Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 36(7-8): 391-407, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35487686

RESUMEN

More than 27 yr ago, the vimentin knockout (Vim-/- ) mouse was reported to develop and reproduce without an obvious phenotype, implying that this major cytoskeletal protein was nonessential. Subsequently, comprehensive and careful analyses have revealed numerous phenotypes in Vim-/- mice and their organs, tissues, and cells, frequently reflecting altered responses in the recovery of tissues following various insults or injuries. These findings have been supported by cell-based experiments demonstrating that vimentin intermediate filaments (IFs) play a critical role in regulating cell mechanics and are required to coordinate mechanosensing, transduction, signaling pathways, motility, and inflammatory responses. This review highlights the essential functions of vimentin IFs revealed from studies of Vim-/- mice and cells derived from them.


Asunto(s)
Filamentos Intermedios , Vimentina/metabolismo , Animales , Fenómenos Fisiológicos Celulares , Filamentos Intermedios/genética , Filamentos Intermedios/metabolismo , Ratones , Vimentina/genética
2.
Cell ; 158(4): 822-832, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126787

RESUMEN

Molecular motors in cells typically produce highly directed motion; however, the aggregate, incoherent effect of all active processes also creates randomly fluctuating forces, which drive diffusive-like, nonthermal motion. Here, we introduce force-spectrum-microscopy (FSM) to directly quantify random forces within the cytoplasm of cells and thereby probe stochastic motor activity. This technique combines measurements of the random motion of probe particles with independent micromechanical measurements of the cytoplasm to quantify the spectrum of force fluctuations. Using FSM, we show that force fluctuations substantially enhance intracellular movement of small and large components. The fluctuations are three times larger in malignant cells than in their benign counterparts. We further demonstrate that vimentin acts globally to anchor organelles against randomly fluctuating forces in the cytoplasm, with no effect on their magnitude. Thus, FSM has broad applications for understanding the cytoplasm and its intracellular processes in relation to cell physiology in healthy and diseased states.


Asunto(s)
Citoplasma/química , Microscopía de Fuerza Atómica/métodos , Animales , Fenómenos Biomecánicos , Embrión de Mamíferos/citología , Fibroblastos/química , Ratones , Proteínas/química , Vimentina/química
3.
Proc Natl Acad Sci U S A ; 119(17): e2121816119, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35439057

RESUMEN

The ability of a cell to regulate its mechanical properties is central to its function. Emerging evidence suggests that interactions between the cell nucleus and cytoskeleton influence cell mechanics through poorly understood mechanisms. Here we conduct quantitative confocal imaging to show that the loss of A-type lamins tends to increase nuclear and cellular volume while the loss of B-type lamins behaves in the opposite manner. We use fluorescence recovery after photobleaching, atomic force microscopy, optical tweezer microrheology, and traction force microscopy to demonstrate that A-type lamins engage with both F-actin and vimentin intermediate filaments (VIFs) through the linker of nucleoskeleton and cytoskeleton (LINC) complexes to modulate cortical and cytoplasmic stiffness as well as cellular contractility in mouse embryonic fibroblasts (MEFs). In contrast, we show that B-type lamins predominantly interact with VIFs through LINC complexes to regulate cytoplasmic stiffness and contractility. We then propose a physical model mediated by the lamin­LINC complex that explains these distinct mechanical phenotypes (mechanophenotypes). To verify this model, we use dominant negative constructs and RNA interference to disrupt the LINC complexes that facilitate the interaction of the nucleus with the F-actin and VIF cytoskeletons and show that the loss of these elements results in mechanophenotypes like those observed in MEFs that lack A- or B-type lamin isoforms. Finally, we demonstrate that the loss of each lamin isoform softens the cell nucleus and enhances constricted cell migration but in turn increases migration-induced DNA damage. Together, our findings uncover distinctive roles for each of the four major lamin isoforms in maintaining nucleocytoskeletal interactions and cellular mechanics.


Asunto(s)
Fibroblastos , Lámina Nuclear , Animales , Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Ratones , Lámina Nuclear/metabolismo , Matriz Nuclear/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
4.
Nucleic Acids Res ; 50(20): e117, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36130229

RESUMEN

The chromatin associated with the nuclear lamina (NL) is referred to as lamina-associated domains (LADs). Here, we present an adaptation of the tyramide-signal amplification sequencing (TSA-seq) protocol, which we call chromatin pull down-based TSA-seq (cTSA-seq), that can be used to map chromatin regions at or near the NL from as little as 50 000 cells. The cTSA-seq mapped regions are composed of previously defined LADs and smaller chromatin regions that fall within the Hi-C defined B-compartment containing nuclear peripheral heterochromatin. We used cTSA-seq to map chromatin at or near the assembling NL in cultured cells progressing through early G1. cTSA-seq revealed that the distal ends of chromosomes are near or at the reassembling NL during early G1, a feature similar to those found in senescent cells. We expand the use of cTSA-seq to the mapping of chromatin at or near the NL from fixed-frozen mouse cerebellar tissue sections. This mapping reveals a general conservation of NL-associated chromatin and identifies global and local changes during cerebellar development. The cTSA-seq method reported here is useful for analyzing chromatin at or near the NL from small numbers of cells derived from both in vitro and in vivo sources.


Asunto(s)
Cromatina , Mapeo Cromosómico , Lámina Nuclear , Análisis de Secuencia de ADN , Animales , Ratones , Ciclo Celular , Cromatina/genética , Cromatina/metabolismo , Heterocromatina/metabolismo , Lámina Nuclear/metabolismo , Análisis de Secuencia de ADN/métodos , Mapeo Cromosómico/métodos
5.
Nature ; 543(7644): 261-264, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28241138

RESUMEN

The nuclear lamina is a fundamental constituent of metazoan nuclei. It is composed mainly of lamins, which are intermediate filament proteins that assemble into a filamentous meshwork, bridging the nuclear envelope and chromatin. Besides providing structural stability to the nucleus, the lamina is involved in many nuclear activities, including chromatin organization, transcription and replication. However, the structural organization of the nuclear lamina is poorly understood. Here we use cryo-electron tomography to obtain a detailed view of the organization of the lamin meshwork within the lamina. Data analysis of individual lamin filaments resolves a globular-decorated fibre appearance and shows that A- and B-type lamins assemble into tetrameric filaments of 3.5 nm thickness. Thus, lamins exhibit a structure that is remarkably different from the other canonical cytoskeletal elements. Our findings define the architecture of the nuclear lamin meshworks at molecular resolution, providing insights into their role in scaffolding the nuclear lamina.


Asunto(s)
Laminas/química , Laminas/ultraestructura , Lámina Nuclear/química , Lámina Nuclear/ultraestructura , Animales , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Cromatina/ultraestructura , Microscopía por Crioelectrón , Citoesqueleto/química , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Humanos , Proteínas de Filamentos Intermediarios/química , Proteínas de Filamentos Intermediarios/metabolismo , Proteínas de Filamentos Intermediarios/ultraestructura , Laminas/metabolismo , Ratones , Lámina Nuclear/metabolismo , Tomografía
6.
Proc Natl Acad Sci U S A ; 117(13): 7326-7337, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32170015

RESUMEN

Neutrophil extracellular traps (NETs) are web-like DNA structures decorated with histones and cytotoxic proteins that are released by activated neutrophils to trap and neutralize pathogens during the innate immune response, but also form in and exacerbate sterile inflammation. Peptidylarginine deiminase 4 (PAD4) citrullinates histones and is required for NET formation (NETosis) in mouse neutrophils. While the in vivo impact of NETs is accumulating, the cellular events driving NETosis and the role of PAD4 in these events are unclear. We performed high-resolution time-lapse microscopy of mouse and human neutrophils and differentiated HL-60 neutrophil-like cells (dHL-60) labeled with fluorescent markers of organelles and stimulated with bacterial toxins or Candida albicans to induce NETosis. Upon stimulation, cells exhibited rapid disassembly of the actin cytoskeleton, followed by shedding of plasma membrane microvesicles, disassembly and remodeling of the microtubule and vimentin cytoskeletons, ER vesiculation, chromatin decondensation and nuclear rounding, progressive plasma membrane and nuclear envelope (NE) permeabilization, nuclear lamin meshwork and then NE rupture to release DNA into the cytoplasm, and finally plasma membrane rupture and discharge of extracellular DNA. Inhibition of actin disassembly blocked NET release. Mouse and dHL-60 cells bearing genetic alteration of PAD4 showed that chromatin decondensation, lamin meshwork and NE rupture and extracellular DNA release required the enzymatic and nuclear localization activities of PAD4. Thus, NETosis proceeds by a stepwise sequence of cellular events culminating in the PAD4-mediated expulsion of DNA.


Asunto(s)
Trampas Extracelulares/inmunología , Neutrófilos/inmunología , Arginina Deiminasa Proteína-Tipo 4/inmunología , Animales , Cromatina/inmunología , Citoesqueleto/inmunología , ADN/inmunología , ADN/metabolismo , Trampas Extracelulares/metabolismo , Células HL-60 , Histonas/inmunología , Humanos , Inmunidad Innata , Inflamación/inmunología , Ratones , Microtúbulos/inmunología , Activación Neutrófila/inmunología , Neutrófilos/metabolismo , Membrana Nuclear/inmunología
7.
Bioessays ; 42(11): e2000078, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32893352

RESUMEN

Intermediate filaments (IFs) formed by vimentin are less understood than their cytoskeletal partners, microtubules and F-actin, but the unique physical properties of IFs, especially their resistance to large deformations, initially suggest a mechanical function. Indeed, vimentin IFs help regulate cell mechanics and contractility, and in crowded 3D environments they protect the nucleus during cell migration. Recently, a multitude of studies, often using genetic or proteomic screenings show that vimentin has many non-mechanical functions within and outside of cells. These include signaling roles in wound healing, lipogenesis, sterol processing, and various functions related to extracellular and cell surface vimentin. Extracellular vimentin is implicated in marking circulating tumor cells, promoting neural repair, and mediating the invasion of host cells by viruses, including SARS-CoV, or bacteria such as Listeria and Streptococcus. These findings underscore the fundamental role of vimentin in not only cell mechanics but also a range of physiological functions. Also see the video abstract here https://youtu.be/YPfoddqvz-g.


Asunto(s)
Filamentos Intermedios/fisiología , Mecanotransducción Celular/fisiología , Vimentina/fisiología , Animales , Fenómenos Fisiológicos Bacterianos , Interacciones Huésped-Patógeno/fisiología , Humanos , Filamentos Intermedios/química , Fenómenos Mecánicos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Vimentina/química , Internalización del Virus
8.
Proc Natl Acad Sci U S A ; 116(35): 17175-17180, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31409716

RESUMEN

In many developmental and pathological processes, including cellular migration during normal development and invasion in cancer metastasis, cells are required to withstand severe deformations. The structural integrity of eukaryotic cells under small deformations has been known to depend on the cytoskeleton including actin filaments (F-actin), microtubules (MT), and intermediate filaments (IFs). However, it remains unclear how cells resist severe deformations since both F-actin and microtubules yield or disassemble under moderate strains. Using vimentin containing IFs (VIFs) as a model for studying the large family of IF proteins, we demonstrate that they dominate cytoplasmic mechanics and maintain cell viability at large deformations. Our results show that cytoskeletal VIFs form a stretchable, hyperelastic network in living cells. This network works synergistically with other cytoplasmic components, substantially enhancing the strength, stretchability, resilience, and toughness of cells. Moreover, we find the hyperelastic VIF network, together with other quickly recoverable cytoskeletal components, forms a mechanically robust structure which can mechanically recover after damage.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Citoplasma/metabolismo , Filamentos Intermedios/metabolismo , Modelos Biológicos , Vimentina/metabolismo , Citoesqueleto de Actina/genética , Animales , Supervivencia Celular , Citoplasma/genética , Filamentos Intermedios/genética , Ratones , Ratones Noqueados , Vimentina/genética
9.
Bioinformatics ; 36(20): 5093-5103, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-32653917

RESUMEN

MOTIVATION: Microscopy images of cytoskeletal, nucleoskeletal and other structures contain complex junctions of overlapping filaments with arbitrary geometry. Yet, state-of-the-art algorithms generally perform single orientation analysis to segment these structures, resulting in gaps near junctions, or assume particular junction geometries to detect them. RESULTS: We developed a fully automated image analysis approach to address the challenge of determining the number of orientations and their values at each point in space to detect both lines and their junctions. Our approach does not assume any fixed number of orientations or any particular geometry in the case of multiple coincident orientations. It is based on analytically resolving coincident orientations revealed by steerable ridge filtering in an adaptive manner that balances orientation resolution and spatial localization. Combining this multiorientation resolution information with a generalization of the concept of non-maximum suppression allowed us to then identify the centers of lines and their junctions in an image. We validated our approach using a wide array of synthetic junctions and by comparison to manual segmentation. We also applied it to light microscopy images of cytoskeletal and nucleoskeletal networks. AVAILABILITY AND IMPLEMENTATION: https://github.com/mkitti/AdaptiveResolutionOrientationSpace. SUPPLEMENTARY INFORMATION: Supplementary information is available at Bioinformatics online.


Asunto(s)
Algoritmos , Procesamiento de Imagen Asistido por Computador , Microscopía
10.
Nature ; 527(7576): 105-9, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26524528

RESUMEN

Macroautophagy (hereafter referred to as autophagy) is a catabolic membrane trafficking process that degrades a variety of cellular constituents and is associated with human diseases. Although extensive studies have focused on autophagic turnover of cytoplasmic materials, little is known about the role of autophagy in degrading nuclear components. Here we report that the autophagy machinery mediates degradation of nuclear lamina components in mammals. The autophagy protein LC3/Atg8, which is involved in autophagy membrane trafficking and substrate delivery, is present in the nucleus and directly interacts with the nuclear lamina protein lamin B1, and binds to lamin-associated domains on chromatin. This LC3-lamin B1 interaction does not downregulate lamin B1 during starvation, but mediates its degradation upon oncogenic insults, such as by activated RAS. Lamin B1 degradation is achieved by nucleus-to-cytoplasm transport that delivers lamin B1 to the lysosome. Inhibiting autophagy or the LC3-lamin B1 interaction prevents activated RAS-induced lamin B1 loss and attenuates oncogene-induced senescence in primary human cells. Our study suggests that this new function of autophagy acts as a guarding mechanism protecting cells from tumorigenesis.


Asunto(s)
Autofagia , Lámina Nuclear/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Familia de las Proteínas 8 Relacionadas con la Autofagia , Transformación Celular Neoplásica , Células Cultivadas , Senescencia Celular , Cromatina/química , Cromatina/metabolismo , Citoplasma/metabolismo , Fibroblastos , Células HEK293 , Humanos , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Lisosomas/metabolismo , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Unión Proteica , Proteolisis
11.
Biophys J ; 119(1): 55-64, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32521238

RESUMEN

Divalent cations behave as effective cross-linkers of intermediate filaments (IFs) such as vimentin IF (VIF). These interactions have been mostly attributed to their multivalency. However, ion-protein interactions often depend on the ion species, and these effects have not been widely studied in IFs. Here, we investigate the effects of two biologically important divalent cations, Zn2+ and Ca2+, on VIF network structure and mechanics in vitro. We find that the network structure is unperturbed at micromolar Zn2+ concentrations, but strong bundle formation is observed at a concentration of 100 µM. Microrheological measurements show that network stiffness increases with cation concentration. However, bundling of filaments softens the network. This trend also holds for VIF networks formed in the presence of Ca2+, but remarkably, a concentration of Ca2+ that is two orders higher is needed to achieve the same effect as with Zn2+, which suggests the importance of salt-protein interactions as described by the Hofmeister effect. Furthermore, we find evidence of competitive binding between the two divalent ion species. Hence, specific interactions between VIFs and divalent cations are likely to be an important mechanism by which cells can control their cytoplasmic mechanics.


Asunto(s)
Citoesqueleto , Filamentos Intermedios , Cationes Bivalentes , Citoplasma , Vimentina
12.
FASEB J ; 33(1): 388-399, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29944446

RESUMEN

Keratin intermediate filaments (IFs) are the major cytoskeletal component in epithelial cells. The dynamics of keratin IFs have been described to depend mostly on the actin cytoskeleton, but the rapid transport of fully polymerized keratin filaments has not been reported. In this work, we used a combination of photoconversion experiments and clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats-associated protein 9 genome editing to study the role of microtubules and microtubule motors in keratin filament transport. We found that long keratin filaments, like other types of IFs, are transported along microtubules by kinesin-1. Our data revealed that keratin and vimentin are nonconventional kinesin-1 cargoes because their transport did not require kinesin light chains, which are a typical adapter for kinesin-dependent cargo transport. Furthermore, we found that the same domain of the kinesin heavy chain tail is involved in keratin and vimentin IF transport, strongly suggesting that multiple types of IFs move along microtubules using an identical mechanism.-Robert, A., Tian, P., Adam, S. A., Kittisopikul, M., Jaqaman, K., Goldman, R. D., Gelfand, V. I. Kinesin-dependent transport of keratin filaments: a unified mechanism for intermediate filament transport.


Asunto(s)
Filamentos Intermedios/metabolismo , Queratina-18/metabolismo , Queratina-8/metabolismo , Cinesinas/fisiología , Microtúbulos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Vimentina/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Sistemas CRISPR-Cas , Células Cultivadas , Cinesinas/antagonistas & inhibidores , Ratones , Ratones Noqueados , Microscopía Fluorescente
13.
Biophys J ; 116(3): 518-529, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30685055

RESUMEN

In development, wound healing, and pathology, cell biomechanical properties are increasingly recognized as being of central importance. To measure these properties, experimental probes of various types have been developed, but how each probe reflects the properties of heterogeneous cell regions has remained obscure. To better understand differences attributable to the probe technology, as well as to define the relative sensitivity of each probe to different cellular structures, here we took a comprehensive approach. We studied two cell types-Schlemm's canal endothelial cells and mouse embryonic fibroblasts (MEFs)-using four different probe technologies: 1) atomic force microscopy (AFM) with sharp tip, 2) AFM with round tip, 3) optical magnetic twisting cytometry (OMTC), and 4) traction microscopy (TM). Perturbation of Schlemm's canal cells with dexamethasone treatment, α-actinin overexpression, or RhoA overexpression caused increases in traction reported by TM and stiffness reported by sharp-tip AFM as compared to corresponding controls. By contrast, under these same experimental conditions, stiffness reported by round-tip AFM and by OMTC indicated little change. Knockout (KO) of vimentin in MEFs caused a diminution of traction reported by TM, as well as stiffness reported by sharp-tip and round-tip AFM. However, stiffness reported by OMTC in vimentin-KO MEFs was greater than in wild type. Finite-element analysis demonstrated that this paradoxical OMTC result in vimentin-KO MEFs could be attributed to reduced cell thickness. Our results also suggest that vimentin contributes not only to intracellular network stiffness but also cortex stiffness. Taken together, this evidence suggests that AFM sharp tip and TM emphasize properties of the actin-rich shell of the cell, whereas round-tip AFM and OMTC emphasize those of the noncortical intracellular network.


Asunto(s)
Citoesqueleto/metabolismo , Fenómenos Mecánicos , Animales , Fenómenos Biomecánicos , Células Endoteliales/citología , Fibroblastos/citología , Técnicas de Inactivación de Genes , Humanos , Ratones , Vimentina/deficiencia , Vimentina/genética
14.
Genes Dev ; 25(24): 2579-93, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22155925

RESUMEN

Nuclear lamin B1 (LB1) is a major structural component of the nucleus that appears to be involved in the regulation of many nuclear functions. The results of this study demonstrate that LB1 expression in WI-38 cells decreases during cellular senescence. Premature senescence induced by oncogenic Ras also decreases LB1 expression through a retinoblastoma protein (pRb)-dependent mechanism. Silencing the expression of LB1 slows cell proliferation and induces premature senescence in WI-38 cells. The effects of LB1 silencing on proliferation require the activation of p53, but not pRb. However, the induction of premature senescence requires both p53 and pRb. The proliferation defects induced by silencing LB1 are accompanied by a p53-dependent reduction in mitochondrial reactive oxygen species (ROS), which can be rescued by growth under hypoxic conditions. In contrast to the effects of LB1 silencing, overexpression of LB1 increases the proliferation rate and delays the onset of senescence of WI-38 cells. This overexpression eventually leads to cell cycle arrest at the G1/S boundary. These results demonstrate the importance of LB1 in regulating the proliferation and senescence of human diploid cells through a ROS signaling pathway.


Asunto(s)
Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Ciclo Celular/genética , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Senescencia Celular/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Especies Reactivas de Oxígeno/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Telómero/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/metabolismo
15.
Am J Physiol Renal Physiol ; 315(4): F769-F780, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29631355

RESUMEN

Most renal transplants ultimately fail secondary to chronic allograft nephropathy (CAN). Vimentin (vim) is a member of the intermediate filament family of proteins and has been shown to be important in the development of CAN. One of the pathways leading to chronic renal fibrosis after transplant is thought to be epithelial to mesenchymal transition (EMT). Even though vim expression is one of the main steps of EMT, it is unknown whether vim expression is required for EMT leading to renal fibrosis and allograft loss. To this end, the role of vim in renal fibrosis was determined via unilateral ureteral obstruction (UUO) in vim knockout mice (129 svs6 vim -/-). Following UUO, kidneys were recovered and analyzed via Western blotting, immunofluorescence, and transcriptomics. Cultured human proximal renal tubular (HK-2) cells were subjected to lentiviral-driven inhibition of vim expression and then treated with transforming growth factor (TGF)-ß to undergo EMT. Immunoblotting as well as wound healing assays were used to determine development of EMT. Western blotting analyses of mice undergoing UUO reveal increased levels of vim soon after UUO. As expected, interstitial collagen deposition increased in control mice following UUO but decreased in vim -/- kidneys. Immunofluorescence analyses also revealed altered localization of ß-catenin in vim -/- mice undergoing UUO without significant changes in mRNA levels. However, RNA sequencing revealed a decrease in ß-catenin-dependent genes in vim -/- kidneys. Finally, vim-silenced HK-2 cell lines undergoing EMT were shown to have decreased cellular migration during wound healing. We conclude that vim inhibition decreases fibrosis following UUO by possibly altering ß-catenin localization and downstream signaling.


Asunto(s)
Fibrosis/patología , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología , Vimentina/metabolismo , Animales , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/fisiología , Fibrosis/metabolismo , Filamentos Intermedios/metabolismo , Filamentos Intermedios/patología , Ratones Noqueados , Transducción de Señal/fisiología , Sistema Urinario/metabolismo , Sistema Urinario/patología
16.
Hum Mol Genet ; 25(11): 2143-2157, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27000625

RESUMEN

Intermediate filaments (IFs) are cytoskeletal polymers that extend from the nucleus to the cell membrane, giving cells their shape and form. Abnormal accumulation of IFs is involved in the pathogenesis of number neurodegenerative diseases, but none as clearly as giant axonal neuropathy (GAN), a ravaging disease caused by mutations in GAN, encoding gigaxonin. Patients display early and severe degeneration of the peripheral nervous system along with IF accumulation, but it has been difficult to link GAN mutations to any particular dysfunction, in part because GAN null mice have a very mild phenotype. We therefore established a robust dorsal root ganglion neuronal model that mirrors key cellular events underlying GAN. We demonstrate that gigaxonin is crucial for ubiquitin-proteasomal degradation of neuronal IF. Moreover, IF accumulation impairs mitochondrial motility and is associated with metabolic and oxidative stress. These results have implications for other neurological disorders whose pathology includes IF accumulation.


Asunto(s)
Proteínas del Citoesqueleto/genética , Neuropatía Axonal Gigante/genética , Proteínas de Filamentos Intermediarios/genética , Filamentos Intermedios/genética , Animales , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Neuropatía Axonal Gigante/patología , Humanos , Proteínas de Filamentos Intermediarios/biosíntesis , Filamentos Intermedios/patología , Ratones , Mitocondrias/genética , Mitocondrias/patología , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis
17.
J Cell Sci ; 129(14): 2732-43, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27235420

RESUMEN

Mutation of the LMNA gene, encoding nuclear lamin A and lamin C (hereafter lamin A/C), is a common cause of familial dilated cardiomyopathy (DCM). Among Finnish DCM patients, the founder mutation c.427T>C (p.S143P) is the most frequently reported genetic variant. Here, we show that p.S143P lamin A/C is more nucleoplasmic and soluble than wild-type lamin A/C and accumulates into large intranuclear aggregates in a fraction of cultured patient fibroblasts as well as in cells ectopically expressing either FLAG- or GFP-tagged p.S143P lamin A. In fluorescence loss in photobleaching (FLIP) experiments, non-aggregated EGFP-tagged p.S143P lamin A was significantly more dynamic. In in vitro association studies, p.S143P lamin A failed to form appropriate filament structures but instead assembled into disorganized aggregates similar to those observed in patient cell nuclei. A whole-genome expression analysis revealed an elevated unfolded protein response (UPR) in cells expressing p.S143P lamin A/C. Additional endoplasmic reticulum (ER) stress induced by tunicamycin reduced the viability of cells expressing mutant lamin further. In summary, p.S143P lamin A/C affects normal lamina structure and influences the cellular stress response, homeostasis and viability.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Estrés del Retículo Endoplásmico , Lamina Tipo A/metabolismo , Mutación/genética , Biomarcadores/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/ultraestructura , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Proteínas Mutantes/metabolismo , Agregado de Proteínas , Transfección , Regulación hacia Arriba
19.
J Biol Chem ; 290(28): 17145-53, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25957409

RESUMEN

Intermediate filaments (IFs) are composed of one or more members of a large family of cytoskeletal proteins, whose expression is cell- and tissue type-specific. Their importance in regulating the physiological properties of cells is becoming widely recognized in functions ranging from cell motility to signal transduction. IF proteins assemble into nanoscale biopolymers with unique strain-hardening properties that are related to their roles in regulating the mechanical integrity of cells. Furthermore, mutations in the genes encoding IF proteins cause a wide range of human diseases. Due to the number of different types of IF proteins, we have limited this short review to cover structure and function topics mainly related to the simpler homopolymeric IF networks composed of vimentin, and specifically for diseases, the related muscle-specific desmin IF networks.


Asunto(s)
Filamentos Intermedios/metabolismo , Animales , Fenómenos Biomecánicos , Movimiento Celular , Forma de la Célula , Desmina/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Filamentos Intermedios/química , Filamentos Intermedios/ultraestructura , Mutación , Orgánulos/metabolismo , Multimerización de Proteína , Vimentina/metabolismo
20.
J Cell Sci ; 127(Pt 12): 2683-96, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24741066

RESUMEN

Nuclear lamins form the major structural elements that comprise the nuclear lamina. Loss of nuclear structural integrity has been implicated as a key factor in the lamin A/C gene mutations that cause laminopathies, whereas the normal regulation of lamin A assembly and organization in interphase cells is still undefined. We assumed phosphorylation to be a major determinant, identifying 20 prime interphase phosphorylation sites, of which eight were high-turnover sites. We examined the roles of these latter sites by site-directed mutagenesis, followed by detailed microscopic analysis - including fluorescence recovery after photobleaching, fluorescence correlation spectroscopy and nuclear extraction techniques. The results reveal three phosphorylation regions, each with dominant sites, together controlling lamin A structure and dynamics. Interestingly, two of these interphase sites are hyper-phosphorylated in mitotic cells and one of these sites is within the sequence that is missing in progerin of the Hutchinson-Gilford progeria syndrome. We present a model where different phosphorylation combinations yield markedly different effects on the assembly, subunit turnover and the mobility of lamin A between, and within, the lamina, the nucleoplasm and the cytoplasm of interphase cells.


Asunto(s)
Interfase , Lamina Tipo A/metabolismo , Procesamiento Proteico-Postraduccional , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Células HeLa , Humanos , Datos de Secuencia Molecular , Lámina Nuclear/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas , Transporte de Proteínas , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA