Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell Rep Med ; 5(5): 101549, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38703767

RESUMEN

There is a compelling need for approaches to predict the efficacy of immunotherapy drugs. Tumor-on-chip technology exploits microfluidics to generate 3D cell co-cultures embedded in hydrogels that recapitulate simplified tumor ecosystems. Here, we present the development and validation of lung tumor-on-chip platforms to quickly and precisely measure ex vivo the effects of immune checkpoint inhibitors on T cell-mediated cancer cell death by exploiting the power of live imaging and advanced image analysis algorithms. The integration of autologous immunosuppressive FAP+ cancer-associated fibroblasts impaired the response to anti-PD-1, indicating that tumors-on-chips are capable of recapitulating stroma-dependent mechanisms of immunotherapy resistance. For a small cohort of non-small cell lung cancer patients, we generated personalized tumors-on-chips with their autologous primary cells isolated from fresh tumor samples, and we measured the responses to anti-PD-1 treatment. These results support the power of tumor-on-chip technology in immuno-oncology research and open a path to future clinical validations.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias Pulmonares , Medicina de Precisión , Receptor de Muerte Celular Programada 1 , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/inmunología , Medicina de Precisión/métodos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Dispositivos Laboratorio en un Chip , Inmunoterapia/métodos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Línea Celular Tumoral
2.
Lab Chip ; 23(18): 3906-3935, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37592893

RESUMEN

Over the past 15 years, the field of oncology research has witnessed significant progress in the development of new cell culture models, such as tumor-on-chip (ToC) systems. In this comprehensive overview, we present a multidisciplinary perspective by bringing together physicists, biologists, clinicians, and experts from pharmaceutical companies to highlight the current state of ToC research, its unique features, and the challenges it faces. To offer readers a clear and quantitative understanding of the ToC field, we conducted an extensive systematic analysis of more than 300 publications related to ToC from 2005 to 2022. ToC offer key advantages over other in vitro models by enabling precise control over various parameters. These parameters include the properties of the extracellular matrix, mechanical forces exerted on cells, the physico-chemical environment, cell composition, and the architecture of the tumor microenvironment. Such fine control allows ToC to closely replicate the complex microenvironment and interactions within tumors, facilitating the study of cancer progression and therapeutic responses in a highly representative manner. Importantly, by incorporating patient-derived cells or tumor xenografts, ToC models have demonstrated promising results in terms of clinical validation. We also examined the potential of ToC for pharmaceutical industries in which ToC adoption is expected to occur gradually. Looking ahead, given the high failure rate of clinical trials and the increasing emphasis on the 3Rs principles (replacement, reduction, refinement of animal experimentation), ToC models hold immense potential for cancer research. In the next decade, data generated from ToC models could potentially be employed for discovering new therapeutic targets, contributing to regulatory purposes, refining preclinical drug testing and reducing reliance on animal models.


Asunto(s)
Técnicas de Cultivo de Célula , Neoplasias , Humanos , Animales , Industria Farmacéutica , Matriz Extracelular , Microambiente Tumoral , Neoplasias/tratamiento farmacológico
3.
Lab Chip ; 22(22): 4443-4455, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36314259

RESUMEN

In vitro cell cultures are most often performed in unphysiological hyperoxia since the oxygen partial pressure of conventional incubators is set at 141 mmHg (18.6%, close to ambient air oxygen 20.1%). This value is higher than human tissue oxygen levels, as the in vivo oxygen partial pressures range from 104 mmHg (lung alveoli) to 8 mmHg (skin epidermis). Importantly, under pathological conditions such as cancer, cells can experience oxygen pressure lower than the healthy tissue. Although hypoxic incubators can regulate gas oxygen, they do not take into account the dissolved oxygen concentration in the cell culture medium. In the context of organ on chip and micro-physiological system development, we present here a new system, called Oxalis (OXygen ALImentation System) that allows fine control of the dissolved oxygen level in the cell culture medium. Oxalis regulates simultaneously the gas composition and the inlet reservoir pressure by modulating the pneumatic valve opening. This dual regulation allows both the pressure driven liquid flowrate and the level of oxygen dissolved in the chip to be controlled independently. Oxalis offers unprecedented features such as an oxygen equilibration time lower than 3 minutes and an accuracy of 3 mmHg. These performances can be reached for chip perfusion flow as low as 1 µL min-1. This low flow rate allows the shear stress experienced by the cells in the chip to be accurately controlled. In addition, the system enables modulation of the pH in the cell culture medium through the modulation of CO2. The fine control and monitoring of both O2 and pH pave the way for new precise investigations on physiological and pathological biological processes. Using Oxalis in the context of tumor-on-chip, we demonstrate the capacity of the system to recapitulate hypoxia-induced gene expression, offering an innovative strategy for future studies on the role of hypoxia in malignant progression and drug resistance.


Asunto(s)
Neoplasias , Oxígeno , Humanos , Hipoxia , Técnicas de Cultivo de Célula , Perfusión
4.
Biosens Bioelectron ; 215: 114571, 2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-35932554

RESUMEN

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.


Asunto(s)
Técnicas Biosensibles , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Microambiente Tumoral , Virus Vaccinia
5.
Cells ; 10(10)2021 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-34685617

RESUMEN

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.


Asunto(s)
Autofagia , Movimiento Celular , Genes ras , Oncogenes , Animales , Autofagia/genética , Bovinos , Línea Celular , Movimiento Celular/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Colágeno/farmacología , Geles/farmacología , Humanos , Procesamiento de Imagen Asistido por Computador , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo
6.
Med Image Anal ; 72: 102124, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34157611

RESUMEN

Biological experiments based on organ-on-chips (OOCs) exploit light Time-Lapse Microscopy (TLM) for a direct observation of cell movement that is an observable signature of underlying biological processes. A high spatial resolution is essential to capture cell dynamics and interactions from recorded experiments by TLM. Unfortunately, due to physical and cost limitations, acquiring high resolution videos is not always possible. To overcome the problem, we present here a new deep learning-based algorithm that extends the well-known Deep Image Prior (DIP) to TLM Video Super Resolution without requiring any training. The proposed Recursive Deep Prior Video method introduces some novelties. The weights of the DIP network architecture are initialized for each of the frames according to a new recursive updating rule combined with an efficient early stopping criterion. Moreover, the DIP loss function is penalized by two different Total Variation-based terms. The method has been validated on synthetic, i.e., artificially generated, as well as real videos from OOC experiments related to tumor-immune interaction. The achieved results are compared with several state-of-the-art trained deep learning Super Resolution algorithms showing outstanding performances.


Asunto(s)
Microscopía , Redes Neurales de la Computación , Algoritmos , Procesamiento de Imagen Asistido por Computador , Imagen de Lapso de Tiempo
7.
PLoS Comput Biol ; 17(3): e1008870, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33784299

RESUMEN

The emerging tumor-on-chip (ToC) approaches allow to address biomedical questions out of reach with classical cell culture techniques: in biomimetic 3D hydrogels they partially reconstitute ex vivo the complexity of the tumor microenvironment and the cellular dynamics involving multiple cell types (cancer cells, immune cells, fibroblasts, etc.). However, a clear bottleneck is the extraction and interpretation of the rich biological information contained, sometime hidden, in the cell co-culture videos. In this work, we develop and apply novel video analysis algorithms to automatically measure the cytotoxic effects on human cancer cells (lung and breast) induced either by doxorubicin chemotherapy drug or by autologous tumor-infiltrating cytotoxic T lymphocytes (CTL). A live fluorescent dye (red) is used to selectively pre-stain the cancer cells before co-cultures and a live fluorescent reporter for caspase activity (green) is used to monitor apoptotic cell death. The here described open-source computational method, named STAMP (spatiotemporal apoptosis mapper), extracts the temporal kinetics and the spatial maps of cancer death, by localizing and tracking cancer cells in the red channel, and by counting the red to green transition signals, over 2-3 days. The robustness and versatility of the method is demonstrated by its application to different cell models and co-culture combinations. Noteworthy, this approach reveals the strong contribution of primary cancer-associated fibroblasts (CAFs) to breast cancer chemo-resistance, proving to be a powerful strategy to investigate intercellular cross-talks and drug resistance mechanisms. Moreover, we defined a new parameter, the 'potential of death induction', which is computed in time and in space to quantify the impact of dying cells on neighbor cells. We found that, contrary to natural death, cancer death induced by chemotherapy or by CTL is transmissible, in that it promotes the death of nearby cancer cells, suggesting the release of diffusible factors which amplify the initial cytotoxic stimulus.


Asunto(s)
Apoptosis/fisiología , Técnicas de Cocultivo/métodos , Linfocitos T Citotóxicos , Microambiente Tumoral/fisiología , Línea Celular Tumoral , Biología Computacional , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Cinética , Técnicas Analíticas Microfluídicas , Microscopía por Video , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/fisiología
8.
Biomaterials ; 269: 120624, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33421710

RESUMEN

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.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Mama , Animales , Biomimética , Neoplasias Óseas/patología , Huesos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Humanos , Metástasis de la Neoplasia/patología , Osteoblastos/patología , Microambiente Tumoral
9.
Nat Commun ; 11(1): 404, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31964880

RESUMEN

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 TGFß 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.


Asunto(s)
Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/metabolismo , Metástasis Linfática/patología , Miofibroblastos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Axila , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Fibroblastos Asociados al Cáncer/patología , Proliferación Celular , Separación Celular , Quimiocina CXCL12/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Citometría de Flujo , Estudios de Seguimiento , Humanos , Estimación de Kaplan-Meier , Ganglios Linfáticos/citología , Ganglios Linfáticos/patología , Persona de Mediana Edad , Miofibroblastos/patología , Invasividad Neoplásica/patología , Cultivo Primario de Células , Pronóstico , Supervivencia sin Progresión , Receptores Notch/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral
10.
EMBO Rep ; 20(11): e48150, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31544310

RESUMEN

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.


Asunto(s)
Autofagia , Núcleo Celular/metabolismo , Carioferinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Portadoras/metabolismo , Cromatografía Liquida , Biología Computacional/métodos , Vía de Señalización Hippo , Humanos , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Transducción de Señal , Espectrometría de Masas en Tándem , Proteína Exportina 1
11.
Sci Rep ; 9(1): 11797, 2019 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-31395941

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Sci Rep ; 9(1): 8910, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31222145

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Transducción de Señal , Proteínas de Unión al GTP ral/metabolismo , Compartimento Celular , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Transporte de Proteínas , Proteínas de Unión al GTP ral/fisiología
13.
Small GTPases ; 10(5): 323-330, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-28498728

RESUMEN

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.


Asunto(s)
Movimiento Celular , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Proteínas ras/metabolismo , Animales , Proteínas Activadoras de GTPasa , Humanos , Neoplasias/patología
14.
Cell Rep ; 25(13): 3884-3893.e3, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30590056

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Fibroblastos Asociados al Cáncer/patología , Inmunocompetencia/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Animales , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Bovinos , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Invasividad Neoplásica , Receptor ErbB-2/metabolismo , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Trastuzumab/farmacología
15.
Elife ; 72018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30320548

RESUMEN

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.


Asunto(s)
Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Proteínas ras/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/efectos de la radiación , Progresión de la Enfermedad , Femenino , Humanos , Luz , Invasividad Neoplásica , Optogenética , Transducción de Señal
17.
Methods Mol Biol ; 1749: 279-289, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29526004

RESUMEN

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.


Asunto(s)
Movimiento Celular/fisiología , Membranas/metabolismo , Actinas/metabolismo , Línea Celular , Humanos
18.
Cancer Cell ; 33(3): 463-479.e10, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29455927

RESUMEN

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.


Asunto(s)
Fibroblastos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Neoplasias de la Mama/inmunología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Factores de Transcripción Forkhead/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Activación de Linfocitos/fisiología
19.
Cell Rep ; 21(7): 1922-1935, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-29141223

RESUMEN

Rac1 is a small RhoGTPase switch that orchestrates actin branching in space and time and protrusion/retraction cycles of the lamellipodia at the cell front during mesenchymal migration. Biosensor imaging has revealed a graded concentration of active GTP-loaded Rac1 in protruding regions of the cell. Here, using single-molecule imaging and super-resolution microscopy, we show an additional supramolecular organization of Rac1. We find that Rac1 partitions and is immobilized into nanoclusters of 50-100 molecules each. These nanoclusters assemble because of the interaction of the polybasic tail of Rac1 with the phosphoinositide lipids PIP2 and PIP3. The additional interactions with GEFs and possibly GAPs, downstream effectors, and other partners are responsible for an enrichment of Rac1 nanoclusters in protruding regions of the cell. Our results show that subcellular patterns of Rac1 activity are supported by gradients of signaling nanodomains of heterogeneous molecular composition, which presumably act as discrete signaling platforms.


Asunto(s)
Microdominios de Membrana/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Células 3T3 , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Ratones , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Imagen Individual de Molécula/métodos , Factores de Transcripción/metabolismo
20.
Lab Chip ; 16(24): 4691-4701, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27797384

RESUMEN

We report here a simple yet robust transient compartmentalization system for microfluidic platforms. Cylindrical microfilaments made of commercially available fishing lines are embedded in a microfluidic chamber and employed as removable walls, dividing the chamber into several compartments. These partitions allow tight sealing for hours, and can be removed at any time by longitudinal sliding with minimal hydrodynamic perturbation. This allows the easy implementation of various functions, previously impossible or requiring more complex instrumentation. In this study, we demonstrate the applications of our strategy, firstly to trigger chemical diffusion, then to make surface co-coating or cell co-culture on a two-dimensional substrate, and finally to form multiple cell-laden hydrogel compartments for three-dimensional cell co-culture in a microfluidic device. This technology provides easy and low-cost solutions, without the use of pneumatic valves or external equipment, for constructing well-controlled microenvironments for biochemical and cellular assays.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Técnicas de Cultivo de Célula/instrumentación , Dispositivos Laboratorio en un Chip , Animales , Hidrogeles/química , Ratones , Neuroglía/citología , Neuronas/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA