Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Mol Cell ; 64(3): 549-564, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27746021

RESUMEN

In advanced stages of cancers, TGF-ß promotes tumor progression in conjunction with inputs from receptor tyrosine kinase pathways. However, mechanisms that underpin the signaling cooperation and convert TGF-ß from a potent growth inhibitor to a tumor promoter are not fully understood. We report here that TGF-ß directly regulates alternative splicing of cancer stem cell marker CD44 through a phosphorylated T179 of SMAD3-mediated interaction with RNA-binding protein PCBP1. We show that TGF-ß and EGF respectively induce SMAD3 and PCBP1 to colocalize in SC35-positive nuclear speckles, and the two proteins interact in the variable exon region of CD44 pre-mRNA to inhibit spliceosome assembly in favor of expressing the mesenchymal isoform CD44s over the epithelial isoform CD44E. We further show that the SMAD3-mediated alternative splicing is essential to the tumor-promoting role of TGF-ß and has a global influence on protein products of genes instrumental to epithelial-to-mesenchymal transition and metastasis.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas/genética , Receptores de Hialuranos/genética , Neoplasias Pulmonares/genética , Proteína smad3/genética , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Exones , Femenino , Perfilación de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Proteínas de Unión al ARN , Transducción de Señal , Proteína smad3/metabolismo , Treonina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
2.
Am J Pathol ; 192(12): 1699-1711, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36063900

RESUMEN

Wound healing is a highly conserved process that restores the integrity and functionality of injured tissues. Transforming growth factor (TGF)-ß is a master regulator of wound healing, whose signaling is attenuated by the E3 ubiquitin ligase Smurf2. Herein, the roles of Smurf2 in cutaneous wound healing were examined using a murine incisional cutaneous model. Loss of Smurf2 increased early inflammation in the wounds and led to narrower wounds with greater breaking strength. Loss of Smurf2 also led to more linearized collagen bundles in normal and wounded skin. Gene expression analyses by real-time quantitative PCR indicated that Smurf2-deficient fibroblasts had increased levels of TGF-ß/Smad3 signaling and changes in expression profile of genes related to matrix turnover. The effect of Smurf2 loss on wound healing and collagen bundling was attenuated by the heterozygous loss of Smad3. Together, these results show that Smurf2 affects inflammation and collagen processing in cutaneous wounds by down-regulating TGF-ß/Smad3 signaling.


Asunto(s)
Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta , Ratones , Animales , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Colágeno , Cicatrización de Heridas , Inflamación , Factores de Crecimiento Transformadores
3.
Proc Natl Acad Sci U S A ; 117(44): 27423-27434, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33060293

RESUMEN

Localization of RNAs at protrusive regions of cells is important for single-cell migration on two-dimensional surfaces. Protrusion-enriched RNAs encode factors linked to cancer progression, such as the RAB13 GTPase and the NET1 guanine nucleotide exchange factor, and are regulated by the tumor-suppressor protein APC. However, tumor cells in vivo often do not move as single cells but rather utilize collective modes of invasion and dissemination. Here, we developed an inducible system of three-dimensional (3D) collective invasion to study the behavior and importance of protrusion-enriched RNAs. We find that, strikingly, both the RAB13 and NET1 RNAs are enriched specifically at the invasive front of leader cells in invasive cell strands. This localization requires microtubules and coincides with sites of high laminin concentration. Indeed, laminin association and integrin engagement are required for RNA accumulation at the invasive front. Importantly, perturbing RNA accumulation reduces collective 3D invasion. Examination of in vivo tumors reveals a similar localization of the RAB13 and NET1 RNAs at potential invasive sites, suggesting that this mechanism could provide a targeting opportunity for interfering with collective cancer cell invasion.


Asunto(s)
Movimiento Celular/genética , Invasividad Neoplásica/genética , Neoplasias/patología , ARN Mensajero/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Células HeLa , Humanos , Hibridación Fluorescente in Situ , Microscopía Intravital , Ratones , Microscopía Confocal , Invasividad Neoplásica/prevención & control , Neoplasias/genética , Proteínas Oncogénicas/genética , ARN Interferente Pequeño , Esferoides Celulares , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión al GTP rab/genética
4.
J Cell Sci ; 131(6)2018 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-29487177

RESUMEN

Many actin filaments in animal cells are co-polymers of actin and tropomyosin. In many cases, non-muscle myosin II associates with these co-polymers to establish a contractile network. However, the temporal relationship of these three proteins in the de novo assembly of actin filaments is not known. Intravital subcellular microscopy of secretory granule exocytosis allows the visualisation and quantification of the formation of an actin scaffold in real time, with the added advantage that it occurs in a living mammal under physiological conditions. We used this model system to investigate the de novo assembly of actin, tropomyosin Tpm3.1 (a short isoform of TPM3) and myosin IIA (the form of non-muscle myosin II with its heavy chain encoded by Myh9) on secretory granules in mouse salivary glands. Blocking actin polymerization with cytochalasin D revealed that Tpm3.1 assembly is dependent on actin assembly. We used time-lapse imaging to determine the timing of the appearance of the actin filament reporter LifeAct-RFP and of Tpm3.1-mNeonGreen on secretory granules in LifeAct-RFP transgenic, Tpm3.1-mNeonGreen and myosin IIA-GFP (GFP-tagged MYH9) knock-in mice. Our findings are consistent with the addition of tropomyosin to actin filaments shortly after the initiation of actin filament nucleation, followed by myosin IIA recruitment.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Tropomiosina/metabolismo , Citoesqueleto de Actina/genética , Actinas/genética , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Cadenas Pesadas de Miosina , Miosina Tipo IIA no Muscular/genética , Unión Proteica , Vesículas Secretoras/genética , Vesículas Secretoras/metabolismo , Tropomiosina/genética
5.
Bioessays ; 40(9): e1800032, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30080263

RESUMEN

The remodeling of biological membranes is crucial for a vast number of cellular activities and is an inherently multiscale process in both time and space. Seminal work has provided important insights into nanometer-scale membrane deformations, and highlighted the remarkable variation and complexity in the underlying molecular machineries and mechanisms. However, how membranes are remodeled at the micron-scale, particularly in vivo, remains poorly understood. Here, we discuss how using regulated exocytosis of large (1.5-2.0 µm) membrane-bound secretory granules in the salivary gland of live mice as a model system, has provided evidence for the importance of the actomyosin cytoskeleton in micron-scale membrane remodeling in physiological conditions. We highlight some of these advances, and present mechanistic hypotheses for how the various biochemical and biophysical properties of distinct actomyosin networks may drive this process.


Asunto(s)
Actomiosina/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Animales , Exocitosis/fisiología , Glándulas Salivales/metabolismo , Vesículas Secretoras/metabolismo
7.
J Dairy Sci ; 102(3): 2760-2782, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30471915

RESUMEN

Milk fat comprises membrane-coated droplets of neutral lipid, which constitute the predominant source of lipids for survival of the suckling neonate. From the perspective of the dairy industry, they are the basis for the manufacture of butter and essential ingredients in the production of cheese, yogurt, and specialty dairy produce. To provide mechanistic insight into the assembly and secretion of lipid droplets during lactation, we developed novel intravital imaging techniques using transgenic mice, which express fluorescently tagged marker proteins. The number 4 mammary glands were surgically prepared under a deep plane of anesthesia and the exposed glands positioned as a skin flap with intact vascular supply on the stage of a laser-scanning confocal microscope. Lipid droplets were stained by prior exposure of the glands to hydrophobic fluorescent BODIPY (boron-dipyrromethene) dyes and their formation and secretion monitored by time-lapse subcellular microscopy over periods of 1 to 2 h. Droplets were transported to the cell apex by directed (superdiffusive) motion at relatively slow and intermittent rates (0-2 µm/min). Regardless of size, droplets grew by numerous fusion events during transport and as they were budding from the cell enveloped by apical membranes. Surprisingly, droplet secretion was not constitutive but required an injection of oxytocin to induce contraction of the myoepithelium with subsequent release of droplets into luminal spaces. These novel results are discussed in the context of the current paradigm for milk fat synthesis and secretion and as a template for future innovations in the dairy industry.


Asunto(s)
Metabolismo de los Lípidos , Leche/metabolismo , Animales , Membrana Celular , Femenino , Microscopía Intravital , Lactancia/metabolismo , Gotas Lipídicas , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Transgénicos , Oxitocina/metabolismo
8.
Blood ; 127(9): 1085-96, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26647393

RESUMEN

Extravascular fibrin deposition accompanies many human diseases and causes chronic inflammation and organ damage, unless removed in a timely manner. Here, we used intravital microscopy to investigate how fibrin is removed from extravascular space. Fibrin placed into the dermis of mice underwent cellular endocytosis and lysosomal targeting, revealing a novel intracellular pathway for extravascular fibrin degradation. A C-C chemokine receptor type 2 (CCR2)-positive macrophage subpopulation constituted the majority of fibrin-uptaking cells. Consequently, cellular fibrin uptake was diminished by elimination of CCR2-expressing cells. The CCR2-positive macrophage subtype was different from collagen-internalizing M2-like macrophages. Cellular fibrin uptake was strictly dependent on plasminogen and plasminogen activator. Surprisingly, however, fibrin endocytosis was unimpeded by the absence of the fibrin(ogen) receptors, αMß2 and ICAM-1, the myeloid cell integrin-binding site on fibrin or the endocytic collagen receptor, the mannose receptor. The study identifies a novel fibrin endocytic pathway engaged in extravascular fibrin clearance and shows that interstitial fibrin and collagen are cleared by different subsets of macrophages employing distinct molecular pathways.


Asunto(s)
Endocitosis , Fibrina/metabolismo , Macrófagos/metabolismo , Receptores CCR2/metabolismo , Animales , Bioensayo , Receptor 1 de Quimiocinas CX3C , Proliferación Celular , Fibrinolisina/metabolismo , Ratones , Células Mieloides/metabolismo , Plasminógeno/metabolismo , Activadores Plasminogénicos/metabolismo , Proteolisis , Receptores de Quimiocina/metabolismo , Receptores de Péptidos/metabolismo
9.
Methods ; 128: 3-11, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28780320

RESUMEN

Head and neck squamous cell carcinoma is one of the most common cancers with a 50% 5-year survival rate. Understanding the mechanisms that control development, progression, and spreading of the tumor to distal sites is of paramount importance to develop effective therapies. Here, we describe a minimally invasive procedure, which enables performing intravital microscopy of the mouse tongue in models for oral cancer and provides structural and dynamic information of the tumors at cellular and subcellular level.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico por imagen , Neoplasias de Cabeza y Cuello/diagnóstico por imagen , Microscopía Intravital/métodos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Línea Celular Tumoral , Femenino , Técnicas de Sustitución del Gen/métodos , Humanos , Ratones , Ratones Desnudos , Ratones SCID
10.
Hepatology ; 64(4): 1317-29, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27396550

RESUMEN

UNLABELLED: Liver kinase B1 (LKB1) and its downstream effector AMP-activated protein kinase (AMPK) play critical roles in polarity establishment by regulating membrane trafficking and energy metabolism. In collagen sandwich-cultured hepatocytes, loss of LKB1 or AMPK impaired apical ABCB11 (Bsep) trafficking and bile canalicular formation. In the present study, we used liver-specific (albumin-Cre) LKB1 knockout mice (LKB1(-/-) ) to investigate the role of LKB1 in the maintenance of functional tight junction (TJ) in vivo. Transmission electron microscopy examination revealed that hepatocyte apical membrane with microvilli substantially extended into the basolateral domain of LKB1(-/-) livers. Immunofluorescence studies revealed that loss of LKB1 led to longer and wider canalicular structures correlating with mislocalization of the junctional protein, cingulin. To test junctional function, we used intravital microscopy to quantify the transport kinetics of 6-carboxyfluorescein diacetate (6-CFDA), which is processed in hepatocytes into its fluorescent derivative 6-carboxyfluorescein (6-CF) and secreted into the canaliculi. In LKB1(-/-) mice, 6-CF remained largely in hepatocytes, canalicular secretion was delayed, and 6-CF appeared in the blood. To test whether 6-CF was transported through permeable TJ, we intravenously injected low molecular weight (3 kDa) dextran in combination with 6-CFDA. In wild-type mice, 3 kDa dextran remained in the vasculature, whereas it rapidly appeared in the abnormal bile canaliculi in LKB1(-/-) mice, confirming that junctional disruption resulted in paracellular exchange between the blood stream and the bile canaliculus. CONCLUSION: LKB1 plays a critical role in regulating the maintenance of TJ and paracellular permeability, which may explain how various drugs, chemicals, and metabolic states that inhibit the LKB1/AMPK pathway result in cholestasis. (Hepatology 2016;64:1317-1329).


Asunto(s)
Hepatocitos/fisiología , Hepatocitos/ultraestructura , Proteínas Serina-Treonina Quinasas/fisiología , Uniones Estrechas/fisiología , Uniones Estrechas/ultraestructura , Proteínas Quinasas Activadas por AMP , Animales , Femenino , Masculino , Ratones , Ratones Noqueados
11.
Exp Eye Res ; 158: 161-170, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27179411

RESUMEN

The promise of revolutionary insights into intraocular pressure (IOP) and aqueous humor outflow homeostasis, IOP pathogenesis, and novel therapy offered by engineered mouse models has been hindered by a lack of appropriate tools for studying the aqueous drainage tissues in their original 3-dimensional (3D) environment. Advances in 2-photon excitation fluorescence imaging (TPEF) combined with availability of modalities such as transgenic reporter mice and intravital dyes have placed us on the cusp of unlocking the potential of the mouse model for unearthing insights into aqueous drainage structure and function. Multimodality 2-photon imaging permits high-resolution visualization not only of tissue structural organization but also cells and cellular function. It is possible to dig deeper into understanding the cellular basis of aqueous outflow regulation as the technique integrates analysis of tissue structure, cell biology and physiology in a way that could also lead to fresh insights into human glaucoma. We outline recent novel applications of two-photon imaging to analyze the mouse conventional drainage system in vivo or in whole tissues: (1) collagen second harmonic generation (SHG) identifies the locations of episcleral vessels, intrascleral plexuses, collector channels, and Schlemm's canal in the distal aqueous drainage tract; (2) the prospero homeobox protein 1-green fluorescent protein (GFP) reporter helps locate the inner wall of Schlemm's canal; (3) Calcein AM, siGLO™, the fluorescent reporters m-Tomato and GFP, and coherent anti-Stokes scattering (CARS), are adjuncts to TPEF to identify live cells by their membrane or cytosolic locations; (4) autofluorescence and sulforhodamine-B to identify elastic fibers in the living eye. These tools greatly expand our options for analyzing physiological and pathological processes in the aqueous drainage tissues of live mice as a model of the analogous human system.


Asunto(s)
Humor Acuoso/diagnóstico por imagen , Glaucoma/diagnóstico por imagen , Limbo de la Córnea/diagnóstico por imagen , Malla Trabecular/diagnóstico por imagen , Animales , Humor Acuoso/metabolismo , Colorantes Fluorescentes/metabolismo , Glaucoma/metabolismo , Humanos , Presión Intraocular/fisiología , Limbo de la Córnea/metabolismo , Ratones , Microscopía de Fluorescencia por Excitación Multifotónica , Malla Trabecular/metabolismo
12.
Proc Natl Acad Sci U S A ; 111(14): 5254-9, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706872

RESUMEN

Multifocal structured illumination microscopy (MSIM) provides a twofold resolution enhancement beyond the diffraction limit at sample depths up to 50 µm, but scattered and out-of-focus light in thick samples degrades MSIM performance. Here we implement MSIM with a microlens array to enable efficient two-photon excitation. Two-photon MSIM gives resolution-doubled images with better sectioning and contrast in thick scattering samples such as Caenorhabditis elegans embryos, Drosophila melanogaster larval salivary glands, and mouse liver tissue.


Asunto(s)
Iluminación , Microscopía/métodos , Fotones , Animales , Caenorhabditis elegans/embriología , Drosophila melanogaster/crecimiento & desarrollo , Larva/química , Hígado/química , Ratones
13.
FASEB J ; 29(3): 1056-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25466898

RESUMEN

Multiple human malignancies rely on C-X-C motif chemokine receptor type 4 (CXCR4) and its ligand, SDF-1/CXCL12 (stroma cell-derived factor 1/C-X-C motif chemokine 12), to metastasize. CXCR4 inhibitors promote the mobilization of bone marrow stem cells, limiting their clinical application for metastasis prevention. We investigated the CXCR4-initiated signaling circuitry to identify new potential therapeutic targets. We used HeLa human cancer cells expressing high levels of CXCR4 endogenously. We found that CXCL12 promotes their migration in Boyden chamber assays and single cell tracking. CXCL12 activated mTOR (mechanistic target of rapamycin) potently in a pertussis-sensitive fashion. Inhibition of mTOR complex 1 (mTORC1) by rapamycin [drug concentration causing 50% inhibition (IC50) = 5 nM] and mTORC1/mTORC2 by Torin2 (IC50 = 6 nM), or by knocking down key mTORC1/2 components, Raptor and Rictor, respectively, decreased directional cell migration toward CXCL12. We developed a CXCR4-mediated spontaneous metastasis model by implanting HeLa cells in the tongue of SCID-NOD mice, in which 80% of the animals develop lymph node metastasis. It is surprising that mTORC1 disruption by Raptor knockdown was sufficient to reduce tumor growth by 60% and spontaneous metastasis by 72%, which were nearly abolished by rapamycin. In contrast, disrupting mTORC2 had no effect in tumor growth or metastasis compared with control short hairpin RNAs. These data suggest that mTORC1 may represent a suitable therapeutic target in human malignancies using CXCR4 for their metastatic spread. .


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/metabolismo , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Complejos Multiproteicos/metabolismo , Receptores CXCR4/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias del Cuello Uterino/secundario , Animales , Apoptosis , Western Blotting , Proliferación Celular , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transducción de Señal , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/metabolismo
14.
Exp Cell Res ; 337(2): 219-25, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26160452

RESUMEN

In this mini-review we focus on the use of time-lapse light microscopy to study membrane remodeling during protein secretion in live animals. In particular, we highlight how subcellular intravital microscopy has enabled imaging the dynamics of both individual secretory vesicles and the plasma membrane, during different steps in the exocytic process. This powerful approach has provided us with the unique opportunity to unravel the role of the actin cytoskeleton in regulating this process under physiological conditions, and to overcome the shortcomings of more reductionist model systems.


Asunto(s)
Citoesqueleto de Actina/química , Membrana Celular/química , Exocitosis/fisiología , Imagen de Lapso de Tiempo/métodos , Animales , Ratones
15.
Proc Natl Acad Sci U S A ; 109(31): E2101-9, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22753477

RESUMEN

Mammalian pigmentation is driven by the intercellular transfer of pigment-containing melanosomes from the tips of melanocyte dendrites to surrounding keratinocytes. Tip accumulation of melanosomes requires myosin Va, because melanosomes concentrate in the center of melanocytes from myosin Va-null (dilute) mice. This distribution defect results in inefficient melanosome transfer and a dilution of coat color. Dilute mice that simultaneously lack melanoregulin, the product of the dilute suppressor locus, exhibit a nearly complete restoration of coat color, but, surprisingly, melanosomes remain concentrated in the center of their melanocytes. Here we show that dilute/dsu melanocytes, but not dilute melanocytes, readily transfer the melanosomes concentrated in their center to surrounding keratinocytes in situ. Using time-lapse imaging of WT melanocyte/keratinocyte cocultures in which the plasma membranes of the two cells are marked with different colors, we define an intercellular melanosome transfer pathway that involves the shedding by the melanocyte of melanosome-rich packages, which subsequently are phagocytosed by the keratinocyte. Shedding, which occurs primarily at dendritic tips but also from more central regions, involves adhesion to the keratinocyte, thinning behind the forming package, and apparent self-abscission. Finally, we show that shedding from the cell center is sixfold more frequent in cultured dilute/dsu melanocytes than in dilute melanocytes, consistent with the in situ data. Together, these results explain how dsu restores the coat color of dilute mice without restoring intracellular melanosome distribution, indicate that melanoregulin is a negative regulator of melanosome transfer, and provide insight into the mechanism of intercellular melanosome transfer.


Asunto(s)
Proteínas Portadoras/metabolismo , Queratinocitos/metabolismo , Melanocitos/metabolismo , Melanosomas/metabolismo , Pigmentación de la Piel/fisiología , Proteínas Adaptadoras del Transporte Vesicular , Animales , Transporte Biológico Activo/fisiología , Proteínas Portadoras/genética , Células Cultivadas , Técnicas de Cocultivo , Péptidos y Proteínas de Señalización Intracelular , Queratinocitos/citología , Melanocitos/citología , Melanosomas/genética , Ratones , Ratones Mutantes , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/genética , Miosina Tipo V/metabolismo
16.
Traffic ; 13(5): 627-34, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22243493

RESUMEN

Regulated exocytosis is a fundamental process that every secretory cell uses to deliver molecules to the cell surface and the extracellular space by virtue of membranous carriers. This process has been extensively studied using various approaches such as biochemistry, electrophysiology and electron microscopy. However, recent developments in time-lapse light microscopy have made possible imaging individual exocytic events, hence, advancing our understanding of this process at a molecular level. In this review, we focus on intravital microscopy (IVM), a light microscopy-based approach that enables imaging subcellular structures in live animals, and discuss its recent application to study regulated exocytosis. IVM has revealed differences in regulation and modality of regulated exocytosis between in vitro and in vivo model systems, unraveled novel aspects of this process that can be appreciated only in in vivo settings and provided valuable and novel information on its molecular machinery. In conclusion, we make the case for IVM being a mature technique that can be used to investigate the molecular machinery of several intracellular events under physiological conditions.


Asunto(s)
Exocitosis , Microscopía/métodos , Actinas/metabolismo , Animales , Citoesqueleto/metabolismo , Diagnóstico por Imagen/métodos , Electrofisiología/métodos , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Microscopía Confocal/métodos , Microscopía Electrónica/métodos , Modelos Biológicos , Neuronas/metabolismo , Ratas
17.
Cell Mol Life Sci ; 70(12): 2099-121, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22986507

RESUMEN

Regulated exocytosis is the main mechanism utilized by specialized secretory cells to deliver molecules to the cell surface by virtue of membranous containers (i.e., secretory vesicles). The process involves a series of highly coordinated and sequential steps, which include the biogenesis of the vesicles, their delivery to the cell periphery, their fusion with the plasma membrane, and the release of their content into the extracellular space. Each of these steps is regulated by the actin cytoskeleton. In this review, we summarize the current knowledge regarding the involvement of actin and its associated molecules during each of the exocytic steps in vertebrates, and suggest that the overall role of the actin cytoskeleton during regulated exocytosis is linked to the architecture and the physiology of the secretory cells under examination. Specifically, in neurons, neuroendocrine, endocrine, and hematopoietic cells, which contain small secretory vesicles that undergo rapid exocytosis (on the order of milliseconds), the actin cytoskeleton plays a role in pre-fusion events, where it acts primarily as a functional barrier and facilitates docking. In exocrine and other secretory cells, which contain large secretory vesicles that undergo slow exocytosis (seconds to minutes), the actin cytoskeleton plays a role in post-fusion events, where it regulates the dynamics of the fusion pore, facilitates the integration of the vesicles into the plasma membrane, provides structural support, and promotes the expulsion of large cargo molecules.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/fisiología , Exocitosis/fisiología , Fusión de Membrana/fisiología , Vías Secretoras/fisiología , Vesículas Secretoras/fisiología , Humanos , Modelos Biológicos , Miosinas/metabolismo
18.
Proc Natl Acad Sci U S A ; 108(33): 13552-7, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21808006

RESUMEN

The regulation and the dynamics of membrane trafficking events have been studied primarily in in vitro models that often do not fully reflect the functional complexity found in a living multicellular organism. Here we used intravital microscopy in the salivary glands of live rodents to investigate regulated exocytosis, a fundamental process in all of the secretory organs. We found that ß-adrenergic stimulation elicits exocytosis of large secretory granules, which gradually collapse with the apical plasma membrane without any evidence of compound exocytosis, as was previously described. Furthermore, we show that the driving force required to complete the collapse of the granules is provided by the recruitment of F-actin and nonmuscle myosin II on the granule membranes that is triggered upon fusion with the plasma membrane. Our results provide information on the machinery controlling regulated secretion and show that intravital microscopy provides unique opportunities to address fundamental questions in cell biology under physiological conditions.


Asunto(s)
Actomiosina/fisiología , Exocitosis , Microscopía Confocal , Actinas/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Membrana Celular , Polaridad Celular , Exocitosis/efectos de los fármacos , Ratones , Ratones Transgénicos , Miosina Tipo IIA no Muscular , Transporte de Proteínas , Glándulas Salivales , Vesículas Secretoras/metabolismo
19.
bioRxiv ; 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-38076999

RESUMEN

PAX3/7 Fusion-negative rhabdomyosarcoma (FN-RMS) is a childhood mesodermal lineage malignancy with a poor prognosis for metastatic or relapsed cases. Towards achieving a more complete understanding of advanced FN-RMS, we developed an orthotopic tongue xenograft model for studies of molecular basis of FN-RMS invasion and metastasis. The behavior of FN-RMS cells injected into murine tongue was examined using in vivo bioluminescence imaging, non-invasive intravital microscopy (IVM), and histopathology and compared to the prevailing hindlimb intramuscular and subcutaneous xenografts. FN-RMS cells were retained in the tongue and invaded locally into muscle mysial spaces and vascular lumen. While evidence of hematogenous dissemination to the lungs occurred in tongue and intramuscular xenografts, evidence of local invasion and lymphatic dissemination to lymph nodes only occurred in tongue xenografts. IVM and RNA-seq of tongue xenografts reveal shifts in cellular phenotype and differentiation state in tongue xenografts. IVM also shows homing to blood and lymphatic vessels, lymphatic intravasation, and dynamic membrane protrusions. Based on these findings, the tongue orthotopic xenograft of FN-RMS is a valuable model for tumor progression studies at the tissue, cellular and subcellular levels providing insight into kinetics and molecular bases of tumor invasion and metastasis and, hence, new therapeutic avenues for advanced FN-RMS.

20.
bioRxiv ; 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38168275

RESUMEN

Membrane remodeling drives a broad spectrum of cellular functions, and it is regulated through mechanical forces exerted on the membrane by cytoplasmic complexes. Here, we investigate how actin filaments dynamically tune their structure to control the active transfer of membranes between cellular compartments with distinct compositions and biophysical properties. Using intravital subcellular microscopy in live rodents we show that: a lattice composed of linear filaments stabilizes the granule membrane after fusion with the plasma membrane; and a network of branched filaments linked to the membranes by Ezrin, a regulator of membrane tension, initiates and drives to completion the integration step. Our results highlight how the actin cytoskeleton tunes its structure to adapt to dynamic changes in the biophysical properties of membranes.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda