamins, intermediate filament proteins that form the nuclear lamina. Point mutations in genes that encode a specific subset of lamins, the A-type lamins, cause a spectrum of diseases termed laminopathies. Recent evidence points to a role for A-type lamins in intracellular redox homeostasis. To determine whether lamin A/C depletion and prelamin A accumulation differentially induce oxidative stress, we have performed a quantitative microscopy-based analysis of reactive oxygen species (ROS) levels and mitochondrial membrane potential (Δψm) in human fibroblasts subjected to sustained siRNA-mediated knockdown of LMNA and ZMPSTE24, respectively. We measured a highly significant increase in basal ROS levels and an even more prominent rise of induced ROS levels in lamin A/C depleted cells, eventually resulting in Δψm hyperpolarization and apoptosis. Depletion of ZMPSTE24 on the other hand, triggered a senescence pathway that was associated with moderately increased ROS levels and a transient Δψm depolarization. Both knockdowns were accompanied by an upregulation of several ROS detoxifying enzymes. Taken together, our data suggest that both persistent prelamin A accumulation and lamin A/C depletion elevate ROS levels, but to a different extent and with different effects on cell fate. This may contribute to the variety of disease phenotypes witnessed in laminopathies.


Subject(s)
Fibroblasts/metabolism , Lamin Type A/metabolism , Mitochondria/metabolism , Nuclear Lamina/metabolism , Reactive Oxygen Species/metabolism , Apoptosis , Fibroblasts/cytology , Gene Expression Regulation , Humans , Lamin Type A/antagonists & inhibitors , Lamin Type A/genetics , Membrane Potential, Mitochondrial , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mitochondria/pathology , Nuclear Lamina/chemistry , Oxidative Stress , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...