Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Leukemia ; 37(12): 2468-2478, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37821581

RESUMEN

Plasma cell disorders are clonal outgrowths of pre-malignant or malignant plasma cells, characterized by extensive chromosomal aberrations. Centrosome abnormalities are a major driver of chromosomal instability in cancer but their origin, incidence, and composition in primary tumor cells is poorly understood. Using cutting-edge, semi-automated high-throughput electron tomography, we characterized at nanoscale 1386 centrioles in CD138pos plasma cells from eight healthy donors and 21 patients with plasma cell disorders, and 722 centrioles from different control populations. In plasma cells from healthy individuals, over-elongated centrioles accumulated with age. In plasma cell disorders, centriole over-elongation was notably frequent in early, pre-malignant disease stages, became less pronounced in overt multiple myeloma, and almost entirely disappeared in aggressive plasma cell leukemia. Centrioles in other types of patient-derived B cell neoplasms showed no over-elongation. In contrast to current belief, centriole length appears to be highly variable in long-lived, healthy plasma cells, and over-elongation and structural aberrations are common in this cell type. Our data suggest that structural centrosome aberrations accumulate with age in healthy CD138pos plasma cells and may thus play an important role in early aneuploidization as an oncogenic driver in plasma cell disorders.


Asunto(s)
Centriolos , Células Plasmáticas , Humanos , Centriolos/metabolismo , Tomografía con Microscopio Electrónico , Centrosoma/metabolismo , Ciclo Celular
2.
Dev Cell ; 58(7): 616-632.e6, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-36990090

RESUMEN

3D cell cultures, in particular organoids, are emerging models in the investigation of healthy or diseased tissues. Understanding the complex cellular sociology in organoids requires integration of imaging modalities across spatial and temporal scales. We present a multi-scale imaging approach that traverses millimeter-scale live-cell light microscopy to nanometer-scale volume electron microscopy by performing 3D cell cultures in a single carrier that is amenable to all imaging steps. This allows for following organoids' growth, probing their morphology with fluorescent markers, identifying areas of interest, and analyzing their 3D ultrastructure. We demonstrate this workflow on mouse and human 3D cultures and use automated image segmentation to annotate and quantitatively analyze subcellular structures in patient-derived colorectal cancer organoids. Our analyses identify local organization of diffraction-limited cell junctions in compact and polarized epithelia. The continuum-resolution imaging pipeline is thus suited to fostering basic and translational organoid research by simultaneously exploiting the advantages of light and electron microscopy.


Asunto(s)
Técnicas de Cultivo Tridimensional de Células , Microscopía , Organoides , Animales , Humanos , Ratones , Técnicas de Cultivo Tridimensional de Células/métodos , Microscopía Electrónica , Organoides/diagnóstico por imagen , Organoides/fisiología , Organoides/ultraestructura , Neoplasias Colorrectales/patología
3.
Cancer Res ; 83(8): 1299-1314, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-36652557

RESUMEN

Crossing the blood-brain barrier is a crucial, rate-limiting step of brain metastasis. Understanding of the mechanisms of cancer cell extravasation from brain microcapillaries is limited as the underlying cellular and molecular processes cannot be adequately investigated using in vitro models and endpoint in vivo experiments. Using ultrastructural and functional imaging, we demonstrate that dynamic changes of activated brain microcapillaries promote the mandatory first steps of brain colonization. Successful extravasation of arrested cancer cells occurred when adjacent capillary endothelial cells (EC) entered into a distinct remodeling process. After extravasation, capillary loops were formed, which was characteristic of aggressive metastatic growth. Upon cancer cell arrest in brain microcapillaries, matrix-metalloprotease 9 (MMP9) was expressed. Inhibition of MMP2/9 and genetic perturbation of MMP9 in cancer cells, but not the host, reduced EC projections, extravasation, and brain metastasis outgrowth. These findings establish an active role of ECs in the process of cancer cell extravasation, facilitated by cross-talk between the two cell types. This extends our understanding of how host cells can contribute to brain metastasis formation and how to prevent it. SIGNIFICANCE: Tracking single extravasating cancer cells using multimodal correlative microscopy uncovers a brain seeding mechanism involving endothelial remodeling driven by cancer cell-derived MMP9, which might enable the development of approaches to prevent brain metastasis. See related commentary by McCarty, p. 1167.


Asunto(s)
Neoplasias Encefálicas , Endotelio Vascular , Humanos , Endotelio Vascular/patología , Células Endoteliales/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/patología , Línea Celular Tumoral
4.
Cell Rep Methods ; 2(11): 100322, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-36452870

RESUMEN

Electron microscopy is the gold standard to characterize centrosomal ultrastructure. However, production of significant morphometrical data is highly limited by acquisition time. We therefore developed a generalizable, semi-automated high-throughput electron tomography strategy to study centrosome aberrations in sparse patient-derived cancer cells at nanoscale. As proof of principle, we present electron tomography data on 455 centrioles of CD138pos plasma cells from one patient with relapsed/refractory multiple myeloma and CD138neg bone marrow mononuclear cells from three healthy donors as a control. Plasma cells from the myeloma patient displayed 122 over-elongated centrioles (48.8%). Particularly mother centrioles also harbored gross structural abnormalities, including fragmentation and disturbed microtubule cylinder formation, while control centrioles were phenotypically unremarkable. These data demonstrate the feasibility of our scalable high-throughput electron tomography strategy to study structural centrosome aberrations in primary tumor cells. Moreover, our electron tomography workflow and data provide a resource for the characterization of cell organelles beyond centrosomes.


Asunto(s)
Centriolos , Mieloma Múltiple , Humanos , Centriolos/patología , Mieloma Múltiple/diagnóstico por imagen , Tomografía con Microscopio Electrónico , Flujo de Trabajo , Centrosoma/ultraestructura
5.
J Cell Biol ; 220(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34160561

RESUMEN

Cells are 3D objects. Therefore, volume EM (vEM) is often crucial for correct interpretation of ultrastructural data. Today, scanning EM (SEM) methods such as focused ion beam (FIB)-SEM are frequently used for vEM analyses. While they allow automated data acquisition, precise targeting of volumes of interest within a large sample remains challenging. Here, we provide a workflow to target FIB-SEM acquisition of fluorescently labeled cells or subcellular structures with micrometer precision. The strategy relies on fluorescence preservation during sample preparation and targeted trimming guided by confocal maps of the fluorescence signal in the resin block. Laser branding is used to create landmarks on the block surface to position the FIB-SEM acquisition. Using this method, we acquired volumes of specific single cells within large tissues such as 3D cultures of mouse mammary gland organoids, tracheal terminal cells in Drosophila melanogaster larvae, and ovarian follicular cells in adult Drosophila, discovering ultrastructural details that could not be appreciated before.


Asunto(s)
Drosophila melanogaster/ultraestructura , Células de la Granulosa/ultraestructura , Glándulas Mamarias Animales/ultraestructura , Microscopía Electrónica de Rastreo/métodos , Coloración y Etiquetado/métodos , Células Tecales/ultraestructura , Tráquea/ultraestructura , Animales , Drosophila melanogaster/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Femenino , Expresión Génica , Genes Reporteros , Células de la Granulosa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Larva/metabolismo , Larva/ultraestructura , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Microscopía Electrónica de Rastreo/instrumentación , Organoides/metabolismo , Organoides/ultraestructura , Análisis de la Célula Individual/instrumentación , Análisis de la Célula Individual/métodos , Células Tecales/metabolismo , Tráquea/metabolismo , Flujo de Trabajo , Proteína Fluorescente Roja
6.
Blood ; 137(9): 1219-1232, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33270819

RESUMEN

Clinically relevant brain metastases (BMs) frequently form in cancer patients, with limited options for effective treatment. Circulating cancer cells must first permanently arrest in brain microvessels to colonize the brain, but the critical factors in this process are not well understood. Here, in vivo multiphoton laser-scanning microscopy of the entire brain metastatic cascade allowed unprecedented insights into how blood clot formation and von Willebrand factor (VWF) deposition determine the arrest of circulating cancer cells and subsequent brain colonization in mice. Clot formation in brain microvessels occurred frequently (>95%) and specifically at intravascularly arrested cancer cells, allowing their long-term arrest. An extensive clot embedded ∼20% of brain-arrested cancer cells, and those were more likely to successfully extravasate and form a macrometastasis. Mechanistically, the generation of tissue factor-mediated thrombin by cancer cells accounted for local activation of plasmatic coagulation in the brain. Thrombin inhibition by treatment with low molecular weight heparin or dabigatran and an anti-VWF antibody prevented clot formation, cancer cell arrest, extravasation, and the formation of brain macrometastases. In contrast, tumor cells were not able to directly activate platelets, and antiplatelet treatments did reduce platelet dispositions at intravascular cancer cells but did not reduce overall formation of BMs. In conclusion, our data show that plasmatic coagulation is activated early by intravascular tumor cells in the brain with subsequent clot formation, which led us to discover a novel and specific mechanism that is crucial for brain colonization. Direct or indirect thrombin and VWF inhibitors emerge as promising drug candidates for trials on prevention of BMs.


Asunto(s)
Coagulación Sanguínea , Neoplasias Encefálicas/sangre , Neoplasias de la Mama/patología , Melanoma/patología , Células Neoplásicas Circulantes/patología , Trombosis/sangre , Animales , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/patología , Neoplasias de la Mama/sangre , Neoplasias de la Mama/complicaciones , Puntos de Control del Ciclo Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Melanoma/sangre , Melanoma/complicaciones , Ratones , Trombosis/etiología , Trombosis/patología , Factor de von Willebrand/análisis
7.
Cell Death Dis ; 10(11): 817, 2019 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-31659149

RESUMEN

Intercellular communication within the bone marrow niche significantly promotes leukemogenesis and provides protection of leukemic cells from therapy. Secreted factors, intercellular transfer of mitochondria and the receptor-ligand interactions have been shown as mediators of this protection. Here we report that tunneling nanotubes (TNTs)-long, thin membranous structures, which have been identified as a novel mode of intercellular cross-talk-are formed in the presence of stroma and mediate transfer of cellular vesicles from stroma to leukemic cells. Importantly, transmission of vesicles via TNTs from stromal cells increases resistance of leukemic cells to the tyrosine kinase inhibitor, imatinib. Using correlative light-electron microscopy and electron tomography we show that stromal TNTs contain vesicles, provide membrane continuity with the cell bodies and can be open-ended. Moreover, trans-SILAC studies to reveal the non-autonomous proteome showed that specific sets of proteins are transferred together with cellular vesicles from stromal to leukemic cells, with a potential role in survival and adaptation. Altogether, our findings provide evidence for the biological role of the TNT-mediated vesicle exchange between stromal and leukemic cells, implicating the direct vesicle and protein transfer in the stroma-provided protection of leukemic cells.


Asunto(s)
Mesilato de Imatinib/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Mitocondrias/genética , Nanotubos/química , Transporte Biológico/genética , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Humanos , Mesilato de Imatinib/química , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Microscopía Electrónica , Mitocondrias/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/ultraestructura
8.
Cell Rep ; 27(12): 3602-3617.e5, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31216478

RESUMEN

The hepatitis C virus (HCV) is a major cause of chronic liver disease, affecting around 71 million people worldwide. Viral RNA replication occurs in a membranous compartment composed of double-membrane vesicles (DMVs), whereas virus particles are thought to form by budding into the endoplasmic reticulum (ER). It is unknown how these steps are orchestrated in space and time. Here, we established an imaging system to visualize HCV structural and replicase proteins in live cells and with high resolution. We determined the conditions for the recruitment of viral proteins to putative assembly sites and studied the dynamics of this event and the underlying ultrastructure. Most notable was the selective recruitment of ER membranes around lipid droplets where structural proteins and the viral replicase colocalize. Moreover, ER membranes wrapping lipid droplets were decorated with double membrane vesicles, providing a topological map of how HCV might coordinate the steps of viral replication and virion assembly.


Asunto(s)
Hepacivirus/fisiología , Hepatitis C/virología , Membranas Intracelulares/virología , Gotas Lipídicas/fisiología , Proteínas no Estructurales Virales/metabolismo , Ensamble de Virus , Replicación Viral , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Hepatitis C/genética , Hepatitis C/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Gotas Lipídicas/virología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , ARN Viral/análisis , ARN Viral/genética , Análisis Espacio-Temporal , Células Tumorales Cultivadas
9.
Dev Cell ; 49(1): 77-88.e7, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30880002

RESUMEN

Phagocytic immune cells such as microglia can engulf and process pathogens and dying cells with high efficiency while still maintaining their dynamic behavior and morphology. Effective intracellular processing of ingested cells is likely to be crucial for microglial function, but the underlying cellular mechanisms are poorly understood. Using both living fish embryos and mammalian macrophages, we show that processing depends on the shrinkage and packaging of phagosomes into a unique cellular compartment, the gastrosome, with distinct molecular and ultra-structural characteristics. Loss of the transporter Slc37a2 blocks phagosomal shrinkage, resulting in the expansion of the gastrosome and the dramatic bloating of the cell. This, in turn, affects the ability of microglia to phagocytose and migrate toward brain injuries. Thus, this work identifies a conserved crucial step in the phagocytic pathway of immune cells and provides a potential entry point for manipulating their behavior in development and disease.


Asunto(s)
Antiportadores/genética , Macrófagos/metabolismo , Proteínas de Transporte de Membrana/genética , Microglía/metabolismo , Fagosomas/ultraestructura , Animales , Apoptosis/genética , Compartimento Celular/genética , Células HeLa , Humanos , Macrófagos/ultraestructura , Ratones , Microglía/ultraestructura , Neuronas/metabolismo , Neuronas/ultraestructura , Fagocitos/ultraestructura , Fagocitosis/genética , Fagosomas/genética , Células RAW 264.7 , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
10.
Dev Cell ; 45(1): 33-52.e12, 2018 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-29634935

RESUMEN

Metastatic seeding is driven by cell-intrinsic and environmental cues, yet the contribution of biomechanics is poorly known. We aim to elucidate the impact of blood flow on the arrest and the extravasation of circulating tumor cells (CTCs) in vivo. Using the zebrafish embryo, we show that arrest of CTCs occurs in vessels with favorable flow profiles where flow forces control the adhesion efficacy of CTCs to the endothelium. We biophysically identified the threshold values of flow and adhesion forces allowing successful arrest of CTCs. In addition, flow forces fine-tune tumor cell extravasation by impairing the remodeling properties of the endothelium. Importantly, we also observe endothelial remodeling at arrest sites of CTCs in mouse brain capillaries. Finally, we observed that human supratentorial brain metastases preferably develop in areas with low perfusion. These results demonstrate that hemodynamic profiles at metastatic sites regulate key steps of extravasation preceding metastatic outgrowth.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Adhesión Celular , Hemodinámica , Neoplasias Pulmonares/patología , Melanoma/patología , Células Neoplásicas Circulantes/patología , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/metabolismo , Ciclo Celular , Circulación Cerebrovascular , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Melanoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Neoplásicas Circulantes/metabolismo , Estudios Retrospectivos , Células Tumorales Cultivadas , Pez Cebra
11.
Cell Rep ; 20(8): 1906-1920, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28834753

RESUMEN

Chromosomal instability is a hallmark of cancer and correlates with the presence of extra centrosomes, which originate from centriole overduplication. Overduplicated centrioles lead to the formation of centriole rosettes, which mature into supernumerary centrosomes in the subsequent cell cycle. While extra centrosomes promote chromosome missegregation by clustering into pseudo-bipolar spindles, the contribution of centriole rosettes to chromosome missegregation is unknown. We used multi-modal imaging of cells with conditional centriole overduplication to show that mitotic rosettes in bipolar spindles frequently harbor unequal centriole numbers, leading to biased chromosome capture that favors binding to the prominent pole. This results in chromosome missegregation and aneuploidy. Rosette mitoses lead to viable offspring and significantly contribute to progeny production. We further show that centrosome abnormalities in primary human malignancies frequently consist of centriole rosettes. As asymmetric centriole rosettes generate mitotic errors that can be propagated, rosette mitoses are sufficient to cause chromosome missegregation in cancer.


Asunto(s)
Centriolos/metabolismo , Inestabilidad Cromosómica/genética , Neoplasias/genética , Polos del Huso/metabolismo , Humanos , Neoplasias/metabolismo
12.
Methods Cell Biol ; 140: 277-301, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28528637

RESUMEN

Combining in vivo imaging with electron microscopy (EM) uniquely allows monitoring rare and critical events in living tissue, followed by their high-resolution visualization in their native context. A major hurdle, however, is to keep track of the region of interest (ROI) when moving from intravital microscopy (IVM) to EM. Here, we present a workflow that relies on correlating IVM and microscopic X-ray computed tomography to predict the position of the ROI inside the EM-processed sample. The ROI can then be accurately and quickly targeted using ultramicrotomy and imaged using EM. We outline how this procedure is used to retrieve and image tumor cells arrested in the vasculature of the mouse brain.


Asunto(s)
Imagenología Tridimensional , Microscopía Electrónica/métodos , Microtomografía por Rayos X , Animales , Línea Celular Tumoral , Humanos , Microscopía Intravital , Ratones , Resinas Sintéticas/química , Rayos X
13.
Trends Cell Biol ; 26(11): 848-863, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27515435

RESUMEN

Studying key biological events within complex model systems relies on dynamic and functional imaging at optimum spatial and temporal resolutions. Intravital correlative light and electron microscopy (intravital CLEM) combines imaging living multicellular model systems with electron microscopy, and offers full ultrastructural details of dynamic or transient events in vivo. However, routine use of intravital CLEM is hindered by multiple technological challenges faced when targeting a micron-size object (e.g., single cells or organelles) in a complex living organism. Recently, various approaches have been developed to overcome these limitations. In this review we outline the current methods and present the power of intravital CLEM in different fields of research. Finally, we describe approaches that will make intravital CLEM a routine, quantitative method for high-resolution cell biology in vivo.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Microscopía Intravital/métodos , Nanotecnología/métodos , Animales , Humanos , Microscopía Electrónica , Neoplasias/diagnóstico , Coloración y Etiquetado
14.
Acta Biomater ; 43: 348-357, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27427227

RESUMEN

UNLABELLED: Nanoparticles which surface adsorb proteins in an uncontrolled and non-reproducible manner will have limited uses as nanomedicinal products. A promising approach to avoid nanoparticle non-specific interactions with proteins is to design bio-hybrids by purposely pre-forming a protein corona around the inorganic cores. Here, we investigate, in vitro and in vivo, the newly acquired bio-identity of superparamagnetic iron oxide nanoparticles (SPIONs) upon their functionalization with a pre-formed and well-defined bovine serum albumin (BSA) corona. Cellular uptake, intracellular particle distribution and cytotoxicity were studied in two cell lines: adherent and non-adherent cells. BSA decreases nanoparticle internalization in both cell lines and protects the iron core once they have been internalized. The physiological response to the nanoparticles is then in vivo evaluated by oral administration to Caenorhabditis elegans, which was selected as a model of a functional intestinal barrier. Nanoparticle biodistribution, at single particle resolution, is studied by transmission electron microscopy. The analysis reveals that the acidic intestinal environment partially digests uncoated SPIONs but does not affect BSA-coated ones. It also discloses that some particles could enter the nematode's enterocytes, likely by endocytosis which is a different pathway than the one described for the worm nutrients. STATEMENT OF SIGNIFICANCE: Unravelling meaningful relationships between the physiological impact of engineered nanoparticles and their synthetic and biological identity is of vital importance when considering nanoparticles biomedical uses and when establishing their nanotoxicological profile. This study contributes to better comprehend the inorganic nanoparticles' behavior in real biological milieus. We synthesized a controlled pre-formed BSA protein corona on SPIONs to lower unspecific cell uptake and decrease nanoparticle fouling with other proteins. Such findings may be of relevance considering clinical translation and regulatory issues of inorganic nanoparticles. Moreover, we have advanced in the validation of C. elegans as a simple animal model for assessing biological responses of engineering nanomaterials. The physiological response of BSA coated SPIONs was evaluated in vivo after their oral administration to C. elegans. Analyzing ultra-thin cross-sections of the worms by TEM with single-particle precision, we could track NP biodistribution along the digestive tract and determine unambiguously their translocation through biological barriers and cell membranes.


Asunto(s)
Caenorhabditis elegans/metabolismo , Materiales Biocompatibles Revestidos/farmacología , Nanopartículas de Magnetita/química , Modelos Biológicos , Albúmina Sérica Bovina/farmacología , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/ultraestructura , Línea Celular Tumoral , Supervivencia Celular , Humanos , Hierro/metabolismo , Nanopartículas de Magnetita/ultraestructura
15.
J Cell Biol ; 212(7): 815-27, 2016 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-27002173

RESUMEN

Centriole elimination is an essential process that occurs in female meiosis of metazoa to reset centriole number in the zygote at fertilization. How centrioles are eliminated remains poorly understood. Here we visualize the entire elimination process live in starfish oocytes. Using specific fluorescent markers, we demonstrate that the two older, mother centrioles are selectively removed from the oocyte by extrusion into polar bodies. We show that this requires specific positioning of the second meiotic spindle, achieved by dynein-driven transport, and anchorage of the mother centriole to the plasma membrane via mother-specific appendages. In contrast, the single daughter centriole remaining in the egg is eliminated before the first embryonic cleavage. We demonstrate that these distinct elimination mechanisms are necessary because if mother centrioles are artificially retained, they cannot be inactivated, resulting in multipolar zygotic spindles. Thus, our findings reveal a dual mechanism to eliminate centrioles: mothers are physically removed, whereas daughters are eliminated in the cytoplasm, preparing the egg for fertilization.


Asunto(s)
Centriolos/fisiología , Meiosis , Oocitos/fisiología , Estrellas de Mar/fisiología , Animales , Centriolos/metabolismo , Fase de Segmentación del Huevo/fisiología , Dineínas/metabolismo , Femenino , Fertilización , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Oocitos/metabolismo , Cuerpos Polares/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Estrellas de Mar/genética , Estrellas de Mar/metabolismo , Factores de Tiempo
16.
J Cell Sci ; 129(2): 444-56, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26659665

RESUMEN

Intravital microscopy provides dynamic understanding of multiple cell biological processes, but its limited resolution has so far precluded structural analysis. Because it is difficult to capture rare and transient events, only a few attempts have been made to observe specific developmental and pathological processes in animal models using electron microscopy. The multimodal correlative approach that we propose here combines intravital microscopy, microscopic X-ray computed tomography and three-dimensional electron microscopy. It enables a rapid (c.a. 2 weeks) and accurate (<5 µm) correlation of functional imaging to ultrastructural analysis of single cells in a relevant context. We demonstrate the power of our approach by capturing single tumor cells in the vasculature of the cerebral cortex and in subcutaneous tumors, providing unique insights into metastatic events. Providing a significantly improved throughput, our workflow enables multiple sampling, a prerequisite for making correlative imaging a relevant tool to study cell biology in vivo. Owing to the versatility of this workflow, we envision broad applications in various fields of biological research, such as cancer or developmental biology.


Asunto(s)
Rastreo Celular/métodos , Animales , Encéfalo/irrigación sanguínea , Encéfalo/patología , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/secundario , Línea Celular Tumoral , Femenino , Microscopía Intravital , Ratones Desnudos , Microscopía Electrónica de Rastreo , Trasplante de Neoplasias , Microambiente Tumoral , Microtomografía por Rayos X
17.
Dev Cell ; 35(2): 186-98, 2015 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-26506308

RESUMEN

Nucleus positioning is key for intracellular organization, cell differentiation, and organ development and is affected in many diseases, including myopathies due to alteration in amphiphysin-2 (BIN1). The actin and microtubule cytoskeletons are essential for nucleus positioning, but their crosstalk in this process is sparsely characterized. Here, we report that impairment of amphiphysin/BIN1 in Caenorhabditis elegans, mammalian cells, or muscles from patients with centronuclear myopathy alters nuclear position and shape. We show that AMPH-1/BIN1 binds to nesprin and actin, as well as to the microtubule-binding protein CLIP170 in both species. Expression of the microtubule-anchoring CAP-GLY domain of CLIP170 fused to the nuclear-envelope-anchoring KASH domain of nesprin rescues nuclear positioning defects of amph-1 mutants. Amphiphysins thus play a central role in linking the nuclear envelope with the actin and microtubule cytoskeletons. We propose that BIN1 has a direct and evolutionarily conserved role in nuclear positioning, altered in myopathies.


Asunto(s)
Núcleo Celular/genética , Proteínas de Microfilamentos/genética , Proteínas Asociadas a Microtúbulos/genética , Miopatías Estructurales Congénitas/genética , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Membrana Nuclear/genética , Proteínas Nucleares/genética , Actinas/genética , Animales , Células COS , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Forma de la Célula/genética , Chlorocebus aethiops , Citoplasma/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Citoesqueleto/patología , Células HEK293 , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/genética , Microtúbulos/metabolismo , Complejos Multiproteicos , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo
18.
J Cell Biol ; 211(1): 27-37, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26459596

RESUMEN

Exosomes are secreted vesicles arising from the fusion of multivesicular bodies (MVBs) with the plasma membrane. Despite their importance in various processes, the molecular mechanisms controlling their formation and release remain unclear. Using nematodes and mammary tumor cells, we show that Ral GTPases are involved in exosome biogenesis. In Caenorhabditis elegans, RAL-1 localizes at the surface of secretory MVBs. A quantitative electron microscopy analysis of RAL-1-deficient animals revealed that RAL-1 is involved in both MVB formation and their fusion with the plasma membrane. These functions do not involve the exocyst complex, a common Ral guanosine triphosphatase (GTPase) effector. Furthermore, we show that the target membrane SNARE protein SYX-5 colocalizes with a constitutively active form of RAL-1 at the plasma membrane, and MVBs accumulate under the plasma membrane when SYX-5 is absent. In mammals, RalA and RalB are both required for the secretion of exosome-like vesicles in cultured cells. Therefore, Ral GTPases represent new regulators of MVB formation and exosome release.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/enzimología , Exosomas/metabolismo , Cuerpos Multivesiculares/metabolismo , Proteínas de Unión al GTP ral/fisiología , Animales , Caenorhabditis elegans/citología , Membrana Celular/enzimología , Fusión de Membrana , Transporte de Proteínas , Proteínas Qa-SNARE/metabolismo
19.
PLoS One ; 9(12): e114448, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25479106

RESUMEN

Correlative microscopy combines the advantages of both light and electron microscopy to enable imaging of rare and transient events at high resolution. Performing correlative microscopy in complex and bulky samples such as an entire living organism is a time-consuming and error-prone task. Here, we investigate correlative methods that rely on the use of artificial and endogenous structural features of the sample as reference points for correlating intravital fluorescence microscopy and electron microscopy. To investigate tumor cell behavior in vivo with ultrastructural accuracy, a reliable approach is needed to retrieve single tumor cells imaged deep within the tissue. For this purpose, fluorescently labeled tumor cells were subcutaneously injected into a mouse ear and imaged using two-photon-excitation microscopy. Using near-infrared branding, the position of the imaged area within the sample was labeled at the skin level, allowing for its precise recollection. Following sample preparation for electron microscopy, concerted usage of the artificial branding and anatomical landmarks enables targeting and approaching the cells of interest while serial sectioning through the specimen. We describe here three procedures showing how three-dimensional (3D) mapping of structural features in the tissue can be exploited to accurately correlate between the two imaging modalities, without having to rely on the use of artificially introduced markers of the region of interest. The methods employed here facilitate the link between intravital and nanoscale imaging of invasive tumor cells, enabling correlating function to structure in the study of tumor invasion and metastasis.


Asunto(s)
Microscopía Electrónica/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Neoplasias Experimentales/mortalidad , Neoplasias Experimentales/ultraestructura , Animales , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias
20.
PLoS One ; 8(4): e60813, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23577166

RESUMEN

Langerin is required for the biogenesis of Birbeck granules (BGs), the characteristic organelles of Langerhans cells. We previously used a Langerin-YFP fusion protein having a C-terminal luminal YFP tag to dynamically decipher the molecular and cellular processes which accompany the traffic of Langerin. In order to elucidate the interactions of Langerin with its trafficking effectors and their structural impact on the biogenesis of BGs, we generated a YFP-Langerin chimera with an N-terminal, cytosolic YFP tag. This latter fusion protein induced the formation of YFP-positive large puncta. Live cell imaging coupled to a fluorescence recovery after photobleaching approach showed that this coalescence of proteins in newly formed compartments was static. In contrast, the YFP-positive structures present in the pericentriolar region of cells expressing Langerin-YFP chimera, displayed fluorescent recovery characteristics compatible with active membrane exchanges. Using correlative light-electron microscopy we showed that the coalescent structures represented highly organized stacks of membranes with a pentalaminar architecture typical of BGs. Continuities between these organelles and the rough endoplasmic reticulum allowed us to identify the stacks of membranes as a form of "Organized Smooth Endoplasmic Reticulum" (OSER), with distinct molecular and physiological properties. The involvement of homotypic interactions between cytoplasmic YFP molecules was demonstrated using an A206K variant of YFP, which restored most of the Langerin traffic and BG characteristics observed in Langerhans cells. Mutation of the carbohydrate recognition domain also blocked the formation of OSER. Hence, a "double-lock" mechanism governs the behavior of YFP-Langerin, where asymmetric homodimerization of the YFP tag and homotypic interactions between the lectin domains of Langerin molecules participate in its retention and the subsequent formation of BG-like OSER. These observations confirm that BG-like structures appear wherever Langerin accumulates and confirm that membrane trafficking effectors dictate their physiology and, illustrate the importance of molecular interactions in the architecture of intracellular membranes.


Asunto(s)
Retículo Endoplásmico/metabolismo , Células de Langerhans/citología , Lectinas Tipo C/genética , Proteínas Luminiscentes/genética , Proteínas Recombinantes de Fusión/genética , Transporte Biológico , Línea Celular Tumoral , Membrana Celular/metabolismo , Retículo Endoplásmico/ultraestructura , Expresión Génica , Humanos , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/metabolismo , Mutación , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA