Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Intervalo de año de publicación
1.
Matrix Biol ; 123: 1-16, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37660739

RESUMEN

Fibrosis is associated with dramatic changes in extracellular matrix (ECM) architecture of unknown etiology. Here we exploit keloid scars as a paradigm to understand fibrotic ECM organization. We reveal that keloid patient fibroblasts uniquely produce a globally aligned ECM network in 2-D culture as observed in scar tissue. ECM anisotropy develops after rapid initiation of a fibroblast supracellular actin network, suggesting that cell alignment initiates ECM patterning. Keloid fibroblasts produce elevated levels of IL-6, and autocrine IL-6 production is both necessary and sufficient to induce cell and ECM alignment, as evidenced by ligand stimulation of normal dermal fibroblasts and treatment of keloid fibroblasts with the function blocking IL-6 receptor monoclonal antibody, tocilizumab. Downstream of IL-6, supracellular organization of keloid fibroblasts is controlled by activation of cell-cell adhesion. Adhesion formation inhibits contact-induced cellular overlap leading to nematic organization of cells and an alignment of focal adhesions. Keloid fibroblasts placed on isotropic ECM align the pre-existing matrix, suggesting that focal adhesion alignment leads to active anisotropic remodeling. These results show that IL-6-induced fibroblast cooperativity can control the development of a nematic ECM, highlighting both IL-6 signaling and cell-cell adhesions as potential therapeutic targets to inhibit this common feature of fibrosis.


Asunto(s)
Queloide , Humanos , Queloide/tratamiento farmacológico , Interleucina-6/genética , Interleucina-6/metabolismo , Anisotropía , Células Cultivadas , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo
2.
Transl Lung Cancer Res ; 12(7): 1384-1390, 2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37577311

RESUMEN

Background: Malignant pleural mesothelioma (MPM) is an incurable, late presenting primary cancer, conferring a survival of 8-14 months. Different intrapleural treatments have been tested as part of a multimodality approach to treat a select group of patients with limited disease, increasing survival. Recently, povidone-iodine has been shown to induce apoptosis in microscopic tumour cells in vitro, with no reported complications. This is the first in vivo study assessing the apoptotic rate caused by intraoperative hyperthermic betadine lavage using routine immunohistochemistry combined with transmission electron microscopy (TEM). Methods: We included surgically fit patients aged >18, undergoing minimally invasive video-assisted thoracoscopic surgery (VATS) pleural biopsy between December 2016 and February 2018, for confirmed or presumed pleural malignancy. Parietal pleural biopsies were obtained at 7.5, 15 and 30 minutes after hyperthermic betadine lavage, and compared to pre-lavage biopsy samples, for apoptotic changes. Viable tumour samples underwent histological, immunohistochemical and ultrastructural analysis as well as TEM for features of apoptosis. Results: N=6. Median age was 76 years. Median overall survival was 26.7 months. There was no statistical impact on survival of side of disease (left vs. right). There was no significant difference in expressions of markers of apoptotic index pre and post betadine treatment upon immunohistochemical analysis. There was no discernible effect on morphological features of apoptosis seen with betadine treatment, on TEM analysis. No side effects were identified post betadine lavage. Conclusions: Although hyperthermic betadine lavage is a safe antiseptic solution with no toxicity when performed intraoperatively, it confers no effect on apoptotic rate or necrosis. It is therefore unlikely that hyperthermic betadine lavage will have an impact on reducing the microscopic residual disease after pleurectomy decortication and enhancing survival.

3.
Ultrason Sonochem ; 97: 106445, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37257208

RESUMEN

Phase-change nanodroplets have attracted increasing interest in recent years as ultrasound theranostic nanoparticles. They are smaller compared to microbubbles and they may distribute better in tissues (e.g. in tumours). They are composed of a stabilising shell and a perfluorocarbon core. Nanodroplets can vaporise into echogenic microbubbles forming cavitation nuclei when exposed to ultrasound. Their perfluorocarbon core phase-change is responsible for the acoustic droplet vaporisation. However, methods to quantify the perfluorocarbon core in nanodroplets are lacking. This is an important feature that can help explain nanodroplet phase change characteristics. In this study, we fabricated nanodroplets using lipids shell and perfluorocarbons. To assess the amount of perfluorocarbon in the core we used two methods, 19F NMR and FTIR. To assess the cavitation after vaporisation we used an ultrasound transducer (1.1 MHz) and a high-speed camera. The 19F NMR based method showed that the fluorine signal correlated accurately with the perfluorocarbon concentration. Using this correlation, we were able to quantify the perfluorocarbon core of nanodroplets. This method was used to assess the content of the perfluorocarbon of the nanodroplets in solutions over time. It was found that perfluoropentane nanodroplets lost their content faster and at higher ratio compared to perfluorohexane nanodroplets. The high-speed imaging indicates that the nanodroplets generate cavitation comparable to that from commercial contrast agent microbubbles. Nanodroplet characterisation should include perfluorocarbon concentration assessment as critical information for their development.


Asunto(s)
Fluorocarburos , Nanopartículas , Ultrasonografía , Nanopartículas/química , Volatilización , Medios de Contraste/química , Fluorocarburos/química , Microburbujas
4.
Science ; 378(6622): eabm7466, 2022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36423280

RESUMEN

Neurons use local protein synthesis to support their morphological complexity, which requires independent control across multiple subcellular compartments up to the level of individual synapses. We identify a signaling pathway that regulates the local synthesis of proteins required to form excitatory synapses on parvalbumin-expressing (PV+) interneurons in the mouse cerebral cortex. This process involves regulation of the TSC subunit 2 (Tsc2) by the Erb-B2 receptor tyrosine kinase 4 (ErbB4), which enables local control of messenger RNA {mRNA} translation in a cell type-specific and synapse type-specific manner. Ribosome-associated mRNA profiling reveals a molecular program of synaptic proteins downstream of ErbB4 signaling required to form excitatory inputs on PV+ interneurons. Thus, specific connections use local protein synthesis to control synapse formation in the nervous system.


Asunto(s)
Corteza Cerebral , Interneuronas , Biosíntesis de Proteínas , Receptor ErbB-4 , Sinapsis , Proteína 2 del Complejo de la Esclerosis Tuberosa , Animales , Ratones , Corteza Cerebral/metabolismo , Interneuronas/metabolismo , Receptor ErbB-4/genética , Receptor ErbB-4/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sinapsis/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo
5.
JCI Insight ; 6(18)2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34403370

RESUMEN

Venous valve (VV) failure causes chronic venous insufficiency, but the molecular regulation of valve development is poorly understood. A primary lymphatic anomaly, caused by mutations in the receptor tyrosine kinase EPHB4, was recently described, with these patients also presenting with venous insufficiency. Whether the venous anomalies are the result of an effect on VVs is not known. VV formation requires complex "organization" of valve-forming endothelial cells, including their reorientation perpendicular to the direction of blood flow. Using quantitative ultrasound, we identified substantial VV aplasia and deep venous reflux in patients with mutations in EPHB4. We used a GFP reporter in mice to study expression of its ligand, ephrinB2, and analyzed developmental phenotypes after conditional deletion of floxed Ephb4 and Efnb2 alleles. EphB4 and ephrinB2 expression patterns were dynamically regulated around organizing valve-forming cells. Efnb2 deletion disrupted the normal endothelial expression patterns of the gap junction proteins connexin37 and connexin43 (both required for normal valve development) around reorientating valve-forming cells and produced deficient valve-forming cell elongation, reorientation, polarity, and proliferation. Ephb4 was also required for valve-forming cell organization and subsequent growth of the valve leaflets. These results uncover a potentially novel cause of primary human VV aplasia.


Asunto(s)
Efrina-B2/genética , Receptor EphB4/genética , Receptor EphB4/metabolismo , Válvulas Venosas/anomalías , Válvulas Venosas/embriología , Animales , Aorta/ultraestructura , Comunicación Celular , Polaridad Celular , Proliferación Celular , Conexina 43/metabolismo , Conexinas/metabolismo , Endotelio , Efrina-B2/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación , Fenotipo , Ultrasonografía , Malformaciones Vasculares/diagnóstico por imagen , Malformaciones Vasculares/genética , Insuficiencia Venosa/diagnóstico por imagen , Válvulas Venosas/diagnóstico por imagen , Proteína alfa-4 de Unión Comunicante
6.
Redox Biol ; 38: 101816, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33340902

RESUMEN

Activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway is critical for vascular endothelial redox homeostasis in regions of high, unidirectional shear stress (USS), however the underlying mechanosensitive mediators are not fully understood. The endothelial glycocalyx is disrupted in arterial areas exposed to disturbed blood flow that also exhibit enhanced oxidative stress leading to atherogenesis. We investigated the contribution of glycocalyx sialic acids (SIA) to Nrf2 signaling in human endothelial cells (EC) exposed to atheroprotective USS or atherogenic low oscillatory shear stress (OSS). Cells exposed to USS exhibited a thicker glycocalyx and enhanced turnover of SIA which was reduced in cells cultured under OSS. Physiological USS, but not disturbed OSS, enhanced Nrf2-mediated expression of antioxidant enzymes, which was attenuated following SIA cleavage with exogenous neuraminidase. SIA removal disrupted kinase signaling involved in the nuclear accumulation of Nrf2 elicited by USS and promoted mitochondrial reactive oxygen species accumulation. Notably, knockdown of the endogenous sialidase NEU1 potentiated Nrf2 target gene expression, directly implicating SIA in regulation of Nrf2 signaling by USS. In the absence of SIA, deficits in Nrf2 responses to physiological flow were also associated with a pro-inflammatory EC phenotype. This study demonstrates that the glycocalyx modulates endothelial redox state in response to shear stress and provides the first evidence of an atheroprotective synergism between SIA and Nrf2 antioxidant signaling. The endothelial glycocalyx therefore represents a potential therapeutic target against EC dysfunction in cardiovascular disease and redox dyshomeostasis in ageing.


Asunto(s)
Células Endoteliales , Factor 2 Relacionado con NF-E2 , Células Endoteliales/metabolismo , Glicocálix/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Ácidos Siálicos , Estrés Mecánico
7.
Dev Cell ; 54(1): 33-42.e9, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32585131

RESUMEN

The extracellular matrix (ECM) is a polymer network hypothesized to form a stable cellular scaffold. While the ECM can undergo acute remodeling during embryogenesis, it is experimentally difficult to determine whether basal turnover is also important. Most studies of homeostatic turnover assume an initial steady-state balance of production and degradation and measure half-life by quantifying the rate of decay after experimental intervention (e.g., pulse labeling). Here, we present an intervention-free approach to mathematically model basal ECM turnover during embryogenesis by exploiting our ability to live image de novo ECM development in Drosophila to quantify production from initiation to homeostasis. This reveals rapid turnover (half-life ∼7-10 h), which we confirmed by in vivo pulse-chase experiments. Moreover, ECM turnover is partially dependent on proteolysis and network interactions, and slowing turnover affects tissue morphogenesis. These data demonstrate that embryonic ECM undergoes constant replacement, which is likely necessary to maintain network plasticity to accommodate growth and morphogenesis.


Asunto(s)
Matriz Extracelular/metabolismo , Homeostasis , Morfogénesis , Animales , Membrana Basal/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Modelos Teóricos
8.
N Engl J Med ; 382(9): 835-844, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32101665

RESUMEN

Mutations in VHL, which encodes von Hippel-Lindau tumor suppressor (VHL), are associated with divergent diseases. We describe a patient with marked erythrocytosis and prominent mitochondrial alterations associated with a severe germline VHL deficiency due to homozygosity for a novel synonymous mutation (c.222C→A, p.V74V). The condition is characterized by early systemic onset and differs from Chuvash polycythemia (c.598C→T) in that it is associated with a strongly reduced growth rate, persistent hypoglycemia, and limited exercise capacity. We report changes in gene expression that reprogram carbohydrate and lipid metabolism, impair muscle mitochondrial respiratory function, and uncouple oxygen consumption from ATP production. Moreover, we identified unusual intermitochondrial connecting ducts. Our findings add unexpected information on the importance of the VHL-hypoxia-inducible factor (HIF) axis to human phenotypes. (Funded by Associazione Italiana Ricerca sul Cancro and others.).


Asunto(s)
Mutación de Línea Germinal , Trastornos del Crecimiento/genética , Hipoglucemia/genética , Factor 1 Inducible por Hipoxia/deficiencia , Mitocondrias/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Expresión Génica , Crecimiento/genética , Humanos , Masculino , Metaboloma/genética , Metaboloma/fisiología , Síndrome , Adulto Joven
9.
J Invest Dermatol ; 140(1): 121-131.e6, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31326396

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is a debilitating genodermatosis caused by loss-of-function mutations in COL7A1 encoding type VII collagen (C7), the main component of anchoring fibrils at the dermal-epidermal junction. With no curative treatments presently available, retrovirally transduced autologous epidermal grafts and intradermal lentivirally engineered fibroblast injections are being investigated. Alternative approaches aim to infuse allogeneic mesenchymal stromal cells (MSCs) to provide a more generalized treatment for RDEB. We investigated whether healthy human MSCs could be engineered to overexpress C7 and correct RDEB in a human:murine chimeric model. Initially, engineered MSCs incorporated ex vivo into RDEB grafts, their presence confirmed by fluorescence in situ hybridization, revealed recovery of function of the dermal-epidermal junction with no signs of blister formation. Importantly, the detection of anchoring fibrils by transmission electron microscopy corroborated structural recovery. Next, MSCs cotransduced to express C7 and luciferase were delivered intradermally into grafted RDEB skin, resulting in localized MSC persistence with deposition of de novo C7 at the site. Notably, C7 expression was sufficient to restore anchoring fibril density to normal levels. In contrast, intravenously injected engineered MSCs were undetectable within grafts and lacked anchoring fibril reconstitution. Our data suggest that although localized correction may be achievable using engineered MSCs, strategies for systemic administration require further modeling.


Asunto(s)
Colágeno Tipo VII/metabolismo , Epidermólisis Ampollosa Distrófica/metabolismo , Células Madre Mesenquimatosas/fisiología , Reticulina/metabolismo , Piel/patología , Animales , Colágeno Tipo VII/genética , Epidermólisis Ampollosa Distrófica/genética , Epidermólisis Ampollosa Distrófica/patología , Ingeniería Genética , Humanos , Ratones , Ratones SCID , Microscopía Electrónica de Transmisión , Mutación/genética , Reticulina/ultraestructura , Trasplante de Piel , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Quimera por Trasplante
10.
Autophagy ; 16(6): 1044-1060, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31517566

RESUMEN

Macroautophagy/autophagy can enable cancer cells to withstand cellular stress and maintain bioenergetic homeostasis by sequestering cellular components into newly formed double-membrane vesicles destined for lysosomal degradation, potentially affecting the efficacy of anti-cancer treatments. Using 13C-labeled choline and 13C-magnetic resonance spectroscopy and western blotting, we show increased de novo choline phospholipid (ChoPL) production and activation of PCYT1A (phosphate cytidylyltransferase 1, choline, alpha), the rate-limiting enzyme of phosphatidylcholine (PtdCho) synthesis, during autophagy. We also discovered that the loss of PCYT1A activity results in compromised autophagosome formation and maintenance in autophagic cells. Direct tracing of ChoPLs with fluorescence and immunogold labeling imaging revealed the incorporation of newly synthesized ChoPLs into autophagosomal membranes, endoplasmic reticulum (ER) and mitochondria during anticancer drug-induced autophagy. Significant increase in the colocalization of fluorescence signals from the newly synthesized ChoPLs and mCherry-MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3) was also found on autophagosomes accumulating in cells treated with autophagy-modulating compounds. Interestingly, cells undergoing active autophagy had an altered ChoPL profile, with longer and more unsaturated fatty acid/alcohol chains detected. Our data suggest that de novo synthesis may be required to increase autophagosomal ChoPL content and alter its composition, together with replacing phospholipids consumed from other organelles during autophagosome formation and turnover. This addiction to de novo ChoPL synthesis and the critical role of PCYT1A may lead to development of agents targeting autophagy-induced drug resistance. In addition, fluorescence imaging of choline phospholipids could provide a useful way to visualize autophagosomes in cells and tissues. ABBREVIATIONS: AKT: AKT serine/threonine kinase; BAX: BCL2 associated X, apoptosis regulator; BECN1: beclin 1; ChoPL: choline phospholipid; CHKA: choline kinase alpha; CHPT1: choline phosphotransferase 1; CTCF: corrected total cell fluorescence; CTP: cytidine-5'-triphosphate; DCA: dichloroacetate; DMEM: dulbeccos modified Eagles medium; DMSO: dimethyl sulfoxide; EDTA: ethylenediaminetetraacetic acid; ER: endoplasmic reticulum; GDPD5: glycerophosphodiester phosphodiesterase domain containing 5; GFP: green fluorescent protein; GPC: glycerophosphorylcholine; HBSS: hanks balances salt solution; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; LPCAT1: lysophosphatidylcholine acyltransferase 1; LysoPtdCho: lysophosphatidylcholine; MRS: magnetic resonance spectroscopy; MTORC1: mechanistic target of rapamycin kinase complex 1; PCho: phosphocholine; PCYT: choline phosphate cytidylyltransferase; PLA2: phospholipase A2; PLB: phospholipase B; PLC: phospholipase C; PLD: phospholipase D; PCYT1A: phosphate cytidylyltransferase 1, choline, alpha; PI3K: phosphoinositide-3-kinase; pMAFs: pancreatic mouse adult fibroblasts; PNPLA6: patatin like phospholipase domain containing 6; Pro-Cho: propargylcholine; Pro-ChoPLs: propargylcholine phospholipids; PtdCho: phosphatidylcholine; PtdEth: phosphatidylethanolamine; PtdIns3P: phosphatidylinositol-3-phosphate; RPS6: ribosomal protein S6; SCD: stearoyl-CoA desaturase; SEM: standard error of the mean; SM: sphingomyelin; SMPD1/SMase: sphingomyelin phosphodiesterase 1, acid lysosomal; SGMS: sphingomyelin synthase; WT: wild-type.


Asunto(s)
Antineoplásicos/farmacología , Autofagosomas/enzimología , Autofagosomas/metabolismo , Citidililtransferasa de Colina-Fosfato/metabolismo , Furanos/farmacología , Macroautofagia , Fosfatidilcolinas/biosíntesis , Piridinas/farmacología , Pirimidinas/farmacología , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/ultraestructura , Células CHO , Línea Celular Tumoral , Colina/metabolismo , Citidililtransferasa de Colina-Fosfato/genética , Cricetulus , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/enzimología , Membranas Intracelulares/metabolismo , Macroautofagia/efectos de los fármacos , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Metabolómica , Ratones , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
11.
Cell Rep ; 27(13): 3956-3971.e6, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31242426

RESUMEN

Senescence is a cellular phenotype present in health and disease, characterized by a stable cell-cycle arrest and an inflammatory response called senescence-associated secretory phenotype (SASP). The SASP is important in influencing the behavior of neighboring cells and altering the microenvironment; yet, this role has been mainly attributed to soluble factors. Here, we show that both the soluble factors and small extracellular vesicles (sEVs) are capable of transmitting paracrine senescence to nearby cells. Analysis of individual cells internalizing sEVs, using a Cre-reporter system, show a positive correlation between sEV uptake and senescence activation. We find an increase in the number of multivesicular bodies during senescence in vivo. sEV protein characterization by mass spectrometry (MS) followed by a functional siRNA screen identify interferon-induced transmembrane protein 3 (IFITM3) as being partially responsible for transmitting senescence to normal cells. We find that sEVs contribute to paracrine senescence.


Asunto(s)
Microambiente Celular , Vesículas Extracelulares/metabolismo , Proteínas de la Membrana/metabolismo , Comunicación Paracrina , Proteínas de Unión al ARN/metabolismo , Femenino , Células HEK293 , Humanos , Células MCF-7 , Masculino
12.
Regen Med ; 13(7): 785-801, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30289057

RESUMEN

AIM: Scaffolds are a promising approach for spinal cord injury (SCI) treatment. FGF-2 is involved in tissue repair but is easily degradable and presents collateral effects in systemic administration. In order to address the stability issue and avoid the systemic effects, FGF-2 was encapsulated into core-shell microfibers by coaxial electrospinning and its in vitro and in vivo potential were studied. Materials & methods: The fibers were characterized by physicochemical and biological parameters. The scaffolds were implanted in a hemisection SCI rat model. Locomotor test was performed weekly for 6 weeks. After this time, histological analyses were performed and expression of nestin and GFAP was quantified by flow cytometry. Results: Electrospinning resulted in uniform microfibers with a core-shell structure, with a sustained liberation of FGF-2 from the fibers. The fibers supported PC12 cells adhesion and proliferation. Implanted scaffolds into SCI promoted locomotor recovery at 28 days after injury and reduced GFAP expression. CONCLUSION: These results indicate the potential of these microfibers in SCI tissue engineering. [Formula: see text].


Asunto(s)
Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Médula Espinal/patología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Ensayo de Materiales , Células PC12 , Ratas , Médula Espinal/metabolismo , Médula Espinal/ultraestructura , Traumatismos de la Médula Espinal/terapia
13.
Nat Cell Biol ; 20(3): 307-319, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29434374

RESUMEN

Reactive oxygen species (ROS) contribute to tissue damage and remodelling mediated by the inflammatory response after injury. Here we show that ROS, which promote axonal dieback and degeneration after injury, are also required for axonal regeneration and functional recovery after spinal injury. We find that ROS production in the injured sciatic nerve and dorsal root ganglia requires CX3CR1-dependent recruitment of inflammatory cells. Next, exosomes containing functional NADPH oxidase 2 complexes are released from macrophages and incorporated into injured axons via endocytosis. Once in axonal endosomes, active NOX2 is retrogradely transported to the cell body through an importin-ß1-dynein-dependent mechanism. Endosomal NOX2 oxidizes PTEN, which leads to its inactivation, thus stimulating PI3K-phosporylated (p-)Akt signalling and regenerative outgrowth. Challenging the view that ROS are exclusively involved in nerve degeneration, we propose a previously unrecognized role of ROS in mammalian axonal regeneration through a NOX2-PI3K-p-Akt signalling pathway.


Asunto(s)
Axones/enzimología , Exosomas/enzimología , Ganglios Espinales/enzimología , NADPH Oxidasa 2/metabolismo , Degeneración Nerviosa , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/enzimología , Especies Reactivas de Oxígeno/metabolismo , Nervio Ciático/enzimología , Traumatismos de la Médula Espinal/enzimología , Animales , Axones/patología , Receptor 1 de Quimiocinas CX3C/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Dineínas/metabolismo , Endocitosis , Endosomas/enzimología , Endosomas/patología , Exosomas/patología , Ganglios Espinales/lesiones , Ganglios Espinales/patología , Macrófagos/enzimología , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 2/deficiencia , NADPH Oxidasa 2/genética , Proteínas Nucleares/metabolismo , Fosfohidrolasa PTEN/metabolismo , Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/patología , Traumatismos de los Nervios Periféricos/fisiopatología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Transducción de Señal , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , beta Carioferinas
14.
Nat Microbiol ; 3(4): 447-455, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29459732

RESUMEN

Malaria parasites replicate within a parasitophorous vacuole in red blood cells (RBCs). Progeny merozoites egress upon rupture of first the parasitophorous vacuole membrane (PVM), then poration and rupture of the RBC membrane (RBCM). Egress is protease-dependent 1 , but none of the effector molecules that mediate membrane rupture have been identified and it is unknown how sequential rupture of the two membranes is controlled. Minutes before egress, the parasite serine protease SUB1 is discharged into the parasitophorous vacuole2-6 where it cleaves multiple substrates2,5,7-9 including SERA6, a putative cysteine protease10-12. Here, we show that Plasmodium falciparum parasites lacking SUB1 undergo none of the morphological transformations that precede egress and fail to rupture the PVM. In contrast, PVM rupture and RBCM poration occur normally in SERA6-null parasites but RBCM rupture does not occur. Complementation studies show that SERA6 is an enzyme that requires processing by SUB1 to function. RBCM rupture is associated with SERA6-dependent proteolytic cleavage within the actin-binding domain of the major RBC cytoskeletal protein ß-spectrin. We conclude that SUB1 and SERA6 play distinct, essential roles in a coordinated proteolytic cascade that enables sequential rupture of the two bounding membranes and culminates in RBCM disruption through rapid, precise, SERA6-mediated disassembly of the RBC cytoskeleton.


Asunto(s)
Proteasas de Cisteína/metabolismo , Eritrocitos/metabolismo , Malaria Falciparum/patología , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Serina Proteasas/metabolismo , Membrana Celular/metabolismo , Proteasas de Cisteína/genética , Citoesqueleto/metabolismo , Eritrocitos/parasitología , Humanos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética
15.
Curr Biol ; 27(22): 3526-3534.e4, 2017 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-29129537

RESUMEN

The basement membrane (BM) is a thin layer of extracellular matrix (ECM) beneath nearly all epithelial cell types that is critical for cellular and tissue function. It is composed of numerous components conserved among all bilaterians [1]; however, it is unknown how all of these components are generated and subsequently constructed to form a fully mature BM in the living animal. Although BM formation is thought to simply involve a process of self-assembly [2], this concept suffers from a number of logistical issues when considering its construction in vivo. First, incorporation of BM components appears to be hierarchical [3-5], yet it is unclear whether their production during embryogenesis must also be regulated in a temporal fashion. Second, many BM proteins are produced not only by the cells residing on the BM but also by surrounding cell types [6-9], and it is unclear how large, possibly insoluble protein complexes [10] are delivered into the matrix. Here we exploit our ability to live image and genetically dissect de novo BM formation during Drosophila development. This reveals that there is a temporal hierarchy of BM protein production that is essential for proper component incorporation. Furthermore, we show that BM components require secretion by migrating macrophages (hemocytes) during their developmental dispersal, which is critical for embryogenesis. Indeed, hemocyte migration is essential to deliver a subset of ECM components evenly throughout the embryo. This reveals that de novo BM construction requires a combination of both production and distribution logistics allowing for the timely delivery of core components.


Asunto(s)
Membrana Basal/fisiología , Matriz Extracelular/metabolismo , Animales , Membrana Basal/metabolismo , Movimiento Celular/fisiología , Colágeno/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Células Epiteliales/metabolismo , Matriz Extracelular/fisiología , Macrófagos/metabolismo
16.
PLoS One ; 12(3): e0173474, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28362799

RESUMEN

Pentamidine is an effective trypanocidal drug used against stage 1 Human African Trypanosomiasis (HAT). At the blood-brain barrier (BBB), it accumulates inside the endothelial cells but has limited entry into the brain. This study examined transporters involved in pentamidine transport at the human and mouse BBB using hCMEC/D3 and bEnd.3 cell lines, respectively. Results revealed that both cell lines expressed the organic cation transporters (OCT1, OCT2 and OCT3), however, P-gp was only expressed in hCMEC/D3 cells. Polarised expression of OCT1 was also observed. Functional assays found that ATP depletion significantly increased [3H]pentamidine accumulation in hCMEC/D3 cells (***p<0.001) but not in bEnd.3 cells. Incubation with unlabelled pentamidine significantly decreased accumulation in hCMEC/D3 and bEnd.3 cells after 120 minutes (***p<0.001). Treating both cell lines with haloperidol and amantadine also decreased [3H]pentamidine accumulation significantly (***p<0.001 and **p<0.01 respectively). However, prazosin treatment decreased [3H]pentamidine accumulation only in hCMEC/D3 cells (*p<0.05), and not bEnd.3 cells. Furthermore, the presence of OCTN, MATE, PMAT, ENT or CNT inhibitors/substrates had no significant effect on the accumulation of [3H]pentamidine in both cell lines. From the data, we conclude that pentamidine interacts with multiple transporters, is taken into brain endothelial cells by OCT1 transporter and is extruded into the blood by ATP-dependent mechanisms. These interactions along with the predominant presence of OCT1 in the luminal membrane of the BBB contribute to the limited entry of pentamidine into the brain. This information is of key importance to the development of pentamidine based combination therapies which could be used to treat CNS stage HAT by improving CNS delivery, efficacy against trypanosomes and safety profile of pentamidine.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Transportador 1 de Catión Orgánico/metabolismo , Pentamidina/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico/genética , Transporte Biológico/fisiología , Western Blotting , Encéfalo/metabolismo , Línea Celular , Electroforesis en Gel de Poliacrilamida , Humanos , Ratones , Microscopía Confocal , Microscopía Electrónica de Transmisión , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 1 de Catión Orgánico/genética , Transportador 2 de Cátion Orgánico
17.
Proc Natl Acad Sci U S A ; 114(13): 3439-3444, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28292906

RESUMEN

In the asexual blood stages of malarial infection, merozoites invade erythrocytes and replicate within a parasitophorous vacuole to form daughter cells that eventually exit (egress) by sequential rupture of the vacuole and erythrocyte membranes. The current model is that PKG, a malarial cGMP-dependent protein kinase, triggers egress, activating malarial proteases and other effectors. Using selective inhibitors of either PKG or cysteine proteases to separately inhibit the sequential steps in membrane perforation, combined with video microscopy, electron tomography, electron energy loss spectroscopy, and soft X-ray tomography of mature intracellular Plasmodium falciparum parasites, we resolve intermediate steps in egress. We show that the parasitophorous vacuole membrane (PVM) is permeabilized 10-30 min before its PKG-triggered breakdown into multilayered vesicles. Just before PVM breakdown, the host red cell undergoes an abrupt, dramatic shape change due to the sudden breakdown of the erythrocyte cytoskeleton, before permeabilization and eventual rupture of the erythrocyte membrane to release the parasites. In contrast to the previous view of PKG-triggered initiation of egress and a gradual dismantling of the host erythrocyte cytoskeleton over the course of schizont development, our findings identify an initial step in egress and show that host cell cytoskeleton breakdown is restricted to a narrow time window within the final stages of egress.


Asunto(s)
Citoesqueleto/metabolismo , Membrana Eritrocítica/parasitología , Eritrocitos/parasitología , Malaria Falciparum/parasitología , Plasmodium falciparum/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Citoesqueleto/genética , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Humanos , Malaria Falciparum/genética , Malaria Falciparum/metabolismo , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
18.
Biochem J ; 474(9): 1495-1508, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28280110

RESUMEN

The twin-arginine translocation (Tat) system is an integral membrane protein complex that accomplishes the remarkable feat of transporting large, fully folded polypeptides across the inner membrane of bacteria, into the periplasm. In Escherichia coli, Tat comprises three membrane proteins: TatA, TatB and TatC. How these proteins arrange themselves in the inner membrane to permit passage of Tat substrates, whilst maintaining membrane integrity, is still poorly understood. TatA is the most abundant component of this complex and facilitates assembly of the transport mechanism. We have utilised immunogold labelling in combination with array tomography to gain insight into the localisation and distribution of the TatA protein in E. coli cells. We show that TatA exhibits a uniform distribution throughout the inner membrane of E. coli and that altering the expression of TatBC shows a previously uncharacterised distribution of TatA in the inner membrane. Array tomography was used to provide our first insight into this altered distribution of TatA in three-dimensional space, revealing that this protein forms linear clusters in the inner membrane of E. coli upon increased expression of TatBC. This is the first indication that TatA organisation in the inner membrane alters in response to changes in Tat subunit stoichiometry.


Asunto(s)
Proteínas de Escherichia coli/ultraestructura , Imagenología Tridimensional/métodos , Proteínas de Transporte de Membrana/ultraestructura , Microscopía Inmunoelectrónica/métodos , Complejos Multiproteicos/ultraestructura , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/ultraestructura , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Operón , Unión Proteica , Transporte de Proteínas
19.
Am J Dermatopathol ; 38(3): 239-43, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26894777

RESUMEN

Woolly hair nevus is a mosaic disorder characterized by unruly, tightly curled hair in a circumscribed area of the scalp. This condition may be associated with epidermal nevi. We describe an 11-year-old boy who initially presented with multiple patches of woolly hair and with epidermal nevi on his left cheek and back. He had no nail, teeth, eye, or cardiac abnormalities. Analysis of plucked hairs from patches of woolly hair showed twisting of the hair shaft and an abnormal hair cuticle. Histopathology of a woolly hair patch showed diffuse hair follicle miniaturization with increased vellus hairs.


Asunto(s)
Enfermedades del Cabello/patología , Folículo Piloso/fisiología , Folículo Piloso/ultraestructura , Niño , Humanos , Masculino , Microscopía Electrónica de Rastreo , Mosaicismo , Nevo/patología
20.
EMBO Mol Med ; 8(3): 191-207, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26881968

RESUMEN

WBP2 encodes the WW domain-binding protein 2 that acts as a transcriptional coactivator for estrogen receptor α (ESR1) and progesterone receptor (PGR). We reported that the loss of Wbp2 expression leads to progressive high-frequency hearing loss in mouse, as well as in two deaf children, each carrying two different variants in the WBP2 gene. The earliest abnormality we detect in Wbp2-deficient mice is a primary defect at inner hair cell afferent synapses. This study defines a new gene involved in the molecular pathway linking hearing impairment to hormonal signalling and provides new therapeutic targets.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cóclea/fisiología , Pérdida Auditiva/patología , Pérdida Auditiva/fisiopatología , Audición , Sinapsis/fisiología , Animales , Humanos , Ratones , Transactivadores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA