24 results on '"Maria Carla Parrini"'
Search Results
2. Discovering the hidden messages within cell trajectories using a deep learning approach for in vitro evaluation of cancer drug treatments
- Author
-
Maria Colomba Comes, C. Di Natale, Lina Ghibelli, Davide Di Giuseppe, Eugenio Martinelli, Arianna Mencattini, Paola Casti, Francesca Romana Bertani, Francesca Corsi, Luca Businaro, and Maria Carla Parrini
- Subjects
0301 basic medicine ,Computer science ,Cancer drugs ,Cell ,time lapse microscopy ,Neural Network ,Motility ,lcsh:Medicine ,Antineoplastic Agents ,Bioengineering ,cell motility ,Machine learning ,computer.software_genre ,Convolutional neural network ,Time-Lapse Imaging ,Settore ING-INF/07 ,Article ,Image analysis ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,Deep Learning ,Image processing ,medicine ,Humans ,lcsh:Science ,Multidisciplinary ,business.industry ,Deep learning ,lcsh:R ,Reproducibility of Results ,Computational Biology ,Molecular Imaging ,030104 developmental biology ,medicine.anatomical_structure ,Cell Tracking ,lcsh:Q ,Artificial intelligence ,Drug Screening Assays, Antitumor ,business ,computer ,Biomedical engineering ,030217 neurology & neurosurgery ,Algorithms - Abstract
We describe a novel method to achieve a universal, massive, and fully automated analysis of cell motility behaviours, starting from time-lapse microscopy images. The approach was inspired by the recent successes in application of machine learning for style recognition in paintings and artistic style transfer. The originality of the method relies i) on the generation of atlas from the collection of single-cell trajectories in order to visually encode the multiple descriptors of cell motility, and ii) on the application of pre-trained Deep Learning Convolutional Neural Network architecture in order to extract relevant features to be used for classification tasks from this visual atlas. Validation tests were conducted on two different cell motility scenarios: 1) a 3D biomimetic gels of immune cells, co-cultured with breast cancer cells in organ-on-chip devices, upon treatment with an immunotherapy drug; 2) Petri dishes of clustered prostate cancer cells, upon treatment with a chemotherapy drug. For each scenario, single-cell trajectories are very accurately classified according to the presence or not of the drugs. This original approach demonstrates the existence of universal features in cell motility (a so called “motility style”) which are identified by the DL approach in the rationale of discovering the unknown message in cell trajectories.
- Published
- 2020
- Full Text
- View/download PDF
3. Direct imaging and automatic analysis in tumor-on-chip reveal cooperative antitumoral activity of immune cells and oncolytic vaccinia virus
- Author
-
Arianna Mencattini, Christine Lansche, Irina Veith, Philippe Erbs, Jean-Marc Balloul, Eric Quemeneur, Stéphanie Descroix, Fatima Mechta-Grigoriou, Gérard Zalcman, Cécile Zaupa, Maria Carla Parrini, and Eugenio Martinelli
- Subjects
Oncolytic Virotherapy ,Biomedical Engineering ,Biophysics ,Cancer immunotherapy ,Vaccinia virus ,General Medicine ,Oncolytic vaccinia virus ,Biosensing Techniques ,Settore ING-INF/07 ,Oncolytic Viruses ,Neoplasms ,Machine learning ,Tumor-on-chip ,Electrochemistry ,Tumor Microenvironment ,Humans ,Biotechnology ,Live imaging - Abstract
Organ-on-chip and tumor-on-chip microfluidic cell cultures represent a fast-growing research field for modelling organ functions and diseases, for drug development, and for promising applications in personalized medicine. Still, one of the bottlenecks of this technology is the analysis of the huge amount of bio-images acquired in these dynamic 3D microenvironments, a task that we propose to achieve by exploiting the interdisciplinary contributions of computer science and electronic engineering. In this work, we apply this strategy to the study of oncolytic vaccinia virus (OVV), an emerging agent in cancer immunotherapy. Infection and killing of cancer cells by OVV were recapitulated and directly imaged in tumor-on-chip. By developing and applying appropriate image analysis strategies and advanced automatic algorithms, we uncovered synergistic cooperation of OVV and immune cells to kill cancer cells. Moreover, we observed that the kinetics of immune cells were modified in presence of OVV and that these immune modulations varied during the course of infection. A correlation between cancer cell infection and cancer-immune interaction time was pointed out, strongly supporting a cause-effect relationship between infection of cancer cells and their recognition by the immune cells. These results shed new light on the mode of action of OVV, and suggest new clinical avenues for immunotherapy developments.
- Published
- 2022
4. Autophagy Is Polarized toward Cell Front during Migration and Spatially Perturbed by Oncogenic Ras
- Author
-
Irina Veith, Mathieu Coppey, Manish Kumar Singh, Jacques Camonis, Giulia Zago, Maria Carla Parrini, Centre de recherche de l'Institut Curie [Paris], Institut Curie [Paris], Unité de génétique et biologie des cancers (U830), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Laboratoire Physico-Chimie Curie [Institut Curie] (PCC), Institut Curie [Paris]-Institut de Chimie du CNRS (INC)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), and Gestionnaire, HAL Sorbonne Université 5
- Subjects
autophagy ,QH301-705.5 ,Cell ,Cellular homeostasis ,Biology ,migration ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Live cell imaging ,Lysosome ,Image Processing, Computer-Assisted ,medicine ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Animals ,Humans ,cancer ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Biology (General) ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,Autophagy ,Cell migration ,Oncogenes ,General Medicine ,Cell biology ,Cell Transformation, Neoplastic ,Genes, ras ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,micro-patterns ,Cattle ,Collagen ,Lysosomes ,Gels ,Microtubule-Associated Proteins ,Intracellular ,Ras - Abstract
International audience; Autophagy is a physiological degradation process that removes unnecessary or dysfunctional components of cells. It is important for normal cellular homeostasis and as a response to a variety of stresses, such as nutrient deprivation. Defects in autophagy have been linked to numerous human diseases, including cancers. Cancer cells require autophagy to migrate and to invade. Here, we study the intracellular topology of this interplay between autophagy and cell migration by an interdisciplinary live imaging approach which combines micro-patterning techniques and an autophagy reporter (RFP-GFP-LC3) to monitor over time, during directed migration, the back–front spatial distribution of LC3-positive compartments (autophagosomes and autolysosomes). Moreover, by exploiting a genetically controlled cell model, we assessed the impact of transformation by the Ras oncogene, one of the most frequently mutated genes in human cancers, which is known to increase both cell motility and basal autophagy. Static cells displayed an isotropic distribution of autophagy LC3-positive compartments. Directed migration globally increased autophagy and polarized both autophagosomes and autolysosomes at the front of the nucleus of migrating cells. In Ras-transformed cells, the front polarization of LC3 compartments was much less organized, spatially and temporally, as compared to normal cells. This might be a consequence of altered lysosome positioning. In conclusion, this work reveals that autophagy organelles are polarized toward the cell front during migration and that their spatial-temporal dynamics are altered in motile cancer cells that express an oncogenic Ras protein.
- Published
- 2021
- Full Text
- View/download PDF
5. The influence of spatial and temporal resolutions on the analysis of cell-cell interaction: a systematic study for time-lapse microscopy applications
- Author
-
C. Di Natale, Luca Businaro, A. De Ninno, Eugenio Martinelli, Fanny Mermet-Meillon, Maria Colomba Comes, Davide Di Giuseppe, Maria Carla Parrini, Arianna Mencattini, and Paola Casti
- Subjects
0301 basic medicine ,Computer science ,Stochastic particle model ,Microfluidics ,Cell ,lcsh:Medicine ,Breast Neoplasms ,Cell Communication ,Time-Lapse Imaging ,Article ,Time-lapse microscopy ,03 medical and health sciences ,Spatio-Temporal Analysis ,0302 clinical medicine ,Cell–cell interaction ,Live cell imaging ,Microscopy ,cell-cell interaction ,Settore ING-INF/07 - Misure Elettriche e Elettroniche ,medicine ,Humans ,Computer Simulation ,Interaction dynamics ,lcsh:Science ,Microscopy, Video ,Multidisciplinary ,lcsh:R ,Electrical and electronic engineering ,030104 developmental biology ,medicine.anatomical_structure ,motility ,Cell Tracking ,Leukocytes, Mononuclear ,Female ,lcsh:Q ,Organ-on-chip ,Biological system ,Biomedical engineering ,Algorithms ,030217 neurology & neurosurgery - Abstract
Cell-cell interactions are an observable manifestation of underlying complex biological processes occurring in response to diversified biochemical stimuli. Recent experiments with microfluidic devices and live cell imaging show that it is possible to characterize cell kinematics via computerized algorithms and unravel the effects of targeted therapies. We study the influence of spatial and temporal resolutions of time-lapse videos on motility and interaction descriptors with computational models that mimic the interaction dynamics among cells. We show that the experimental set-up of time-lapse microscopy has a direct impact on the cell tracking algorithm and on the derived numerical descriptors. We also show that, when comparing kinematic descriptors in two diverse experimental conditions, too low resolutions may alter the descriptors’ discriminative power, and so the statistical significance of the difference between the two compared distributions. The conclusions derived from the computational models were experimentally confirmed by a series of video-microscopy acquisitions of co-cultures of unlabelled human cancer and immune cells embedded in 3D collagen gels within microfluidic devices. We argue that the experimental protocol of acquisition should be adapted to the specific kind of analysis involved and to the chosen descriptors in order to derive reliable conclusions and avoid biasing the interpretation of results.
- Published
- 2019
- Full Text
- View/download PDF
6. In vitro bone metastasis dwelling in a 3D bioengineered niche
- Author
-
Frédéric Deschaseaux, Stéphanie Descroix, Anne Vincent-Salomon, Elodie Montaudon, Rania El Botty, Weijing Han, Nicolas Espagnolle, Laurent Malaquin, Luc Sensebé, Sergio Roman-Roman, Jacques Camonis, Pascal Silberzan, Franck Assayag, Paul Cottu, Maria Carla Parrini, Elisabetta Marangoni, Gérard Zalcman, Guillaume Dutertre, Unité de génétique et biologie des cancers (U830), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Curie [Paris], Équipe Ingénierie pour les sciences du vivant (LAAS-ELIA), Laboratoire d'analyse et d'architecture des systèmes (LAAS), Université Toulouse Capitole (UT Capitole), Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Université de Toulouse (UT)-Institut National des Sciences Appliquées (INSA)-Université Toulouse - Jean Jaurès (UT2J), Université de Toulouse (UT)-Université Toulouse III - Paul Sabatier (UT3), Université de Toulouse (UT)-Centre National de la Recherche Scientifique (CNRS)-Institut National Polytechnique (Toulouse) (Toulouse INP), Université de Toulouse (UT)-Université Toulouse Capitole (UT Capitole), Université de Toulouse (UT), STROMALab, Université Toulouse III - Paul Sabatier (UT3), Université de Toulouse (UT)-Université de Toulouse (UT)-Ecole Nationale Vétérinaire de Toulouse (ENVT), Institut National Polytechnique (Toulouse) (Toulouse INP), Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National Polytechnique (Toulouse) (Toulouse INP), Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement Français du Sang-Centre National de la Recherche Scientifique (CNRS), Laboratoire Physico-Chimie Curie [Institut Curie] (PCC), Institut Curie [Paris]-Institut de Chimie du CNRS (INC)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), European Project: 760927,H2020-HoliFAB project, Université Toulouse - Jean Jaurès (UT2J)-Université Toulouse 1 Capitole (UT1), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Toulouse - Jean Jaurès (UT2J)-Université Toulouse 1 Capitole (UT1), Université Fédérale Toulouse Midi-Pyrénées, Centre National de la Recherche Scientifique (CNRS)-Ecole Nationale Vétérinaire de Toulouse (ENVT), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Etablissement Français du Sang-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Université Fédérale Toulouse Midi-Pyrénées-Institut National des Sciences Appliquées (INSA)-Université Toulouse - Jean Jaurès (UT2J)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS)-Institut National Polytechnique (Toulouse) (Toulouse INP), Université Fédérale Toulouse Midi-Pyrénées-Université Toulouse Capitole (UT Capitole), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Ecole Nationale Vétérinaire de Toulouse (ENVT), Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement Français du Sang-Centre National de la Recherche Scientifique (CNRS), Silberzan, Pascal, and HoliFAB - H2020-HoliFAB project - 760927 - INCOMING
- Subjects
3D-printing ,Patient-derived-xenograft ,Biophysics ,Bone Neoplasms ,Breast Neoplasms ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Bioengineering ,02 engineering and technology ,Bone and Bones ,Metastasis ,Biomaterials ,03 medical and health sciences ,Breast cancer ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Biomimetics ,In vivo ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,[SDV.IB.BIO]Life Sciences [q-bio]/Bioengineering/Biomaterials ,030304 developmental biology ,0303 health sciences ,Osteoblasts ,business.industry ,Bone metastasis ,Cancer ,021001 nanoscience & nanotechnology ,medicine.disease ,Metastatic breast cancer ,In vitro ,3. Good health ,[SDV.IB.BIO] Life Sciences [q-bio]/Bioengineering/Biomaterials ,Mechanics of Materials ,Cancer cell ,Ceramics and Composites ,Cancer research ,0210 nano-technology ,business ,Model - Abstract
International audience; Bone is the most frequent metastasis site for breast cancer. As well as dramatically increasing disease burden, bone metastases are also an indicator of poor prognosis. One of the main challenges in investigating bone metastasis in breast cancer is engineering in vitro models that replicate the features of in vivo bone environments. Such in vitro models ideally enable the biology of the metastatic cells to mimic their in vivo behavior as closely as possible. Here, taking benefit of cutting-edge technologies both in microfabrication and cancer cell biology, we have developed an in vitro breast cancer bone-metastasis model. To do so we first 3D printed a bone scaffold that reproduces the trabecular architecture and that can be conditioned with osteoblast-like cells, a collagen matrix, and mineralized calcium. We thus demonstrated that this device offers an adequate soil to seed primary breast cancer bone metastatic cells. In particular, patient-derived xenografts being considered as a better approach than cell lines to achieve clinically relevant results, we demonstrate the ability of this biomimetic bone niche model to host patient-derived xenografted metastatic breast cancer cells. These patient-derived xenograft cells show a long-term survival in the bone model and maintain their cycling propensity, and exhibit the same modulated drug response as in vivo. This experimental system enables access to the idiosyncratic features of the bone microenvironment and cancer bone metastasis, which has implications for drug testing.
- Published
- 2021
- Full Text
- View/download PDF
7. Cancer-associated fibroblast heterogeneity in axillary lymph nodes drives metastases in breast cancer through complementary mechanisms
- Author
-
Fatima Mechta-Grigoriou, Yann Kieffer, Anne-Marie Givel, Maria Carla Parrini, Anne Vincent-Salomon, Stéphanie Descroix, Pascal Silberzan, Claire Bonneau, Isabelle Bonnet, Laetitia Fuhrmann, Ana Catarina Costa, Ilaria Magagna, Floriane Pelon, Brigitte Bourachot, Danijela Matic Vignjevic, Jorge Barbazan, Youmna Attieh, Fanny Mermet-Meillon, Unité de génétique et biologie des cancers (U830), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5), Sorbonne Université (SU), Laboratoire Physico-Chimie Curie [Institut Curie] (PCC), Institut Curie [Paris]-Institut de Chimie du CNRS (INC)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Université Paris Descartes - Paris 5 (UPD5), Pathology [Paris], Department of Tumor Biology [Paris], Institut Curie [Paris]-Institut Curie [Paris], Physico-Chimie-Curie (PCC), Centre National de la Recherche Scientifique (CNRS)-Institut Curie [Paris]-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Chimie du CNRS (INC), Compartimentation et dynamique cellulaires (CDC), Centre National de la Recherche Scientifique (CNRS)-Institut Curie [Paris]-Université Pierre et Marie Curie - Paris 6 (UPMC), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Curie-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Institut Curie-Institut Curie, Centre National de la Recherche Scientifique (CNRS)-Institut Curie-Université Pierre et Marie Curie - Paris 6 (UPMC), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), and Institut Pierre-Gilles de Gennes pour la Microfluidique
- Subjects
0301 basic medicine ,[SDV]Life Sciences [q-bio] ,General Physics and Astronomy ,Cell Separation ,Kaplan-Meier Estimate ,Metastasis ,Breast cancer ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Transforming Growth Factor beta ,Tumor Cells, Cultured ,Tumor Microenvironment ,lcsh:Science ,Myofibroblasts ,Cancer ,Aged, 80 and over ,Multidisciplinary ,Receptors, Notch ,Middle Aged ,Flow Cytometry ,Prognosis ,Progression-Free Survival ,3. Good health ,medicine.anatomical_structure ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Female ,Signal Transduction ,Adult ,Cancer microenvironment ,Epithelial-Mesenchymal Transition ,Axillary lymph nodes ,Tumour heterogeneity ,Science ,Primary Cell Culture ,Notch signaling pathway ,Breast Neoplasms ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Aged ,Cell Proliferation ,Tumor microenvironment ,General Chemistry ,medicine.disease ,Chemokine CXCL12 ,030104 developmental biology ,Axilla ,Cancer cell ,Cancer research ,lcsh:Q ,Lymph Nodes ,Follow-Up Studies - Abstract
Although fibroblast heterogeneity is recognized in primary tumors, both its characterization in and its impact on metastases remain unknown. Here, combining flow cytometry, immunohistochemistry and RNA-sequencing on breast cancer samples, we identify four Cancer-Associated Fibroblast (CAF) subpopulations in metastatic lymph nodes (LN). Two myofibroblastic subsets, CAF-S1 and CAF-S4, accumulate in LN and correlate with cancer cell invasion. By developing functional assays on primary cultures, we demonstrate that these subsets promote metastasis through distinct functions. While CAF-S1 stimulate cancer cell migration and initiate an epithelial-to-mesenchymal transition through CXCL12 and TGFβ pathways, highly contractile CAF-S4 induce cancer cell invasion in 3-dimensions via NOTCH signaling. Patients with high levels of CAFs, particularly CAF-S4, in LN at diagnosis are prone to develop late distant metastases. Our findings suggest that CAF subset accumulation in LN is a prognostic marker, suggesting that CAF subsets could be examined in axillary LN at diagnosis., Cancer associated fibroblasts are known to promote the progression of cancer. Here, the authors show that two particular subsets of cancer associated fibroblasts induce metastasis but work via distinct mechanisms including, chemokine signalling and Notch signalling.
- Published
- 2020
- Full Text
- View/download PDF
8. Localization of RalB signaling at endomembrane compartments and its modulation by autophagy
- Author
-
Alexandre P. J. Martin, Jacques Camonis, Manish Kumar Singh, Mathieu Coppey, Maria Carla Parrini, Carine Joffre, Giulia Zago, Laboratoire Aimé Cotton (LAC), Centre National de la Recherche Scientifique (CNRS)-Université Paris-Sud - Paris 11 (UP11)-École normale supérieure - Cachan (ENS Cachan), Institut des Sciences de la mécanique et Applications industrielles (IMSIA - UMR 9219), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-École Nationale Supérieure de Techniques Avancées (ENSTA Paris)-Centre National de la Recherche Scientifique (CNRS)-Université Paris-Saclay-EDF R&D (EDF R&D), EDF (EDF)-EDF (EDF), Institut Universitaire du Cancer de Toulouse - Oncopole (IUCT Oncopole - UMR 1037), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-CHU Toulouse [Toulouse]-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Curie [Paris], Unité de génétique et biologie des cancers (U830), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5), Laboratoire Physico-Chimie Curie [Institut Curie] (PCC), Institut Curie [Paris]-Institut de Chimie du CNRS (INC)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), coppey, mathieu, École normale supérieure - Cachan (ENS Cachan)-Université Paris-Sud - Paris 11 (UP11)-Centre National de la Recherche Scientifique (CNRS), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-École Nationale Supérieure de Techniques Avancées (ENSTA Paris)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-EDF R&D (EDF R&D), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), and Université de Toulouse (UT)-Université de Toulouse (UT)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Autophagosome ,Endosome ,[SDV]Life Sciences [q-bio] ,lcsh:Medicine ,GTPase ,Article ,Imaging ,03 medical and health sciences ,0302 clinical medicine ,Autophagy ,Guanine Nucleotide Exchange Factors ,Humans ,Endomembrane system ,lcsh:Science ,ComputingMilieux_MISCELLANEOUS ,RALB ,Multidisciplinary ,Chemistry ,lcsh:R ,Intracellular Membranes ,Publisher Correction ,Cell biology ,Cell Compartmentation ,[SDV] Life Sciences [q-bio] ,Protein Transport ,030104 developmental biology ,ral GTP-Binding Proteins ,lcsh:Q ,Guanine nucleotide exchange factor ,Signal transduction ,030217 neurology & neurosurgery ,Cell signalling ,Signal Transduction - Abstract
The monomeric GTPase RalB controls crucial physiological processes, including autophagy and invasion, but it still remains unclear how this multi-functionality is achieved. Previously, we reported that the RalGEF (Guanine nucleotide Exchange Factor) RGL2 binds and activates RalB to promote invasion. Here we show that RGL2, a major activator of RalB, is also required for autophagy. Using a novel automated image analysis method, Endomapper, we quantified the endogenous localization of the RGL2 activator and its substrate RalB at different endomembrane compartments, in an isogenic normal and Ras-transformed cell model. In both normal and Ras-transformed cells, we observed that RGL2 and RalB substantially localize at early and recycling endosomes, and to lesser extent at autophagosomes, but not at trans-Golgi. Interestingly the use of a FRET-based RalB biosensor indicated that RalB signaling is active at these endomembrane compartments at basal level in rich medium. Furthermore, induction of autophagy by nutrient starvation led to a considerable reduction of early and recycling endosomes, in contrast to the expected increase of autophagosomes, in both normal and Ras-transformed cells. However, autophagy mildly affected relative abundances of both RGL2 and RalB at early and recycling endosomes, and at autophagosomes. Interestingly, RalB activity increased at autophagosomes upon starvation in normal cells. These results suggest that the contribution of endosome membranes (carrying RGL2 and RalB molecules) increases total pool of RGL2-RalB at autophagosome forming compartments and might contribute to amplify RalB signaling to support autophagy.
- Published
- 2019
- Full Text
- View/download PDF
9. STK38 kinase acts as XPO1 gatekeeper regulating the nuclear export of autophagy proteins and other cargoes
- Author
-
Manish Kumar Singh, Joanna Lipecka, Gérard Zalcman, Jacques Camonis, Maria Carla Parrini, Brigitte Meunier, Nicolas Carpi, Maarten Jacquemyn, Patrice Codogno, Alexandre P. J. Martin, Alexander Hergovich, Cerina Chhuon, Matthieu Piel, Vasily N. Aushev, Ida Chiara Guerrera, and Dirk Daelemans
- Subjects
Receptors, Cytoplasmic and Nuclear ,Karyopherins ,Protein Serine-Threonine Kinases ,Biochemistry ,03 medical and health sciences ,XPO1 ,0302 clinical medicine ,Tandem Mass Spectrometry ,Protein Interaction Mapping ,Genetics ,Autophagy ,Humans ,Hippo Signaling Pathway ,Kinase activity ,Phosphorylation ,Nuclear export signal ,Protein kinase A ,Molecular Biology ,030304 developmental biology ,YAP1 ,Cell Nucleus ,0303 health sciences ,Hippo signaling pathway ,Chemistry ,Computational Biology ,Articles ,Cell biology ,Protein Transport ,Carrier Proteins ,030217 neurology & neurosurgery ,Chromatography, Liquid ,Protein Binding ,Signal Transduction - Abstract
STK38 (also known as NDR1) is a Hippo pathway serine/threonine protein kinase with multifarious functions in normal and cancer cells. Using a context-dependent proximity-labeling assay, we identify more than 250 partners of STK38 and find that STK38 modulates its partnership depending on the cellular context by increasing its association with cytoplasmic proteins upon nutrient starvation-induced autophagy and with nuclear ones during ECM detachment. We show that STK38 shuttles between the nucleus and the cytoplasm and that its nuclear exit depends on both XPO1 (aka exportin-1, CRM1) and STK38 kinase activity. We further uncover that STK38 modulates XPO1 export activity by phosphorylating XPO1 on serine 1055, thus regulating its own nuclear exit. We expand our model to other cellular contexts by discovering that XPO1 phosphorylation by STK38 regulates also the nuclear exit of Beclin1 and YAP1, key regulator of autophagy and transcriptional effector, respectively. Collectively, our results reveal STK38 as an activator of XPO1, behaving as a gatekeeper of nuclear export. These observations establish a novel mechanism of XPO1-dependent cargo export regulation by phosphorylation of XPO1's C-terminal auto-inhibitory domain.
- Published
- 2019
10. RalB directly triggers invasion downstream Ras by mobilizing the Wave complex
- Author
-
Marjorie Palmeri, Irina Veith, Ahmed El Marjou, Laetitia Fuhrmann, Nathalie Brandon, Mathieu Coppey, Giulia Zago, Saori Takaoka, Manish Kumar Singh, Maria Carla Parrini, Amanda Remorino, Simon de Beco, Frédérique Berger, Anne Vincent-Salomon, and Jacques Camonis
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Light ,QH301-705.5 ,Science ,Breast Neoplasms ,Exocyst ,GTPase ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,breast cancer ,Invasion ,Humans ,Neoplasm Invasiveness ,Wave ,Biology (General) ,optogenetics ,PI3K/AKT/mTOR pathway ,RALB ,General Immunology and Microbiology ,Effector ,General Neuroscience ,Cell Membrane ,Cell Biology ,General Medicine ,RALA ,Wiskott-Aldrich Syndrome Protein Family ,Cell biology ,Ral ,030104 developmental biology ,Disease Progression ,ras Proteins ,Medicine ,Female ,ral GTP-Binding Proteins ,Cell Surface Extensions ,Guanine nucleotide exchange factor ,Research Article ,Human ,Signal Transduction - Abstract
The two Ral GTPases, RalA and RalB, have crucial roles downstream Ras oncoproteins in human cancers; in particular, RalB is involved in invasion and metastasis. However, therapies targeting Ral signalling are not available yet. By a novel optogenetic approach, we found that light-controlled activation of Ral at plasma-membrane promotes the recruitment of the Wave Regulatory Complex (WRC) via its effector exocyst, with consequent induction of protrusions and invasion. We show that active Ras signals to RalB via two RalGEFs (Guanine nucleotide Exchange Factors), RGL1 and RGL2, to foster invasiveness; RalB contribution appears to be more important than that of MAPK and PI3K pathways. Moreover, on the clinical side, we uncovered a potential role of RalB in human breast cancers by determining that RalB expression at protein level increases in a manner consistent with progression toward metastasis. This work highlights the Ras-RGL1/2-RalB-exocyst-WRC axis as appealing target for novel anticancer strategies., eLife digest Cancers develop when cells in the body divide rapidly in an uncontroled manner. It is generally possible to cure cancers that remain contained within a small area. However, if the tumor cells start to move, the cancer may spread in the body and become life threatening. Currently, most of the anti-cancer treatments act to reduce the multiplication of these cells, but not their ability to migrate. A signal protein called Ras stimulates human cells to grow and move around. In healthy cells, the activity of Ras is tightly controled to ensure cells only divide and migrate at particular times, but in roughly 30% of all human cancers, Ras is abnormally active. Ras switches on another protein, named RalB, which is also involved in inappropriate cell migration. Yet, it is not clear how RalB is capable to help Ras trigger the migration of cells. Zago et al. used an approach called optogenetics to specifically activate the RalB protein in human cells using a laser that produces blue light. When activated, the light-controlled RalB started abnormal cell migration; this was used to dissect which molecules and mechanisms were involved in the process. Taken together, the experiments showed that, first, Ras ‘turns on’ RalB by changing the location of two proteins that control RalB. Then, the activated RalB regulates the exocyst, a group of proteins that travel within the cell. In turn, the exocyst recruits another group of proteins, named the Wave complex, which is part of the molecular motor required for cells to migrate. Zago et al. also found that, in patients, the RalB protein was present at abnormally high levels in samples of breast cancer cells that had migrated to another part of the body. Overall, these findings indicate that the role of RalB protein in human cancers is larger than previously thought, and they highlight a new pathway that could be a target for new anti-cancer drugs.
- Published
- 2018
11. A Biologist-Friendly Method to Analyze Cross-Correlation Between Protrusion Dynamics and Membrane Recruitment of Actin Regulators
- Author
-
Perrine, Paul-Gilloteaux, François, Waharte, Manish Kumar, Singh, and Maria Carla, Parrini
- Subjects
Membranes ,Cell Movement ,Humans ,Actins ,Cell Line - Abstract
During mesenchymal cell motility, various actin regulators are recruited to the leading edge with exquisite precision in time and space to generate protrusion and retraction cycles. We present here an automated method, named CorRecD (from Correlation Recruitment Dynamics), which quantifies cell edge dynamics, protein recruitment and analyze their cross-correlation. The Wave Regulatory Complex (WRC), a master driver of protrusions, is used as a case-of-study. This biologist-friendly method relies on free software tools and can be applied to any fluorescently tagged protein of interest.
- Published
- 2018
12. Fibroblast Heterogeneity and Immunosuppressive Environment in Human Breast Cancer
- Author
-
Yann Kieffer, Claire Bonneau, Inna Kuperstein, Anne Vincent-Salomon, Laetitia Fuhrmann, Ana Catarina Costa, Fatima Mechta-Grigoriou, Anne-Marie Givel, Vassili Soumelis, Ilaria Magagna, Maria Carla Parrini, Andrei Zinovyev, Alix Scholer-Dahirel, Melissa Cardon, Philémon Sirven, Brigitte Bourachot, Maria Kondratova, Charles Bernard, Floriane Pelon, Mechta-Grigoriou, Fatima, Equipements d'excellence - Equipement de biologie intégrative du cancer pour une médecine personnalisée - - ICGex2010 - ANR-10-EQPX-0003 - EQPX - VALID, Unité de génétique et biologie des cancers (U830), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Immunité et cancer (U932), Institut Curie [Paris], Cancer et génome: Bioinformatique, biostatistiques et épidémiologie d'un système complexe, Mines Paris - PSL (École nationale supérieure des mines de Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de Pathologie [Institut Curie], A.C. was supported by funding from the Fondation de France (2012-00034102) and the Fondation Pierre-Gilles de Gennes (FPGG0048). Y.K. was supported by the Institut National Du Cancer (INCa-DGOS-9963) and the Fondation pour la Recherche Médicale (FRM ING20130526797). M.C. was supported by the SiRIC-Curie program (INCa-DGOS-4654). The experimental work was supported by grants from the Institut National de la Santé et de la Recherche Médicale (Inserm), the Institut Curie, in particular the PIC TME and the PIC3i CAFi, the Ligue Nationale Contre le Cancer (Labelisation), the ICGex (ANR-10-EQPX-03), and INCa (INCa-DGOS-9963). We are very grateful to our funders for providing support these last years., ANR-10-EQPX-0003,ICGex,Equipement de biologie intégrative du cancer pour une médecine personnalisée(2010), and MINES ParisTech - École nationale supérieure des mines de Paris
- Subjects
0301 basic medicine ,regulatory T cell ,Cancer Research ,FOXP3 ,Regulatory T cell ,[SDV]Life Sciences [q-bio] ,Cellular differentiation ,T lymphocytes ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Breast Neoplasms ,Biology ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,stroma ,medicine ,Immune Tolerance ,Tumor Microenvironment ,Humans ,IL-2 receptor ,CAF ,Cell Proliferation ,Tumor microenvironment ,triple-negative ,breast cancers ,Cancer ,Cell Differentiation ,Forkhead Transcription Factors ,T lymphocyte ,Fibroblasts ,medicine.disease ,Treg ,[SDV] Life Sciences [q-bio] ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Cancer-Associated Fibroblasts ,heterogeneity - Abstract
Comment inBreast cancer: Fibroblast subtypes alter the microenvironment. [Nat Rev Clin Oncol. 2018]; International audience; Carcinoma-associated fibroblasts (CAF) are key players in the tumor microenvironment. Here, we characterize four CAF subsets in breast cancer with distinct properties and levels of activation. Two myofibroblastic subsets (CAF-S1, CAF-S4) accumulate differentially in triple-negative breast cancers (TNBC). CAF-S1 fibroblasts promote an immunosuppressive environment through a multi-step mechanism. By secreting CXCL12, CAF-S1 attracts CD4+CD25+ T lymphocytes and retains them by OX40L, PD-L2, and JAM2. Moreover, CAF-S1 increases T lymphocyte survival and promotes their differentiation into CD25HighFOXP3High, through B7H3, CD73, and DPP4. Finally, in contrast to CAF-S4, CAF-S1 enhances the regulatory T cell capacity to inhibit T effector proliferation. These data are consistent with FOXP3+ T lymphocyte accumulation in CAF-S1-enriched TNBC and show how a CAF subset contributes to immunosuppression.
- Published
- 2017
- Full Text
- View/download PDF
13. A family affair: A Ral-exocyst-centered network links Ras, Rac, Rho signaling to control cell migration
- Author
-
Jacques Camonis, Giulia Zago, Marco Biondini, and Maria Carla Parrini
- Subjects
rac1 GTP-Binding Protein ,animal structures ,Mini-Review - Commissioned ,Exocyst ,RAC1 ,GTPase ,Biology ,Biochemistry ,Exocytosis ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Neoplasms ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,RALB ,Effector ,GTPase-Activating Proteins ,Cell migration ,Cell Biology ,RALA ,Cell biology ,Neoplasm Proteins ,030220 oncology & carcinogenesis ,ras Proteins ,ral GTP-Binding Proteins ,Signal Transduction - Abstract
Cell migration is central to many developmental, physiologic and pathological processes, including cancer progression. The Ral GTPases (RalA and RalB) which act down-stream the Ras oncogenes, are key players in the coordination between membrane trafficking and actin polymerization. A major direct effector of Ral, the exocyst complex, works in polarized exocytosis and is at the center of multiple protein-protein interactions that support cell migration by promoting protrusion formation, front-rear polarization, and extra-cellular matrix degradation. In this review we describe the recent advancements in deciphering the molecular mechanisms underlying this role of Ral via exocyst on cell migration. Among others, we will discuss the recently identified cross-talk between Ral and Rac1 pathways: exocyst binds to a negative regulator (the RacGAP SH3BP1) and to the major effector (the Wave Regulatory Complex, WRC) of Rac1, the master regulator of protrusions. Next challenge will be to better characterize the dynamics in space and in time of these molecular interplays, to better understand the pleiotropic functions of Ral in both normal and cancer cells.
- Published
- 2017
14. Direct interaction between exocyst and Wave complexes promotes cell protrusions and motility
- Author
-
Maud Hertzog, Amel Sadou-Dubourgnoux, Jacques Camonis, Alexis Gautreau, Raphael Guerois, Giulia Zago, Marco Biondini, Perrine Paul-Gilloteaux, François Waharte, Melis D Arslanhan, Giorgio Scita, Etienne Formstecher, Maria Carla Parrini, Jinchao Yu, Institut Curie, Unité de génétique et biologie des cancers ( U830 ), Université Paris Descartes - Paris 5 ( UPD5 ) -Institut Curie-Institut National de la Santé et de la Recherche Médicale ( INSERM ), Compartimentation et dynamique cellulaires ( CDC ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -INSTITUT CURIE-Centre National de la Recherche Scientifique ( CNRS ), BioImaging Cell and Tissue Core Facility ( PICT-IBiSA ), INSTITUT CURIE, Mécanismes moléculaires du transport intracellulaire, Université Pierre et Marie Curie - Paris 6 ( UPMC ) -INSTITUT CURIE-Centre National de la Recherche Scientifique ( CNRS ) -Université Pierre et Marie Curie - Paris 6 ( UPMC ) -INSTITUT CURIE-Centre National de la Recherche Scientifique ( CNRS ), Hybrigenics [Paris], Hybrigenics, Institut de Biologie Intégrative de la Cellule ( I2BC ), Université Paris-Sud - Paris 11 ( UP11 ) -Commissariat à l'énergie atomique et aux énergies alternatives ( CEA ) -Université Paris-Saclay-Centre National de la Recherche Scientifique ( CNRS ), Laboratoire de Biochimie de l'Ecole polytechnique ( BIOC ), Centre National de la Recherche Scientifique ( CNRS ) -École polytechnique ( X ), Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Institut Curie [Paris], Unité de génétique et biologie des cancers (U830), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Compartimentation et dynamique cellulaires (CDC), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut Curie [Paris]-Centre National de la Recherche Scientifique (CNRS), BioImaging Cell and Tissue Core Facility (PICT-IBiSA), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut Curie [Paris]-Centre National de la Recherche Scientifique (CNRS)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut Curie [Paris]-Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Intégrative de la Cellule (I2BC), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Assemblage moléculaire et intégrité du génome (AMIG), Département Biochimie, Biophysique et Biologie Structurale (B3S), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Intégrative de la Cellule (I2BC), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Laboratoire de Biochimie de l'Ecole polytechnique (BIOC), École polytechnique (X)-Centre National de la Recherche Scientifique (CNRS), IFOM, Istituto FIRC di Oncologia Molecolare (IFOM), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5), Centre National de la Recherche Scientifique (CNRS)-Institut Curie [Paris]-Université Pierre et Marie Curie - Paris 6 (UPMC), Centre National de la Recherche Scientifique (CNRS)-Institut Curie [Paris]-Université Pierre et Marie Curie - Paris 6 (UPMC)-Centre National de la Recherche Scientifique (CNRS)-Institut Curie [Paris]-Université Pierre et Marie Curie - Paris 6 (UPMC), Centre National de la Recherche Scientifique ( CNRS ) -INSTITUT CURIE-Université Pierre et Marie Curie - Paris 6 ( UPMC ), Centre National de la Recherche Scientifique ( CNRS ) -INSTITUT CURIE-Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Centre National de la Recherche Scientifique ( CNRS ) -INSTITUT CURIE-Université Pierre et Marie Curie - Paris 6 ( UPMC ), École polytechnique ( X ) -Centre National de la Recherche Scientifique ( CNRS ), IFOM, Istituto FIRC di Oncologia Molecolare ( IFOM ), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS)-Institut Curie-Université Pierre et Marie Curie - Paris 6 (UPMC), Centre National de la Recherche Scientifique (CNRS)-Institut Curie-Université Pierre et Marie Curie - Paris 6 (UPMC)-Centre National de la Recherche Scientifique (CNRS)-Institut Curie-Université Pierre et Marie Curie - Paris 6 (UPMC), Université Paris-Sud - Paris 11 (UP11)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS)-Université Paris-Saclay, Université Paris-Sud - Paris 11 (UP11)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS)-Université Paris-Saclay-Université Paris-Sud - Paris 11 (UP11)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS)-Université Paris-Saclay-Institut de Biologie Intégrative de la Cellule (I2BC), and Université Paris-Sud - Paris 11 (UP11)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS)-Université Paris-Saclay-Université Paris-Sud - Paris 11 (UP11)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Centre National de la Recherche Scientifique (CNRS)-Université Paris-Saclay
- Subjects
0301 basic medicine ,[SDV]Life Sciences [q-bio] ,Cell ,Vesicular Transport Proteins ,Motility ,Exocyst ,Biology ,Exocytosis ,03 medical and health sciences ,Cell Movement ,medicine ,Humans ,Wave ,Actin ,Adaptor Proteins, Signal Transducing ,[ SDV ] Life Sciences [q-bio] ,Effector ,Rac1 gtpase ,Cell migration ,Cell Biology ,Wiskott-Aldrich Syndrome Protein Family ,Cell biology ,Ral ,Cytoskeletal Proteins ,Protein Subunits ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,Multiprotein Complexes ,Cell Surface Extensions ,Protein Binding - Abstract
International audience; Coordination between membrane trafficking and actin polymerization is fundamental in cell migration, but a dynamic view of the underlying molecular mechanisms is still missing. The Rac1 GTPase controls actin polymerization at protrusions by interacting with its effector, the Wave regulatory complex (WRC). The exocyst complex, which functions in polarized exocytosis, has been involved in the regulation of cell motility. Here, we show a physical and functional connection between exocyst and WRC. Purified components of exocyst and WRC directly associate in vitro, and interactions interfaces are identified. The exocyst-WRC interaction is confirmed in cells by co-immunoprecipitation and is shown to occur independently of the Arp2/3 complex. Disruption of the exocyst-WRC interaction leads to impaired migration. By using time-lapse microscopy coupled to image correlation analysis, we visualized the trafficking of the WRC towards the front of the cell in nascent protrusions. The exocyst is necessary for WRC recruitment at the leading edge and for resulting cell edge movements. This direct link between the exocyst and WRC provides a new mechanistic insight into the spatio-temporal regulation of cell migration.
- Published
- 2016
- Full Text
- View/download PDF
15. Dissecting Effects of Anti-cancer Drugs and Cancer-Associated Fibroblasts by On-Chip Reconstitution of Immunocompetent Tumor Microenvironments
- Author
-
Eugenio Martinelli, Luca Businaro, Vassili Soumelis, Ayako Yamada, Fanny Mermet-Meillon, Maria Carla Parrini, Weijing Han, Jacques Camonis, Floriane Pelon, Yasmine Khira, Giulia Fornabaio, Francesca Romana Bertani, Annamaria Gerardino, Gérard Zalcman, Stéphanie Descroix, Mélissande Cossutta, Marie Nguyen, Fatima Mechta-Grigoriou, Sophia S. Evans, Adele De Ninno, Davide Di Giuseppe, Philémon Sirven, Arianna Mencattini, Università degli Studi di Roma Tor Vergata [Roma], Croissance cellulaire, réparation et régénération tissulaires (CRRET), Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12)-Centre National de la Recherche Scientifique (CNRS), Immunité et cancer (U932), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie-Institut National de la Santé et de la Recherche Médicale (INSERM), Institute of Photonics and Nanotechnology, Consiglio Nazionale delle Ricerche [Roma] (CNR), Physico-Chimie-Curie (PCC), Centre National de la Recherche Scientifique (CNRS)-Institut Curie-Université Pierre et Marie Curie - Paris 6 (UPMC), Laboratoire d'Immunologie Clinique, Institut Curie, Institut Curie-Institut Curie, Service de pneumologie [CHU Caen], Université de Caen Normandie (UNICAEN), Normandie Université (NU)-Normandie Université (NU)-CHU Caen, Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Tumorothèque de Caen Basse-Normandie (TCBN), Département de Recherche Translationnelle (Plateforme BioPhenics), AP-HP Groupe Hospitalier Saint-Louis (Paris)-Institut Curie (Paris), Institut Curie-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS)-Institut Curie [Paris]-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Chimie du CNRS (INC), Institut Curie [Paris], Hopital Saint-Louis [AP-HP] (AP-HP), and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut Curie [Paris]
- Subjects
0301 basic medicine ,Receptor, ErbB-2 ,[SDV]Life Sciences [q-bio] ,medicine.medical_treatment ,Settore ING-INF/01 ,Cell Communication ,ErbB-2 ,Cancer-Associated Fibroblasts ,Trastuzumab ,Tumor Microenvironment ,skin and connective tissue diseases ,lcsh:QH301-705.5 ,Antibody-dependent cell-mediated cytotoxicity ,Tumor ,3. Good health ,tumor-on-chip ,HER2(+) breast cancer ,cancer-associated fibroblasts ,immunotherapy ,live cell imaging ,microfluidics ,organ-on-chip ,pre-clinical models ,trastuzumab ,tumor microenvironment ,Animals ,Antineoplastic Agents ,Cattle ,Cell Line, Tumor ,Human Umbilical Vein Endothelial Cells ,Humans ,Immunocompetence ,Neoplasm Invasiveness ,Stromal Cells ,Receptor ,medicine.drug ,Stromal cell ,cancer on chip ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Immune system ,medicine ,neoplasms ,Tumor microenvironment ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,030104 developmental biology ,lcsh:Biology (General) ,Cancer research ,organs on chip ,business - Abstract
Summary: A major challenge in cancer research is the complexity of the tumor microenvironment, which includes the host immunological setting. Inspired by the emerging technology of organ-on-chip, we achieved 3D co-cultures in microfluidic devices (integrating four cell populations: cancer, immune, endothelial, and fibroblasts) to reconstitute ex vivo a human tumor ecosystem (HER2+ breast cancer). We visualized and quantified the complex dynamics of this tumor-on-chip, in the absence or in the presence of the drug trastuzumab (Herceptin), a targeted antibody therapy directed against the HER2 receptor. We uncovered the capacity of the drug trastuzumab to specifically promote long cancer-immune interactions (>50 min), recapitulating an anti-tumoral ADCC (antibody-dependent cell-mediated cytotoxicity) immune response. Cancer-associated fibroblasts (CAFs) antagonized the effects of trastuzumab. These observations constitute a proof of concept that tumors-on-chip are powerful platforms to study ex vivo immunocompetent tumor microenvironments, to characterize ecosystem-level drug responses, and to dissect the roles of stromal components. : Inspired by the emerging technology of tumor-on-chip, Nguyen et al. reconstituted ex vivo a human tumor microenvironment (HER2+ breast cancer), characterized the ecosystem-level responses to the drug trastuzumab (Herceptin), and dissected the roles of stromal components (immune cells and fibroblasts), demonstrating the power of immunocompetent tumors-on-chip for preclinical drug studies. Keywords: tumor microenvironment, organ-on-chip, tumor-on-chip, trastuzumab, HER2+ breast cancer, cancer-associated fibroblasts, live cell imaging, microfluidics, pre-clinical models, immunotherapy
- Published
- 2018
- Full Text
- View/download PDF
16. Correction: Mitochondrial clearance by the STK38 kinase supports oncogenic Ras-induced cell transformation
- Author
-
Marta Gomez, Alexander Hergovich, Ramazan Gundogdu, Michael A. White, Didier Surdez, Giulia Zago, Audrey Bettoun, Maria Carla Parrini, Ilaria Cascone, Patrice Codogno, David Vallerand, Brigitte Meunier, Carine Joffre, Ahmad A.D. Sharif, and Jacques Camonis
- Subjects
Ubiquitin-Protein Ligases ,Transplantation, Heterologous ,Cell ,Mice, Nude ,Apoptosis ,Protein Serine-Threonine Kinases ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Cell Line, Tumor ,Autophagy ,medicine ,Animals ,Humans ,030304 developmental biology ,selective autophagy ,STK38 ,0303 health sciences ,cellular transformation ,Chemistry ,business.industry ,Kinase ,Mitophagy ,Correction ,Anoikis ,HCT116 Cells ,Transformation (genetics) ,Cell Transformation, Neoplastic ,HEK293 Cells ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,ras Proteins ,Cancer research ,Ras GTPase ,RNA Interference ,business ,Research Paper - Abstract
Oncogenic Ras signalling occurs frequently in many human cancers. However, no effective targeted therapies are currently available to treat patients suffering from Ras-driven tumours. Therefore, it is imperative to identify downstream effectors of Ras signalling that potentially represent promising new therapeutic options. Particularly, considering that autophagy inhibition can impair the survival of Ras-transformed cells in tissue culture and mouse models, an understanding of factors regulating the balance between autophagy and apoptosis in Ras-transformed human cells is needed. Here, we report critical roles of the STK38 protein kinase in oncogenic Ras transformation. STK38 knockdown impaired anoikis resistance, anchorage-independent soft agar growth, and in vivo xenograft growth of Ras-transformed human cells. Mechanistically, STK38 supports Ras-driven transformation through promoting detachment-induced autophagy. Even more importantly, upon cell detachment STK38 is required to sustain the removal of damaged mitochondria by mitophagy, a selective autophagic process, to prevent excessive mitochondrial reactive oxygen species production that can negatively affect cancer cell survival. Significantly, knockdown of PINK1 or Parkin, two positive regulators of mitophagy, also impaired anoikis resistance and anchorage-independent growth of Ras-transformed human cells, while knockdown of USP30, a negative regulator of PINK1/Parkin-mediated mitophagy, restored anchorage-independent growth of STK38-depleted Ras-transformed human cells. Therefore, our findings collectively reveal novel molecular players that determine whether Ras-transformed human cells die or survive upon cell detachment, which potentially could be exploited for the development of novel strategies to target Ras-transformed cells.
- Published
- 2018
- Full Text
- View/download PDF
17. Dissecting Activation of the PAK1 Kinase at Protrusions in Living Cells
- Author
-
Jean de Gunzburg, Maria Carla Parrini, Michiyuki Matsuda, and Jacques Camonis
- Subjects
rac1 GTP-Binding Protein ,Motility ,RAC1 ,Biosensing Techniques ,CDC42 ,Biology ,Cell Membrane Structures ,Models, Biological ,Biochemistry ,PAK1 ,Cell Movement ,Chlorocebus aethiops ,Fluorescence Resonance Energy Transfer ,Animals ,Humans ,Phosphorylation ,cdc42 GTP-Binding Protein ,Molecular Biology ,Actin ,COS cells ,Effector ,Mechanisms of Signal Transduction ,Cell Biology ,Rats ,Cell biology ,Enzyme Activation ,p21-Activated Kinases ,Cdc42 GTP-Binding Protein ,COS Cells - Abstract
The p21-activated kinase (PAK) 1 kinase, an effector of the Cdc42 and Rac1 GTPases, regulates cell protrusions and motility by controlling actin and adhesion dynamics. Its deregulation has been linked to human cancer. We show here that activation of PAK1 is necessary for protrusive activity during cell spreading. To investigate PAK1 activation dynamics at live protrusions, we developed a conformational biosensor, based on fluorescence resonance energy transfer. This novel PAK1 biosensor allowed the spatiotemporal visualization of PAK1 activation during spreading of COS-7 cells and during motility of normal rat kidney cells. By using this imaging approach in COS-7 cells, the following new insights on PAK1 regulation were unveiled. First, PAK1 acquires an intermediate semi-open conformational state upon recruitment to the plasma membrane. This semi-open PAK1 species is selectively autophosphorylated on serines in the N-terminal regulatory region but not on the critical threonine 423 in the catalytic site. Second, this intermediate PAK1 state is hypersensitive to stimulation by Cdc42 and Rac1. Third, interaction with PIX proteins contributes to PAK1 stimulation at membrane protrusions, in a GTPase-independent way. Finally, trans-phosphorylation events occur between PAK1 molecules at the membrane possibly playing a relevant role for its activation. This study leads to a model for the complex and accurate regulation of PAK1 kinase in vivo at cell protrusions.
- Published
- 2009
- Full Text
- View/download PDF
18. Cell Type-specific Regulation of RhoA Activity during Cytokinesis
- Author
-
Naoki Mochizuki, Hisayoshi Yoshizaki, Anne R. Bresnick, Maria Carla Parrini, Michiyuki Matsuda, Yusuke Ohba, and Natalya G. Dulyaninova
- Subjects
G2 Phase ,Botulinum Toxins ,DNA, Complementary ,Time Factors ,RHOA ,Myosin light-chain kinase ,Cell division ,Pyridines ,Green Fluorescent Proteins ,RAC1 ,Naphthalenes ,Septin ,Heterocyclic Compounds, 4 or More Rings ,Biochemistry ,Adenoviridae ,Cell Line ,Mice ,Multinucleate ,Proto-Oncogene Proteins ,Fluorescence Resonance Energy Transfer ,Animals ,Humans ,Molecular Biology ,Cytokinesis ,Genes, Dominant ,ADP Ribose Transferases ,biology ,Cell Membrane ,G1 Phase ,Azepines ,Cell Biology ,Amides ,Rats ,Cell biology ,Gene Expression Regulation ,Cell culture ,Mutation ,NIH 3T3 Cells ,biology.protein ,rhoA GTP-Binding Protein ,HeLa Cells ,Plasmids - Abstract
Rho family GTPases play pivotal roles in cytokinesis. By using probes based on the principle of fluorescence resonance energy transfer (FRET), we have shown that in HeLa cells RhoA activity increases with the progression of cytokinesis. Here we show that in Rat1A cells RhoA activity remained suppressed during most of the cytokinesis. Consistent with this observation, the expression of C3 toxin inhibited cytokinesis in HeLa cells but not in Rat1A cells. Furthermore, the expression of a dominant negative mutant of Ect2, a Rho GEF, or Y-27632, an inhibitor of the Rho-dependent kinase ROCK, inhibited cytokinesis in HeLa cells but not in Rat1A cells. In contrast to the activity of RhoA, the activity of Rac1 was suppressed during cytokinesis and started increasing at the plasma membrane of polar sides before the abscission of the daughter cells in both HeLa and Rat1A cells. This type of Rac1 suppression was shown to be essential for cytokinesis because a constitutively active mutant of Rac1 induced a multinucleated phenotype in both HeLa and Rat1A cells. Moreover, the involvement of MgcRacGAP/CYK-4 in this suppression of Rac1 during cytokinesis was shown by the use of a dominant negative mutant. Because ML-7, an inhibitor of myosin light chain kinase, delayed the cytokinesis of Rat1A cells and because Pak, a Rac1 effector, is known to suppress myosin light chain kinase, the suppression of the Rac1-Pak pathway by MgcRacGAP may play a pivotal role in the cytokinesis of Rat1A cells.
- Published
- 2004
- Full Text
- View/download PDF
19. Automated velocity mapping of migrating cell populations (AVeMap)
- Author
-
Maxime Deforet, Laurence Petitjean, Jacques Camonis, Pascal Silberzan, Marco Biondini, Maria Carla Parrini, Axel Buguin, Laboratoire Jean Perrin (LJP), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Curie [Paris], Laboratoire Physico-Chimie Curie [Institut Curie] (PCC), Institut Curie [Paris]-Institut de Chimie du CNRS (INC)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Génétique et expression des oncogènes, and Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
Dynamic mapping ,Computer science ,Computation ,Population ,Video microscopy ,Image processing ,Biochemistry ,Bottleneck ,Cell Line ,Computational science ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Velocity mapping ,Image Processing, Computer-Assisted ,Humans ,education ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Wound Healing ,0303 health sciences ,education.field_of_study ,Microscopy, Video ,business.industry ,Robotics ,Cell Biology ,Cell Tracking ,Biophysics ,Artificial intelligence ,business ,[PHYS.COND.CM-SCM]Physics [physics]/Condensed Matter [cond-mat]/Soft Condensed Matter [cond-mat.soft] ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Characterizing the migration of a population of cells remains laborious and somewhat subjective. Advances in genetics and robotics allow researchers to perform many experiments in parallel, but analyzing the large sets of data remains a bottleneck. Here we describe a rapid, fully automated correlation-based method for cell migration analysis, compatible with standard video microscopy. This method allows for the computation of quantitative migration parameters via an extensive dynamic mapping of cell displacements.
- Published
- 2012
- Full Text
- View/download PDF
20. Properties and regulation of the catalytic domain of Ira2p, Saccharomyces cerevisiae GTPase-activating protein of Ras2p
- Author
-
Alberto Bernardi, Michel Jacquet, Eric Jacquet, Andrea Parmeggiani, and Maria Carla Parrini
- Subjects
Saccharomyces cerevisiae Proteins ,GTPase-activating protein ,GTP' ,Recombinant Fusion Proteins ,Saccharomyces cerevisiae ,GTPase ,Biochemistry ,GTP Phosphohydrolases ,Fungal Proteins ,Proto-Oncogene Proteins p21(ras) ,Affinity chromatography ,Tubulin ,Humans ,chemistry.chemical_classification ,Binding Sites ,Neurofibromin 1 ,biology ,Chemistry ,GTPase-Activating Proteins ,Temperature ,Proteins ,Hydrogen-Ion Concentration ,biology.organism_classification ,Fusion protein ,Peptide Fragments ,Amino acid ,Kinetics ,ras GTPase-Activating Proteins ,ras Proteins ,biology.protein ,Electrophoresis, Polyacrylamide Gel - Abstract
This work describes the biochemical characterization of the catalytic domain of Ira2p, a Saccharomyces cerevisiae GTPase-activating protein (GAP) regulating the RAS gene products. A fragment of 383 residues (amino acids 1644-2026) was produced in Escherichia coli as glutathione S-transferase fusion protein (GST-Ira2p-383) and highly purified (> 90%) by affinity chromatography. The affinity of Ras2p for the GST-fused Ira2p-383 was 18 microM and the maximal stimulation of the Ras2p GTPase activity 6,000 times. The Ira2p activity was confirmed to be strictly specific for Ras2p, no stimulatory effect on human c-H-ras p21 GTPase being detectable. Comparison with the GAP-like domain of mammalian p120-GAP and neurofibromin using yeast Ras2p as substrate showed that Ira2p-383 has an affinity and turnover intermediary between GAP-334 and NF1-414. The activity of Ira2p-383 was strongly inhibited by monovalent and divalent salts. The simultaneous presence of the catalytic domains of Ira2p and the yeast GDP/GTP exchange factor Cdc25p induced on Ras2p a multiple-round reaction of GTP hydrolysis and GDP/GTP exchange, showing that it is possible to reconstitute in vitro a S. cerevisiae system suitable for the study of the regulation of the Ras2p GDP/GTP cycle. The tubulin partially inhibited (25%) the GAP activity of the Ira2p-383. A larger Ira2p catalytic fragment, Ira2p-505 (amino acids 1549-2053), that showed the same Km for Ras2p as Ira2p-383, was also inhibited by tubulin to the same extent but with a higher affinity than Ira2p-383.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1995
- Full Text
- View/download PDF
21. Oxidative stress promotes myofibroblast differentiation and tumour spreading
- Author
-
Anne Vincent-Salomon, Brigitte Bourachot, Olivier Delattre, Dorine Bellanger, Xavier Sastre-Garau, Marion Richardson, Guillem Rigaill, Carlo Lucchesi, Damien Gerald, Melissa Cardon, Marc-Henri Stern, Thierry Dubois, Gilles Despouy, Aurore Toullec, Maria Carla Parrini, Sylvain Lefort, Fatima Mechta-Grigoriou, Unité de génétique et biologie des cancers (U830), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5), Institut Curie [Paris], Institut National de la Santé et de la Recherche Médicale (INSERM), Service de Pathologie, Mechta-Grigoriou, Fatima, and ProdInra, Migration
- Subjects
Chemokine ,stromametastasis ,Proto-Oncogene Proteins c-jun ,Cellular differentiation ,[SDV]Life Sciences [q-bio] ,medicine.disease_cause ,Metastasis ,Mice ,0302 clinical medicine ,HIF‐1 ,Neoplasm Metastasis ,Research Articles ,Mice, Knockout ,chemistry.chemical_classification ,Microscopy ,0303 health sciences ,Histocytochemistry ,Incidence ,Cell Differentiation ,Immunohistochemistry ,SDF‐1 ,3. Good health ,[SDV] Life Sciences [q-bio] ,AP‐1 ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Myofibroblast ,Locomotion ,Breast Neoplasms ,Mammary Neoplasms, Animal ,Biology ,Models, Biological ,Cell Line ,SDF-1 ,03 medical and health sciences ,Stroma ,Proto-Oncogene Proteins ,stroma ,medicine ,Animals ,Humans ,metastasis ,Transcription factor ,030304 developmental biology ,Reactive oxygen species ,HIF-1 ,Fibroblasts ,Hypoxia-Inducible Factor 1, alpha Subunit ,AP-1 ,medicine.disease ,Survival Analysis ,Molecular biology ,Chemokine CXCL12 ,Oxidative Stress ,Microscopy, Fluorescence ,chemistry ,Cancer research ,biology.protein ,Reactive Oxygen Species ,Oxidative stress - Abstract
International audience; JunD regulates genes involved in antioxidant defence. We took advantage of the chronic oxidative stress resulting from junD deletion to examine the role of reactive oxygen species (ROS) in tumour development. In a model of mammary carcinogenesis, junD inactivation increased tumour incidence and revealed an associated reactive stroma. junD‐inactivation in the stroma was sufficient to shorten tumour‐free survival rate and enhance metastatic spread. ROS promoted conversion of fibroblasts into highly migrating myofibroblasts through accumulation of the hypoxia‐inducible factor (HIF)‐1α transcription factor and the CXCL12 chemokine. Accordingly, treatment with an antioxidant reduced the levels of HIF and CXCL12 and numerous myofibroblast features. CXCL12 accumulated in the stroma of HER2‐human breast adenocarcinomas. Moreover, HER2 tumours exhibited a high proportion of myofibroblasts, which was significantly correlated to nodal metastases. Interestingly, this subset of tumours exhibited a significant nuclear exclusion of JunD and revealed an associated oxido‐reduction signature, further demonstrating the relevance of our findings in human cancers. Collectively, our data uncover a new mechanism by which oxidative stress increases the migratory properties of stromal fibroblasts, which in turn potentiate tumour dissemination.
- Published
- 2010
- Full Text
- View/download PDF
22. Pak1 kinase homodimers are autoinhibited in trans and dissociated upon activation by Cdc42 and Rac1
- Author
-
Maria Carla Parrini, Ming Lei, Bruce J. Mayer, and Stephen C. Harrison
- Subjects
rac1 GTP-Binding Protein ,Molecular Sequence Data ,Cell Cycle Proteins ,GTPase ,CDC42 ,Biology ,Protein Serine-Threonine Kinases ,Ligands ,Transfection ,Cell Line ,PAK1 ,Guanine Nucleotide Exchange Factors ,Humans ,Amino Acid Sequence ,Phosphorylation ,cdc42 GTP-Binding Protein ,Molecular Biology ,Effector ,Cell Biology ,Actin cytoskeleton ,Cell biology ,Enzyme Activation ,Cdc42 GTP-Binding Protein ,p21-Activated Kinases ,Mutation ,Guanine nucleotide exchange factor ,Dimerization ,Rho Guanine Nucleotide Exchange Factors ,GTPase binding - Abstract
Pak1, a serine/threonine kinase that regulates the actin cytoskeleton, is an effector of the Rho family GTPases Cdc42 and Rac1. The crystal structure of Pak1 revealed an autoinhibited dimer that must dissociate upon GTPase binding. We show that Pak1 forms homodimers in vivo and that its dimerization is regulated by the intracellular level of GTP-Cdc42 or GTP-Rac1. The dimerized Pak1 adopts a trans-inhibited conformation: the N-terminal inhibitory portion of one Pak1 molecule in the dimer binds and inhibits the catalytic domain of the other. One GTPase interaction can result in activation of both partners. Another ligand, betaPIX, can stably associate with dimerized Pak1. Dimerization does not facilitate Pak1 trans-phosphorylation. We conclude that the functional significance of dimerization is to allow trans-inhibition.
- Published
- 2002
23. Determinants of Ras proteins specifying the sensitivity to yeast Ira2p and human p120-GAP
- Author
-
Andrea Parmeggiani, Maria Carla Parrini, and Alberto Bernardi
- Subjects
Models, Molecular ,Saccharomyces cerevisiae Proteins ,GTPase-activating protein ,Recombinant Fusion Proteins ,Saccharomyces cerevisiae ,Molecular Sequence Data ,GTPase ,In Vitro Techniques ,General Biochemistry, Genetics and Molecular Biology ,Fungal Proteins ,Proto-Oncogene Proteins p21(ras) ,In vivo ,Escherichia coli ,Humans ,Amino Acid Sequence ,Molecular Biology ,Genetics ,Binding Sites ,General Immunology and Microbiology ,biology ,General Neuroscience ,GTPase-Activating Proteins ,Proteins ,biology.organism_classification ,Yeast ,In vitro ,Cell biology ,ras GTPase-Activating Proteins ,Mutagenesis, Site-Directed ,ras Proteins ,Guanine nucleotide exchange factor ,Function (biology) ,Research Article - Abstract
Human and Saccharomyces cerevisiae Ras proteins and their regulators GAP (GTPase activating protein)and GEF (guanine nucleotide exchange factor) display structural similarities and are functionally interchangeable in vivo and in vitro, indicating that the molecular mechanism regulating Ras proteins has been conserved during evolution. As the only exceptions, the two S.cerevisiae GAPs, Ira1p and Ira2p, are strictly specific for yeast Ras proteins and cannot stimulate the GTPase of mammalian Ras. This study searches for the reasons for the different sensitivity to Ira2p of human H-ras p21 and yeast Ras2p. Construction of H-ras/Ras2p chimaeras showed that Gly18 of Ras2p (Ala11 of H-ras p21) is an important determinant for the specificity of Ira2p, revealing for the first time a function for this position. A second even more crucial determinant was found to be the 89-102 region of Ras2p (82-95 of H-ras p21) including the distal part of strand beta4, loop L6 and the proximal part of helix alpha3. It was possible to construct Ras2p's resistant to Ira2p but still sensitive to human p120-GAP and, conversely, a H-ras p21 sensitive to Ira2p. This work helps clarify specific aspects of the conserved molecular mechanism of interaction between Ras proteins and their negative GAP regulators.
- Published
- 1996
24. Engineering temperature-sensitive SH3 domains
- Author
-
Maria Carla Parrini and Bruce J. Mayer
- Subjects
animal structures ,DNA Mutational Analysis ,Molecular Sequence Data ,Clinical Biochemistry ,Protein domain ,Mutant ,Plasma protein binding ,temperature-sensitive mutation ,Biology ,Protein Engineering ,Polymerase Chain Reaction ,Biochemistry ,SH3 domain ,Homology (biology) ,Cell Line ,src Homology Domains ,Proto-Oncogene Proteins ,Drug Discovery ,Humans ,Amino Acid Sequence ,Molecular Biology ,Pharmacology ,Genetics ,Temperature ,Nck ,General Medicine ,Proto-Oncogene Proteins c-crk ,Yeast ,Cell biology ,SH2/SH3 adaptors ,Mutagenesis ,Molecular Medicine ,Signal transduction ,Carrier Proteins ,signal transduction ,Protein Binding ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Background: The ability to control specific protein-protein interactions conditionally in vivo would be extremely helpful for analyzing protein-protein interaction networks. SH3 (Src homology 3) modular protein binding domains are found in many signaling proteins and they play a crucial role in signal transduction by binding to proline-rich sequences. Results: Random in vitro mutagenesis coupled with yeast two-hybrid screening was used to identify mutations in the second SH3 domain of Nck that render interaction with its ligand temperature sensitive. Four of the mutants were functionally temperature sensitive in mammalian cells, where temperature sensitivity was correlated with a pronounced instability of the mutant domains at the nonpermissive temperature. Two of the mutations affect conserved residues in the hydrophobic core (Val133 and Val160), suggesting a general strategy for engineering temperature-sensitive SH3-containing proteins. Indeed mutagenesis of the corresponding positions in another SH3 domain, that of Crk-1, rendered the full-length Crk-1 protein temperature sensitive for function and stability in mammalian cells. Conclusions: Construction of temperature-sensitive SH3 domains is a novel approach to regulating the function of SH3 domains in vivo. Such mutants will be valuable in dissecting SH3-mediated signaling pathways. Furthermore, the methodology described here to isolate temperature-sensitive domains should be widely applicable to any domain involved in protein-protein interactions.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.