Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 137(7): 1308-19, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19563761

RESUMEN

Post-Golgi vesicles target and deliver most biosynthetic cargoes to the cell surface. However, the molecules and mechanisms involved in fusion of these vesicles are not well understood. We have employed a system to simultaneously monitor release of luminal and membrane biosynthetic cargoes from individual post-Golgi vesicles. Exocytosis of these vesicles is not calcium triggered. Release of luminal cargo can be accompanied by complete, partial, or no release of membrane cargo. Partial and no release of membrane cargo of a fusing vesicle are fates associated with kiss-and-run exocytosis, and we find that these are the predominant mode of post-Golgi vesicle exocytosis. Partial cargo release by post-Golgi vesicles occurs because of premature closure of the fusion pore and is modulated by the activity of clathrin, actin, and dynamin. Our results demonstrate that these components of the endocytic machinery modulate the nature and extent of biosynthetic cargo delivery by post-Golgi vesicles at the cell membrane.


Asunto(s)
Exocitosis , Vesículas Transportadoras/metabolismo , Actinas/metabolismo , Membrana Celular/metabolismo , Clatrina/metabolismo , Dinaminas/metabolismo , Aparato de Golgi/metabolismo , Humanos , Microscopía Fluorescente , Mutación , Proteína 1A de Unión a Tacrolimus/genética
2.
Genome Res ; 30(6): 885-897, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32660935

RESUMEN

RNA-seq is widely used for studying gene expression, but commonly used sequencing platforms produce short reads that only span up to two exon junctions per read. This makes it difficult to accurately determine the composition and phasing of exons within transcripts. Although long-read sequencing improves this issue, it is not amenable to precise quantitation, which limits its utility for differential expression studies. We used long-read isoform sequencing combined with a novel analysis approach to compare alternative splicing of large, repetitive structural genes in muscles. Analysis of muscle structural genes that produce medium (Nrap: 5 kb), large (Neb: 22 kb), and very large (Ttn: 106 kb) transcripts in cardiac muscle, and fast and slow skeletal muscles identified unannotated exons for each of these ubiquitous muscle genes. This also identified differential exon usage and phasing for these genes between the different muscle types. By mapping the in-phase transcript structures to known annotations, we also identified and quantified previously unannotated transcripts. Results were confirmed by endpoint PCR and Sanger sequencing, which revealed muscle-type-specific differential expression of these novel transcripts. The improved transcript identification and quantification shown by our approach removes previous impediments to studies aimed at quantitative differential expression of ultralong transcripts.


Asunto(s)
Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , ARN Mensajero , Análisis de Secuencia de ARN , Transcriptoma , Empalme Alternativo , Biología Computacional/métodos , Exones , Perfilación de la Expresión Génica/métodos , Humanos , Anotación de Secuencia Molecular , Especificidad de Órganos/genética , Secuencias Repetitivas de Ácidos Nucleicos
3.
FASEB J ; 35(9): e21819, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34405910

RESUMEN

Skeletal muscle contains multiple cell types that work together to maintain tissue homeostasis. Among these, satellite cells (SC) and fibroadipogenic progenitors cells (FAPs) are the two main stem cell pools. Studies of these cells using animal models have shown the importance of interactions between these cells in repair of healthy muscle, and degeneration of dystrophic muscle. Due to the unavailability of fresh patient muscle biopsies, similar analysis of interactions between human FAPs and SCs is limited especially among the muscular dystrophy patients. To address this issue here we describe a method that allows the use of frozen human skeletal muscle biopsies to simultaneously isolate and grow SCs and FAPs from healthy or dystrophic patients. We show that while the purified SCs differentiate into mature myotubes, purified FAPs can differentiate into adipocytes or fibroblasts demonstrating their multipotency. We find that these FAPs can be immortalized and the immortalized FAPs (iFAPs) retain their multipotency. These approaches open the door for carrying out personalized analysis of patient FAPs and interactions with the SCs that lead to muscle loss.


Asunto(s)
Biopsia , Separación Celular , Criopreservación , Músculo Esquelético/citología , Músculo Esquelético/patología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/patología , Adolescente , Adulto , Anciano , Diferenciación Celular , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Células Madre Multipotentes/citología , Células Madre Multipotentes/patología , Distrofia Muscular de Duchenne/patología , Adulto Joven
4.
Ann Neurol ; 88(2): 332-347, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32403198

RESUMEN

OBJECTIVE: A hitherto undescribed phenotype of early onset muscular dystrophy associated with sensorineural hearing loss and primary ovarian insufficiency was initially identified in 2 siblings and in subsequent patients with a similar constellation of findings. The goal of this study was to understand the genetic and molecular etiology of this condition. METHODS: We applied whole exome sequencing (WES) superimposed on shared haplotype regions to identify the initial biallelic variants in GGPS1 followed by GGPS1 Sanger sequencing or WES in 5 additional families with the same phenotype. Molecular modeling, biochemical analysis, laser membrane injury assay, and the generation of a Y259C knock-in mouse were done. RESULTS: A total of 11 patients in 6 families carrying 5 different biallelic pathogenic variants in specific domains of GGPS1 were identified. GGPS1 encodes geranylgeranyl diphosphate synthase in the mevalonate/isoprenoid pathway, which catalyzes the synthesis of geranylgeranyl pyrophosphate, the lipid precursor of geranylgeranylated proteins including small guanosine triphosphatases. In addition to proximal weakness, all but one patient presented with congenital sensorineural hearing loss, and all postpubertal females had primary ovarian insufficiency. Muscle histology was dystrophic, with ultrastructural evidence of autophagic material and large mitochondria in the most severe cases. There was delayed membrane healing after laser injury in patient-derived myogenic cells, and a knock-in mouse of one of the mutations (Y259C) resulted in prenatal lethality. INTERPRETATION: The identification of specific GGPS1 mutations defines the cause of a unique form of muscular dystrophy with hearing loss and ovarian insufficiency and points to a novel pathway for this clinical constellation. ANN NEUROL 2020;88:332-347.


Asunto(s)
Dimetilaliltranstransferasa/genética , Farnesiltransferasa/genética , Geraniltranstransferasa/genética , Pérdida Auditiva/genética , Distrofias Musculares/genética , Mutación/genética , Insuficiencia Ovárica Primaria/genética , Adolescente , Adulto , Animales , Femenino , Técnicas de Sustitución del Gen/métodos , Pérdida Auditiva/diagnóstico por imagen , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Distrofias Musculares/diagnóstico por imagen , Linaje , Insuficiencia Ovárica Primaria/diagnóstico por imagen , Estructura Secundaria de Proteína , Análisis de Secuencia de ADN/métodos , Secuenciación del Exoma/métodos , Adulto Joven
5.
Biochem Soc Trans ; 48(5): 1995-2002, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-32985660

RESUMEN

Mitochondria are central to the health of eukaryotic cells. While commonly known for their bioenergetic role, mitochondria also function as signaling organelles that regulate cell stress responses capable of restoring homeostasis or leading the stressed cell to eventual death. Damage to the plasma membrane is a potentially fatal stressor incurred by all cells. Repairing plasma membrane damage requires cells to mount a rapid and localized response to injury. Accumulating evidence has identified a role for mitochondria as an important facilitator of this acute and localized repair response. However, as mitochondria are organized in a cell-wide, interconnected network, it is unclear how they collectively sense and respond to a focal injury. Here we will discuss how mitochondrial shape change is an integral part of this localized repair response. Mitochondrial fragmentation spatially restricts beneficial repair signaling, enabling a localized response to focal injury. Conservation of mitochondrial fragmentation in response to cell and tissue damage across species demonstrates that this is a universal pro-survival adaptation to injury and suggests that mitochondrial fragmentation may provide cells a mechanism to facilitate localized signaling in contexts beyond repairing plasma membrane injury.


Asunto(s)
Membrana Celular/metabolismo , Supervivencia Celular , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno , Transducción de Señal , Animales , Calcio/metabolismo , Muerte Celular , Citoesqueleto/metabolismo , Daño del ADN , Metabolismo Energético , Ambiente , Homeostasis , Humanos , Oxidación-Reducción , Cicatrización de Heridas
6.
J Pathol ; 249(2): 215-226, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31135059

RESUMEN

The etiology of myositis is unknown. Although attempts to identify viruses in myositis skeletal muscle have failed, several studies have identified the presence of a viral signature in myositis patients. Here we postulate that in individuals with susceptible genetic backgrounds, viral infection alters the epigenome to activate the pathological pathways leading to disease onset. To identify epigenetic changes, methylation profiling of Coxsackie B infected human myotubes and muscle biopsies from polymyositis (PM) and dermatomyositis (DM) patients were compared to changes in global transcript expression induced by in vitro Coxsackie B infection. Gene and protein expression analysis and live cell imaging were performed to examine the mechanisms. Analysis of methylation and gene expression changes identified that a mitochondria-localized activator of apoptosis - harakiri (HRK) - is upregulated in myositis skeletal muscle cells. Muscle cells with higher HRK expression have reduced mitochondrial potential and poor ability to repair from injury as compared to controls. In cells from myositis patient toll-like receptor 7 (TLR7) activates and sustains high HRK expression. Forced over expression of HRK in healthy muscle cells is sufficient to compromise their membrane repair ability. Endurance exercise that is associated with improved muscle and mitochondrial function in PM and DM patients decreased TLR7 and HRK expression identifying these as therapeutic targets. Increased HRK and TLR7 expression causes mitochondrial damage leading to poor myofiber repair, myofiber death and muscle weakness in myositis patients and exercise induced reduction of HRK and TLR7 expression in patients is associated with disease amelioration. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Dermatomiositis/metabolismo , Enterovirus Humano B/patogenicidad , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Polimiositis/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Estudios de Casos y Controles , Células Cultivadas , Metilación de ADN , Dermatomiositis/patología , Dermatomiositis/fisiopatología , Dermatomiositis/virología , Epigénesis Genética , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Mitocondrias Musculares/genética , Mitocondrias Musculares/patología , Mitocondrias Musculares/virología , Fuerza Muscular , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Músculo Esquelético/virología , Mioblastos Esqueléticos/patología , Mioblastos Esqueléticos/virología , Resistencia Física , Polimiositis/patología , Polimiositis/fisiopatología , Polimiositis/virología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Regulación hacia Arriba
7.
Mol Cell ; 47(5): 707-21, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22857951

RESUMEN

Doublecortin (Dcx) defines a growing family of microtubule (MT)-associated proteins (MAPs) involved in neuronal migration and process outgrowth. We show that Dcx is essential for the function of Kif1a, a kinesin-3 motor protein that traffics synaptic vesicles. Neurons lacking Dcx and/or its structurally conserved paralogue, doublecortin-like kinase 1 (Dclk1), show impaired Kif1a-mediated transport of Vamp2, a cargo of Kif1a, with decreased run length. Human disease-associated mutations in Dcx's linker sequence (e.g., W146C, K174E) alter Kif1a/Vamp2 transport by disrupting Dcx/Kif1a interactions without affecting Dcx MT binding. Dcx specifically enhances binding of the ADP-bound Kif1a motor domain to MTs. Cryo-electron microscopy and subnanometer-resolution image reconstruction reveal the kinesin-dependent conformational variability of MT-bound Dcx and suggest a model for MAP-motor crosstalk on MTs. Alteration of kinesin run length by MAPs represents a previously undiscovered mode of control of kinesin transport and provides a mechanism for regulation of MT-based transport by local signals.


Asunto(s)
Cinesinas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Quinasas Similares a Doblecortina , Femenino , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Microtúbulos/metabolismo , Neuronas/citología , Neuropéptidos/deficiencia , Proteínas Serina-Treonina Quinasas/deficiencia
8.
Int J Mol Sci ; 21(15)2020 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-32759720

RESUMEN

Deficits in plasma membrane repair have been identified in dysferlinopathy and Duchenne Muscular Dystrophy, and contribute to progressive myopathy. Although Facioscapulohumeral Muscular Dystrophy (FSHD) shares clinicopathological features with these muscular dystrophies, it is unknown if FSHD is characterized by plasma membrane repair deficits. Therefore, we exposed immortalized human FSHD myoblasts, immortalized myoblasts from unaffected siblings, and myofibers from a murine model of FSHD (FLExDUX4) to focal, pulsed laser ablation of the sarcolemma. Repair kinetics and success were determined from the accumulation of intracellular FM1-43 dye post-injury. We subsequently treated FSHD myoblasts with a DUX4-targeting antisense oligonucleotide (AON) to reduce DUX4 expression, and with the antioxidant Trolox to determine the role of DUX4 expression and oxidative stress in membrane repair. Compared to unaffected myoblasts, FSHD myoblasts demonstrate poor repair and a greater percentage of cells that failed to repair, which was mitigated by AON and Trolox treatments. Similar repair deficits were identified in FLExDUX4 myofibers. This is the first study to identify plasma membrane repair deficits in myoblasts from individuals with FSHD, and in myofibers from a murine model of FSHD. Our results suggest that DUX4 expression and oxidative stress may be important targets for future membrane-repair therapies.


Asunto(s)
Proteínas de Homeodominio/genética , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapulohumeral/genética , Estrés Oxidativo/genética , Adulto , Anciano , Animales , Antioxidantes/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/antagonistas & inhibidores , Humanos , Masculino , Ratones , Persona de Mediana Edad , Fibras Musculares Esqueléticas/patología , Distrofia Muscular Facioescapulohumeral/metabolismo , Distrofia Muscular Facioescapulohumeral/patología , Distrofia Muscular Facioescapulohumeral/terapia , Mioblastos/metabolismo , Miofibrillas/genética , Miofibrillas/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacología , Estrés Oxidativo/efectos de los fármacos
9.
Hum Mol Genet ; 26(11): 1979-1991, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334824

RESUMEN

Repair of skeletal muscle after sarcolemmal damage involves dysferlin and dysferlin-interacting proteins such as annexins. Mice and patient lacking dysferlin exhibit chronic muscle inflammation and adipogenic replacement of the myofibers. Here, we show that similar to dysferlin, lack of annexin A2 (AnxA2) also results in poor myofiber repair and progressive muscle weakening with age. By longitudinal analysis of AnxA2-deficient muscle we find that poor myofiber repair due to the lack of AnxA2 does not result in chronic inflammation or adipogenic replacement of the myofibers. Further, deletion of AnxA2 in dysferlin deficient mice reduced muscle inflammation, adipogenic replacement of myofibers, and improved muscle function. These results identify multiple roles of AnxA2 in muscle repair, which includes facilitating myofiber repair, chronic muscle inflammation and adipogenic replacement of dysferlinopathic muscle. It also identifies inhibition of AnxA2-mediated inflammation as a novel therapeutic avenue for treating muscle loss in dysferlinopathy.


Asunto(s)
Anexina A2/metabolismo , Anexina A2/fisiología , Adipogénesis , Animales , Anexina A2/genética , Disferlina , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/terapia , Miofibrillas/fisiología , Sarcolema/metabolismo
10.
Mol Ther ; 26(9): 2231-2242, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30166241

RESUMEN

Mutations of the DYSF gene leading to reduced dysferlin protein level causes limb girdle muscular dystrophy type 2B (LGMD2B). Dysferlin facilitates sarcolemmal membrane repair in healthy myofibers, thus its deficit compromises myofiber repair and leads to chronic muscle inflammation. An experimental therapeutic approach for LGMD2B is to protect damage or improve repair of myofiber sarcolemma. Here, we compared the effects of prednisolone and vamorolone (a dissociative steroid; VBP15) on dysferlin-deficient myofiber repair. Vamorolone, but not prednisolone, stabilized dysferlin-deficient muscle cell membrane and improved repair of dysferlin-deficient mouse (B6A/J) myofibers injured by focal sarcolemmal damage, eccentric contraction-induced injury or injury due to spontaneous in vivo activity. Vamorolone decreased sarcolemmal lipid mobility, increased muscle strength, and decreased late-stage myofiber loss due to adipogenic infiltration. In contrast, the conventional glucocorticoid prednisolone failed to stabilize dysferlin deficient muscle cell membrane or improve repair of dysferlinopathic patient myoblasts and mouse myofibers. Instead, prednisolone treatment increased muscle weakness and myofiber atrophy in B6A/J mice-findings that correlate with reports of prednisolone worsening symptoms of LGMD2B patients. Our findings showing improved cellular and pre-clinical efficacy of vamorolone compared to prednisolone and better safety profile of vamorolone indicates the suitability of vamorolone for clinical trials in LGMD2B.


Asunto(s)
Disferlina/deficiencia , Distrofias Musculares/tratamiento farmacológico , Esteroides/uso terapéutico , Adolescente , Animales , Células Cultivadas , Disferlina/metabolismo , Humanos , Masculino , Ratones , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Prednisolona/uso terapéutico , Pregnadienodioles/uso terapéutico
11.
Cell Mol Life Sci ; 75(20): 3751-3770, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30051163

RESUMEN

Plasma membrane forms the barrier between the cytoplasm and the environment. Cells constantly and selectively transport molecules across their plasma membrane without disrupting it. Any disruption in the plasma membrane compromises its selective permeability and is lethal, if not rapidly repaired. There is a growing understanding of the organelles, proteins, lipids, and small molecules that help cells signal and efficiently coordinate plasma membrane repair. This review aims to summarize how these subcellular responses are coordinated and how cellular signals generated due to plasma membrane injury interact with each other to spatially and temporally coordinate repair. With the involvement of calcium and redox signaling in single cell and tissue repair, we will discuss how these and other related signals extend from single cell repair to tissue level repair. These signals link repair processes that are activated immediately after plasma membrane injury with longer term processes regulating repair and regeneration of the damaged tissue. We propose that investigating cell and tissue repair as part of a continuum of wound repair mechanisms would be of value in treating degenerative diseases.


Asunto(s)
Membrana Celular/metabolismo , Transducción de Señal/fisiología , Animales , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Humanos , Fosfolipasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Proteínas de Unión al GTP rho/metabolismo
12.
Curr Top Membr ; 84: 67-98, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31610866

RESUMEN

The plasma membrane forms the physical barrier between the cytoplasm and extracellular space, allowing for biochemical reactions necessary for life to occur. Plasma membrane damage needs to be rapidly repaired to avoid cell death. This relies upon the coordinated action of the machinery that polarizes the repair response to the site of injury, resulting in resealing of the damaged membrane and subsequent remodeling to return the injured plasma membrane to its pre-injury state. As lipids comprise the bulk of the plasma membrane, the acts of injury, resealing, and remodeling all directly impinge upon the plasma membrane lipids. In addition to their structural role in shaping the physical properties of the plasma membrane, lipids also play an important signaling role in maintaining plasma membrane integrity. While much attention has been paid to the involvement of proteins in the membrane repair pathway, the role of lipids in facilitating plasma membrane repair remains poorly studied. Here we will discuss the current knowledge of how lipids facilitate plasma membrane repair by regulating membrane structure and signaling to coordinate the repair response, and will briefly note how lipid involvement extends beyond plasma membrane repair to the tissue repair response.


Asunto(s)
Membrana Celular/química , Membrana Celular/metabolismo , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Transducción de Señal , Animales , Humanos , Estructura Molecular
13.
J Am Soc Nephrol ; 28(3): 862-875, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27811066

RESUMEN

Studies have shown that podocytes and renal tubular epithelial cells from patients with HIV-associated nephropathy (HIVAN) express HIV-1 transcripts, suggesting that productive infection of renal epithelial cells precipitates development of HIVAN. However, podocytes and renal tubular epithelial cells do not express CD4 receptors, and it is unclear how these cells become productively infected in vivo We investigated the mechanisms underlying the infection by HIV-1 of podocytes cultured from the urine of children with HIVAN. We observed low-level productive infection on exposure of these cells to primary cell-free HIV-1 supernatants. However, envelope-defective recombinant HIV-1 did not infect the renal epithelial cell lines. Moreover, treatment of podocytes to inhibit endocytic transport or dynamin activity or remove cell surface heparan sulfate proteoglycans reduced infection efficiency. Transfection of CD4- 293T cells with a cDNA expression library developed from a podocyte cell line derived from a child with HIVAN led to the identification of TNF-α as a possible mediator of HIV-1 infection. Overexpression of transmembrane TNF-α in cultured CD4- renal tubular epithelial cells, 293T cells, and HeLa cells enabled the infection of these cells; exposure to soluble TNF-α did not. Immunohistochemistry showed TNF-α expression in podocytes of renal sections from children with HIVAN. Furthermore, we found that TNF-α enhanced NF-κB activation and integration of HIV-1 into the podocyte DNA. Finally, inhibition of dynamin activity blocked TNF-α-mediated infection. These data establish a role for transmembrane TNF-α in facilitating the viral entry and integration of HIV-1 into the DNA of renal epithelial cells.


Asunto(s)
Nefropatía Asociada a SIDA/virología , VIH-1/fisiología , Podocitos/virología , Factor de Necrosis Tumoral alfa/fisiología , Células Cultivadas , Niño , Humanos , Proteínas de la Membrana
15.
Med Microbiol Immunol ; 204(3): 449-60, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25724304

RESUMEN

The endoplasmic reticulum (ER) membrane is closely apposed to the outer mitochondrial membrane (OMM), which facilitates communication between these organelles. These contacts, known as mitochondria-associated membranes (MAM), facilitate calcium signaling, lipid transfer, as well as antiviral and stress responses. How cellular proteins traffic to the MAM, are distributed therein, and interact with ER and mitochondrial proteins are subject of great interest. The human cytomegalovirus UL37 exon 1 protein or viral mitochondria-localized inhibitor of apoptosis (vMIA) is crucial for viral growth. Upon synthesis at the ER, vMIA traffics to the MAM and OMM, where it reprograms the organization and function of these compartments. vMIA significantly changes the abundance of cellular proteins at the MAM and OMM, including proteins that regulate calcium homeostasis and cell death. Through the use of superresolution imaging, we have shown that vMIA is distributed at the OMM in nanometer scale clusters. This is similar to the clusters reported for the mitochondrial calcium channel, VDAC, as well as electron transport chain, translocase of the OMM complex, and mitochondrial inner membrane organizing system components. Thus, aside from addressing how vMIA targets the MAM and regulates survival of infected cells, biochemical studies and superresolution imaging of vMIA offer insights into the formation, organization, and functioning of MAM. Here, we discuss these insights into trafficking, function, and organization of vMIA at the MAM and OMM and discuss how the use of superresolution imaging is contributing to the study of the formation and trafficking of viruses.


Asunto(s)
Imagen Molecular , Proteínas Virales/metabolismo , Animales , Apoptosis , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Retículo Endoplásmico/metabolismo , Humanos , Espacio Intracelular/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Imagen Molecular/métodos , Imagen Óptica/métodos , Transporte de Proteínas , Replicación Viral
16.
Mol Cell Proteomics ; 12(5): 1061-73, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23297347

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular disorder caused by a mutation in the dystrophin gene. DMD is characterized by progressive weakness of skeletal, cardiac, and respiratory muscles. The molecular mechanisms underlying dystrophy-associated muscle weakness and damage are not well understood. Quantitative proteomics techniques could help to identify disease-specific pathways. Recent advances in the in vivo labeling strategies such as stable isotope labeling in mouse (SILAC mouse) with (13)C6-lysine or stable isotope labeling in mammals (SILAM) with (15)N have enabled accurate quantitative analysis of the proteomes of whole organs and tissues as a function of disease. Here we describe the use of the SILAC mouse strategy to define the underlying pathological mechanisms in dystrophin-deficient skeletal muscle. Differential SILAC proteome profiling was performed on the gastrocnemius muscles of 3-week-old (early stage) dystrophin-deficient mdx mice and wild-type (normal) mice. The generated data were further confirmed in an independent set of mdx and normal mice using a SILAC spike-in strategy. A total of 789 proteins were quantified; of these, 73 were found to be significantly altered between mdx and normal mice (p < 0.05). Bioinformatics analyses using Ingenuity Pathway software established that the integrin-linked kinase pathway, actin cytoskeleton signaling, mitochondrial energy metabolism, and calcium homeostasis are the pathways initially affected in dystrophin-deficient muscle at early stages of pathogenesis. The key proteins involved in these pathways were validated by means of immunoblotting and immunohistochemistry in independent sets of mdx mice and in human DMD muscle biopsies. The specific involvement of these molecular networks early in dystrophic pathology makes them potential therapeutic targets. In sum, our findings indicate that SILAC mouse strategy has uncovered previously unidentified pathological pathways in mouse models of human skeletal muscle disease.


Asunto(s)
Redes y Vías Metabólicas , Distrofia Muscular de Duchenne/metabolismo , Proteoma/metabolismo , Animales , Encéfalo/metabolismo , Distrofina/deficiencia , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/metabolismo , Especificidad de Órganos , Proteómica
17.
J Neurosci ; 33(2): 709-21, 2013 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23303949

RESUMEN

Doublecortin (Dcx) is the causative gene for X-linked lissencephaly, which encodes a microtubule-binding protein. Axon tracts are abnormal in both affected individuals and in animal models. To determine the reason for the axon tract defect, we performed a semiquantitative proteomic analysis of the corpus callosum in mice mutant for Dcx. In axons from mice mutant for Dcx, widespread differences are found in actin-associated proteins as compared with wild-type axons. Decreases in actin-binding proteins α-actinin-1 and α-actinin-4 and actin-related protein 2/3 complex subunit 3 (Arp3), are correlated with dysregulation in the distribution of filamentous actin (F-actin) in the mutant neurons with increased F-actin around the cell body and decreased F-actin in the neurites and growth cones. The actin distribution defect can be rescued by full-length Dcx and further enhanced by Dcx S297A, the unphosphorylatable mutant, but not with the truncation mutant of Dcx missing the C-terminal S/P-rich domain. Thus, the C-terminal region of Dcx dynamically regulates formation of F-actin features in developing neurons, likely through interaction with spinophilin, but not through α-actinin-4 or Arp3. We show with that the phenotype of Dcx/Doublecortin-like kinase 1 deficiency is consistent with actin defect, as these axons are selectively deficient in axon guidance, but not elongation.


Asunto(s)
Actinas/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas de Neurofilamentos/fisiología , Neuronas/fisiología , Neuropéptidos/fisiología , Proteína 3 Relacionada con la Actina/metabolismo , Actinina/metabolismo , Actinas/metabolismo , Animales , Axones/fisiología , Western Blotting , Células Cultivadas , Cuerpo Calloso/citología , Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Calloso/fisiología , Bases de Datos Factuales , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Electroforesis en Gel de Poliacrilamida , Femenino , Inmunohistoquímica , Masculino , Espectrometría de Masas , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuropéptidos/genética , Proteómica
18.
J Cell Sci ; 125(Pt 23): 5721-32, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22992464

RESUMEN

The protein clathrin mediates one of the major pathways of endocytosis from the extracellular milieu and plasma membrane. In single-cell eukaryotes, such as Saccharomyces cerevisiae, the gene encoding clathrin is not an essential gene, raising the question of whether clathrin conveys specific advantages for multicellularity. Furthermore, in contrast to mammalian cells, endocytosis in S. cerevisiae is not dependent on either clathrin or adaptor protein 2 (AP2), an endocytic adaptor molecule. In this study, we investigated the requirement for components of clathrin-mediated endocytosis (CME) in another unicellular organism, the amoeba Dictyostelium. We identified a heterotetrameric AP2 complex in Dictyostelium that is similar to that which is found in higher eukaryotes. By simultaneously imaging fluorescently tagged clathrin and AP2, we found that, similar to higher eukaryotes, these proteins colocalized to membrane puncta that move into the cell together. In addition, the contractile vacuole marker protein, dajumin-green fluorescent protein (GFP), is trafficked via the cell membrane and internalized by CME in a clathrin-dependent, AP2-independent mechanism. This pathway is distinct from other endocytic mechanisms in Dictyostelium. Our finding that CME is required for the internalization of contractile vacuole proteins from the cell membrane explains the contractile vacuole biogenesis defect in Dictyostelium cells lacking clathrin. Our results also suggest that the machinery for CME and its role in organelle maintenance appeared early during eukaryotic evolution. We hypothesize that dependence of endocytosis on specific components of the CME pathway evolved later, as demonstrated by internalization independent of AP2 function.


Asunto(s)
Clatrina/metabolismo , Dictyostelium/citología , Dictyostelium/metabolismo , Endocitosis/fisiología , Complejo 2 de Proteína Adaptadora/metabolismo , Biogénesis de Organelos , Saccharomyces cerevisiae
19.
Nat Commun ; 15(1): 1574, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383560

RESUMEN

Annexins are cytosolic proteins with conserved three-dimensional structures that bind acidic phospholipids in cellular membranes at elevated Ca2+ levels. Through this they act as Ca2+-regulated membrane binding modules that organize membrane lipids, facilitating cellular membrane transport but also displaying extracellular activities. Recent discoveries highlight annexins as sensors and regulators of cellular and organismal stress, controlling inflammatory reactions in mammals, environmental stress in plants, and cellular responses to plasma membrane rupture. Here, we describe the role of annexins as Ca2+-regulated membrane binding modules that sense and respond to cellular stress and share our view on future research directions in the field.


Asunto(s)
Anexinas , Gusto , Animales , Anexinas/química , Membrana Celular/metabolismo , Transducción de Señal , Transporte Biológico , Calcio/metabolismo , Mamíferos/metabolismo
20.
J Biol Chem ; 287(36): 30455-67, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22778268

RESUMEN

Skeletal muscles are proficient at healing from a variety of injuries. Healing occurs in two phases, early and late phase. Early phase involves healing the injured sarcolemma and restricting the spread of damage to the injured myofiber. Late phase of healing occurs a few days postinjury and involves interaction of injured myofibers with regenerative and inflammatory cells. Of the two phases, cellular and molecular processes involved in the early phase of healing are poorly understood. We have implemented an improved sarcolemmal proteomics approach together with in vivo labeling of proteins with modified amino acids in mice to study acute changes in the sarcolemmal proteome in early phase of myofiber injury. We find that a notable early phase response to muscle injury is an increased association of mitochondria with the injured sarcolemma. Real-time imaging of live myofibers during injury demonstrated that the increased association of mitochondria with the injured sarcolemma involves translocation of mitochondria to the site of injury, a response that is lacking in cultured myoblasts. Inhibiting mitochondrial function at the time of injury inhibited healing of the injured myofibers. This identifies a novel role of mitochondria in the early phase of healing injured myofibers.


Asunto(s)
Mitocondrias Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Proteómica , Sarcolema/metabolismo , Cicatrización de Heridas/fisiología , Animales , Línea Celular Transformada , Femenino , Masculino , Ratones , Fibras Musculares Esqueléticas/patología , Sarcolema/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA