Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(3): e1011281, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37000891

RESUMEN

During the blood stage of a malaria infection, malaria parasites export both soluble and membrane proteins into the erythrocytes in which they reside. Exported proteins are trafficked via the parasite endoplasmic reticulum and secretory pathway, before being exported across the parasitophorous vacuole membrane into the erythrocyte. Transport across the parasitophorous vacuole membrane requires protein unfolding, and in the case of membrane proteins, extraction from the parasite plasma membrane. We show that trafficking of the exported Plasmodium protein, Pf332, differs from that of canonical eukaryotic soluble-secreted and transmembrane proteins. Pf332 is initially ER-targeted by an internal hydrophobic sequence that unlike a signal peptide, is not proteolytically removed, and unlike a transmembrane segment, does not span the ER membrane. Rather, both termini of the hydrophobic sequence enter the ER lumen and the ER-lumenal species is a productive intermediate for protein export. Furthermore, we show in intact cells, that two other exported membrane proteins, SBP1 and MAHRP2, assume a lumenal topology within the parasite secretory pathway. Although the addition of a C-terminal ER-retention sequence, recognised by the lumenal domain of the KDEL receptor, does not completely block export of SBP1 and MAHRP2, it does enhance their retention in the parasite ER. This indicates that a sub-population of each protein adopts an ER-lumenal state that is an intermediate in the export process. Overall, this suggests that although many exported proteins traverse the parasite secretory pathway as typical soluble or membrane proteins, some exported proteins that are ER-targeted by a transmembrane segment-like, internal, non-cleaved hydrophobic segment, do not integrate into the ER membrane, and form an ER-lumenal species that is a productive export intermediate. This represents a novel means, not seen in typical membrane proteins found in model systems, by which exported transmembrane-like proteins can be targeted and trafficked within the lumen of the secretory pathway.


Asunto(s)
Malaria , Plasmodium , Humanos , Transporte de Proteínas , Proteínas Protozoarias/metabolismo , Plasmodium/metabolismo , Retículo Endoplásmico/metabolismo , Eritrocitos/parasitología , Malaria/metabolismo , Proteínas de la Membrana/metabolismo , Plasmodium falciparum/metabolismo
2.
Gene Ther ; 30(9): 723-735, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37386155

RESUMEN

Adeno-associated virus serotype 2 (AAV2) is a viral vector that can be used to deliver therapeutic genes to diseased cells in the retina. One strategy for altering AAV2 vectors involves the mutation of phosphodegron residues, which are thought to be phosphorylated/ubiquitinated in the cytosol, facilitating degradation of the vector and the inhibition of transduction. As such, mutation of phosphodegron residues have been correlated with increased transduction of target cells, however, an assessment of the immunobiology of wild-type and phosphodegron mutant AAV2 vectors following intravitreal (IVT) delivery to immunocompetent animals is lacking in the current literature. In this study, we show that IVT of a triple phosphodegron mutant AAV2 capsid is associated with higher levels of humoral immune activation, infiltration of CD4 and CD8 T-cells into the retina, generation of splenic germinal centre reactions, activation of conventional dendritic cell subsets, and elevated retinal gliosis compared to wild-type AAV2 capsids. However, we did not detect significant changes in electroretinography arising after vector administration. We also demonstrate that the triple AAV2 mutant capsid is less susceptible to neutralisation by soluble heparan sulphate and anti-AAV2 neutralising antibodies, highlighting a possible utility for the vector in terms of circumventing pre-existing humoral immunity. In summary, the present study highlights novel aspects of rationally-designed vector immunobiology, which may be relevant to their application in preclinical and clinical settings.


Asunto(s)
Cápside , Parvovirinae , Ratones , Animales , Cápside/metabolismo , Serogrupo , Transducción Genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Parvovirinae/genética , Dependovirus/metabolismo , Vectores Genéticos/genética
3.
Gene Ther ; 30(6): 503-519, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36635457

RESUMEN

Recombinant adeno-associated viral vectors (AAVs) are an effective system for gene transfer. AAV serotype 2 (AAV2) is commonly used to deliver transgenes to retinal ganglion cells (RGCs) via intravitreal injection. The AAV serotype however is not the only factor contributing to the effectiveness of gene therapies. Promoters influence the strength and cell-selectivity of transgene expression. This study compares five promoters designed to maximise AAV2 cargo space for gene delivery: chicken ß-actin (CBA), cytomegalovirus (CMV), short CMV early enhancer/chicken ß-actin/short ß-globulin intron (sCAG), mouse phosphoglycerate kinase (PGK), and human synapsin (SYN). The promoters driving enhanced green fluorescent protein (eGFP) were examined in adult C57BL/6J mice eyes and tissues of the visual system. eGFP expression was strongest in the retina, optic nerves and brain when driven by the sCAG and SYN promoters. CBA, CMV, and PGK had moderate expression by comparison. The SYN promoter had almost exclusive transgene expression in RGCs. The PGK promoter had predominant expression in both RGCs and AII amacrine cells. The ubiquitous CBA, CMV, and sCAG promoters expressed eGFP in a variety of cell types across multiple retinal layers including Müller glia and astrocytes. We also found that these promoters could transduce human retina ex vivo, although expression was predominantly in glial cells due to low RGC viability. Taken together, this promoter comparison study contributes to optimising AAV-mediated transduction in the retina, and could be valuable for research in ocular disorders, particularly those with large or complex genetic cargos.


Asunto(s)
Infecciones por Citomegalovirus , Parvovirinae , Ratones , Animales , Humanos , Células Ganglionares de la Retina/metabolismo , Actinas/genética , Actinas/metabolismo , Transducción Genética , Ratones Endogámicos C57BL , Transgenes , Dependovirus/genética , Dependovirus/metabolismo , Parvovirinae/genética , Proteínas Fluorescentes Verdes/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Vectores Genéticos/genética
4.
Exp Eye Res ; 213: 108793, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34656549

RESUMEN

Membrane contact sites (MCS) play crucial roles in cell physiology with dysfunction in several MCS proteins being linked with neurological and optic nerve diseases. Although there have been significant advances in imaging these interactions over the past two decades with advanced electron microscopy techniques, super-resolution imaging and proximity-dependent fluorescent reporters, a technique to observe and quantify MCS in mammalian optic nerve tissues has not yet been reported. We demonstrate for the first time that proximity ligation assay (PLA), a technique already used in mammalian cell lines, can be used as an efficient method of quantifying inter-organelle contact sites, namely mitochondria-endoplasmic reticulum (ER) and mitochondria-late-endosomes, in mammalian optic nerve tissues treated with adeno-associated virus (AAV) gene therapy with wild-type or phosphomimetic (active) protrudin. PLA utilises complementary single-stranded DNA oligomers bound to secondary antibodies that hybridise and complete a circular piece of DNA when the primary antibodies of interest interact. These interactions can be detected by amplifying the circular DNA and adding fluorescent probes. We show that PLA is a useful method that can be used to quantify MCS in optic nerve tissues. We have found that upregulation of protrudin with gene therapy significantly increases the number of mitochondria-ER and mitochondria-Rab7-late endosomes contact sites in optic nerves.


Asunto(s)
Bioensayo/métodos , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Membranas Mitocondriales/metabolismo , Nervio Óptico/metabolismo , Animales , Sitios de Unión , Dependovirus/genética , Femenino , Expresión Génica , Terapia Genética , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Orgánulos , Fijación del Tejido , Proteínas de Transporte Vesicular/genética
5.
Sensors (Basel) ; 21(6)2021 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-33801076

RESUMEN

Radiation detectors installed at major ports of entry are a key component of the overall strategy to protect countries from nuclear terrorism. While the goal of deploying these systems is to intercept special nuclear material as it enters the country, no detector system is foolproof. Mobile, distributed sensors have been proposed to detect nuclear materials in transit should portal monitors fail to prevent their entry in the first place. In large metropolitan areas, a mobile distributed sensor network could be deployed using vehicle platforms such as taxis, Ubers, and Lyfts, which are already connected to communications infrastructure. However, performance and coverage that could be achieved using a network of sensors mounted on commercial passenger vehicles has not been established. Here, we evaluate how a mobile sensor network could perform in New York City using a combination of radiation transport and geographic information systems. The geographic information system is used in conjunction with OpenStreetMap data to isolate roads and construct a grid over the streets. Vehicle paths are built using pickup and drop off data from Uber, and from the New York State Department of Transportation. The results show that the time to first detection increases with source velocity, decreases with the number of mobile detectors, and reaches a plateau that depends on the strength of the source.

6.
Stem Cells ; 36(1): 65-78, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29044808

RESUMEN

Optic neuropathies such as glaucoma occur when retinal ganglion cells (RGCs) in the eye are injured. Strong evidence suggests mesenchymal stem cells (MSCs) could be a potential therapy to protect RGCs; however, little is known regarding their effect on the human retina. We, therefore, investigated if human MSCs (hMSCs), or platelet-derived growth factor (PDGF) as produced by hMSC, could delay RGC death in a human retinal explant model of optic nerve injury. Our results showed hMSCs and the secreted growth factor PDGF-AB could substantially reduce human RGC loss and apoptosis following axotomy. The neuroprotective pathways AKT, ERK, and STAT3 were activated in the retina shortly after treatments with labeling seen in the RGC layer. A dose dependent protective effect of PDGF-AB was observed in human retinal explants but protection was not as substantial as that achieved by culturing hMSCs on the retina surface which resulted in RGC cell counts similar to those immediately post dissection. These results demonstrate that hMSCs and PDGF have strong neuroprotective action on human RGCs and may offer a translatable, therapeutic strategy to reduce degenerative visual loss. Stem Cells 2018;36:65-78.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Fármacos Neuroprotectores/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Ganglionares de la Retina/metabolismo , Humanos , Transducción de Señal
7.
Neural Plast ; 2018: 7108948, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29853847

RESUMEN

The eye is at the forefront of the application of gene therapy techniques to medicine. In the United States, a gene therapy treatment for Leber's congenital amaurosis, a rare inherited retinal disease, recently became the first gene therapy to be approved by the FDA for the treatment of disease caused by mutations in a specific gene. Phase III clinical trials of gene therapy for other single-gene defect diseases of the retina and optic nerve are also currently underway. However, for optic nerve diseases not caused by single-gene defects, gene therapy strategies are likely to focus on slowing or preventing neuronal death through the expression of neuroprotective agents. In addition to these strategies, there has also been recent interest in the potential use of precise genome editing techniques to treat ocular disease. This review focuses on recent developments in gene therapy techniques for the treatment of glaucoma and Leber's hereditary optic neuropathy (LHON). We discuss recent successes in clinical trials for the treatment of LHON using gene supplementation therapy, promising neuroprotective strategies that have been employed in animal models of glaucoma and the potential use of genome editing techniques in treating optic nerve disease.


Asunto(s)
Terapia Genética , Glaucoma/terapia , Atrofia Óptica Hereditaria de Leber/terapia , Células Ganglionares de la Retina/patología , Animales , Glaucoma/patología , Humanos , Atrofia Óptica Hereditaria de Leber/patología
8.
J Biol Chem ; 291(50): 26188-26207, 2016 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-27777305

RESUMEN

Repetitive low complexity sequences, mostly assumed to have no function, are common in proteins that are exported by the malaria parasite into its host erythrocyte. We identify a group of exported proteins containing short lysine-rich tandemly repeated sequences that are sufficient to localize to the erythrocyte periphery, where key virulence-related modifications to the plasma membrane and the underlying cytoskeleton are known to occur. Efficiency of targeting is dependent on repeat number, indicating that novel targeting modules could evolve by expansion of short lysine-rich sequences. Indeed, analysis of fragments of GARP from different species shows that two novel targeting sequences have arisen via the process of repeat expansion in this protein. In the protein Hyp12, the targeting function of a lysine-rich sequence is masked by a neighboring repetitive acidic sequence, further highlighting the importance of repetitive low complexity sequences. We show that sequences capable of targeting the erythrocyte periphery are present in at least nine proteins from Plasmodium falciparum and one from Plasmodium knowlesi We find these sequences in proteins known to be involved in erythrocyte rigidification and cytoadhesion as well as in previously uncharacterized exported proteins. Together, these data suggest that expansion and contraction of lysine-rich repeats could generate targeting sequences de novo as well as modulate protein targeting efficiency and function in response to selective pressure.


Asunto(s)
Eritrocitos/parasitología , Plasmodium falciparum , Plasmodium knowlesi , Proteínas Protozoarias , Animales , Eritrocitos/metabolismo , Humanos , Ratones , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium knowlesi/genética , Plasmodium knowlesi/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Secuencias Repetitivas de Aminoácido
9.
Stem Cells ; 33(10): 3006-16, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26175331

RESUMEN

A variety of diseases lead to degeneration of retinal ganglion cells (RGCs) and their axons within the optic nerve resulting in loss of visual function. Although current therapies may delay RGC loss, they do not restore visual function or completely halt disease progression. Regenerative medicine has recently focused on stem cell therapy for both neuroprotective and regenerative purposes. However, significant problems remain to be addressed, such as the long-term impact of reactive gliosis occurring in the host retina in response to transplanted stem cells. The aim of this work was to investigate retinal glial responses to intravitreally transplanted bone marrow mesenchymal stem cells (BM-MSCs) to help identify factors able to modulate graft-induced reactive gliosis. We found in vivo that intravitreal BM-MSC transplantation is associated with gliosis-mediated retinal folding, upregulation of intermediate filaments, and recruitment of macrophages. These responses were accompanied by significant JAK/STAT3 and MAPK (ERK1/2 and JNK) cascade activation in retinal Muller glia. Lipocalin-2 (Lcn-2) was identified as a potential new indicator of graft-induced reactive gliosis. Pharmacological inhibition of STAT3 in BM-MSC cocultured retinal explants successfully reduced glial fibrillary acidic protein expression in retinal Muller glia and increased BM-MSC retinal engraftment. Inhibition of stem cell-induced reactive gliosis is critical for successful transplantation-based strategies for neuroprotection, replacement, and regeneration of the optic nerve.


Asunto(s)
Gliosis/terapia , Trasplante de Células Madre Mesenquimatosas , Neuroglía/patología , Medicina Regenerativa , Animales , Axones/patología , Células de la Médula Ósea/citología , Células Ependimogliales/patología , Gliosis/patología , Humanos , Células Madre Mesenquimatosas , Ratones , Nervio Óptico/patología , Retina/crecimiento & desarrollo , Retina/patología , Células Ganglionares de la Retina/patología
10.
Exp Eye Res ; 143: 28-38, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26432917

RESUMEN

There is a growing need for models of human diseases that utilise native, donated human tissue in order to model disease processes and develop novel therapeutic strategies. In this paper we assessed the suitability of adult human retinal explants as a potential model of chronic retinal ganglion cell (RGC) degeneration. Our results confirmed that RGC markers commonly used in rodent studies (NeuN, ßIII Tubulin and Thy-1) were appropriate for labelling human RGCs and followed the expected differential expression patterns across, as well as throughout, the macular and para-macular regions of the retina. Furthermore, we showed that neither donor age nor post-mortem time (within 24 h) significantly affected the initial expression levels of RGC markers. In addition, the feasibility of using human post mortem donor tissue as a long-term model of RGC degeneration was determined with RGC protein being detectable up to 4 weeks in culture with an associated decline in RGC mRNA and significant, progressive, apoptotic labelling of NeuN(+) cells. Differences in RGC apoptosis might have been influenced by medium compositions indicating that media constituents could play a role in supporting axotomised RGCs. We propose that using ex vivo human explants may prove to be a useful model for testing the effectiveness of neuroprotective strategies.


Asunto(s)
Modelos Teóricos , Degeneración Nerviosa/patología , Células Ganglionares de la Retina/patología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos Nucleares/genética , Apoptosis , Biomarcadores/metabolismo , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Etiquetado Corte-Fin in Situ , Persona de Mediana Edad , Degeneración Nerviosa/metabolismo , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Ganglionares de la Retina/metabolismo , Antígenos Thy-1/genética , Donantes de Tejidos , Tubulina (Proteína)/genética
11.
Brain ; 137(Pt 2): 503-19, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24176979

RESUMEN

The development of neuroprotective strategies to attenuate retinal ganglion cell death could lead to novel therapies for chronic optic neuropathies such as glaucoma. Intravitreal transplantation of mesenchymal stem cells slows retinal ganglion cell death in models of optic nerve injury, but the mechanism of action remains unclear. Here we characterized the neuroprotective effects of mesenchymal stem cells and mesenchymal stem cell-derived factors in organotypic retinal explant culture and an in vivo model of ocular hypertensive glaucoma. Co-culture of rat and human bone marrow-derived mesenchymal stem cells with retinal explants increased retinal ganglion cell survival, after 7 days ex vivo, by ∼2-fold and was associated with reduced apoptosis and increased nerve fibre layer and inner plexiform layer thicknesses. These effects were not demonstrated by co-culture with human or mouse fibroblasts. Conditioned media from mesenchymal stem cells conferred neuroprotection, suggesting that the neuroprotection is mediated, at least partly, by secreted factors. We compared the concentrations of 29 factors in human mesenchymal stem cell and fibroblast conditioned media, and identified 11 enriched in the mesenchymal stem cell secretome. Treatment of retinal explants with a cocktail of these factors conferred retinal ganglion cell neuroprotection, with factors from the platelet-derived growth factor family being the most potent. Blockade of platelet-derived growth factor signalling with neutralizing antibody or with small molecule inhibitors of platelet-derived growth factor receptor kinase or downstream phosphatidylinositol 3 kinase eliminated retinal ganglion cell neuroprotection conferred by mesenchymal stem cell co-culture. Intravitreal injection of platelet-derived growth factor -AA or -AB led to profound optic nerve neuroprotection in vivo following experimental induction of elevated intraocular pressure. These data demonstrate that mesenchymal stem cells secrete a number of neuroprotective proteins and suggest that platelet-derived growth factor secretion in particular may play an important role in mesenchymal stem cell-mediated retinal ganglion cell neuroprotection. Furthermore, platelet-derived growth factor may represent an independent target for achieving retinal ganglion cell neuroprotection.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Fármacos Neuroprotectores/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Ganglionares de la Retina/metabolismo , Animales , Axotomía/efectos adversos , Técnicas de Cocultivo/métodos , Humanos , Células Madre Mesenquimatosas/patología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Células Ganglionares de la Retina/patología
12.
Mol Microbiol ; 87(4): 835-50, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23279267

RESUMEN

The malaria parasite exports proteins across its plasma membrane and a surrounding parasitophorous vacuole membrane, into its host erythrocyte. Most exported proteins contain a Host Targeting motif (HT motif) that targets them for export. In the parasite secretory pathway, the HT motif is cleaved by the protease plasmepsin V, but the role of the newly generated N-terminal sequence in protein export is unclear. Using a model protein that is cleaved by an exogenous viral protease, we show that the new N-terminal sequence, normally generated by plasmepsin V cleavage, is sufficient to target a protein for export, and that cleavage by plasmepsin V is not coupled directly to the transfer of a protein to the next component in the export pathway. Mutation of the fourth and fifth positions of the HT motif, as well as amino acids further downstream, block or affect the efficiency of protein export indicating that this region is necessary for efficient export. We also show that the fifth position of the HT motif is important for plasmepsin V cleavage. Our results indicate that plasmepsin V cleavage is required to generate a new N-terminal sequence that is necessary and sufficient to mediate protein export by the malaria parasite.


Asunto(s)
Malaria Falciparum/parasitología , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Secuencias de Aminoácidos , Ácido Aspártico Endopeptidasas/metabolismo , Interacciones Huésped-Parásitos , Humanos , Malaria Falciparum/enzimología , Plasmodium falciparum/química , Plasmodium falciparum/genética , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Protozoarias/genética
13.
Nature ; 455(7215): 984-7, 2008 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-18923526

RESUMEN

An important step in the biosynthesis of many proteins is their partial or complete translocation across the plasma membrane in prokaryotes or the endoplasmic reticulum membrane in eukaryotes. In bacteria, secretory proteins are generally translocated after completion of their synthesis by the interaction of the cytoplasmic ATPase SecA and a protein-conducting channel formed by the SecY complex. How SecA moves substrates through the SecY channel is unclear. However, a recent structure of a SecA-SecY complex raises the possibility that the polypeptide chain is moved by a two-helix finger domain of SecA that is inserted into the cytoplasmic opening of the SecY channel. Here we have used disulphide-bridge crosslinking to show that the loop at the tip of the two-helix finger of Escherichia coli SecA interacts with a polypeptide chain right at the entrance into the SecY pore. Mutagenesis demonstrates that a tyrosine in the loop is particularly important for translocation, but can be replaced by some other bulky, hydrophobic residues. We propose that the two-helix finger of SecA moves a polypeptide chain into the SecY channel with the tyrosine providing the major contact with the substrate, a mechanism analogous to that suggested for hexameric, protein-translocating ATPases.


Asunto(s)
Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Escherichia coli/enzimología , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Adenosina Trifosfatasas/genética , Secuencias de Aminoácidos , Proteínas Bacterianas/genética , Reactivos de Enlaces Cruzados , Disulfuros/química , Disulfuros/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Transporte de Membrana/genética , Modelos Biológicos , Modelos Moleculares , Conformación Proteica , Transporte de Proteínas , Canales de Translocación SEC , Proteína SecA , Relación Estructura-Actividad , Tirosina/metabolismo
14.
Nat Commun ; 15(1): 1143, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326364

RESUMEN

The major ion chemistry of the ocean has been assumed to be controlled by river input, hydrothermal circulation at mid-ocean ridges, carbonate production, and low-temperature alteration of seafloor basalt, but marine chemical budgets remain difficult to balance. Here we propose that large-scale groundwater flow and diagenetic reactions in continental shelf sediments have been overlooked as an important contributor to major ion budgets in the ocean. Based on data synthesized from 17 passive margin basins, continental shelves contribute fluid exchanges comparable to hydrothermal circulation at mid-ocean ridges. Chemical exchange is similarly significant, indicating removal of Mg2+ from the oceans at rates similar to mid-ocean ridge convection. Continental shelves likely contribute Ca2+ and K+ to the oceans at rates that, in combination with low-temperature basalt alteration, can close current budget deficits. Flow and reaction in continental shelf sediments should be included in a new generation of studies addressing marine isotope budgets.

15.
Infect Control Hosp Epidemiol ; 45(1): 132-134, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37529841

RESUMEN

A wall-mounted, far-ultraviolet-C light technology reduced aerosolized bacteriophage MS2 by >3 log10 plaque-forming units within 30 minutes. Vegetative bacterial pathogens on steel disk carriers in the center of the room were reduced by >3 log10 after 45 minutes of exposure, but Candida auris and Clostridioides difficile spores were not.


Asunto(s)
Clostridioides difficile , Descontaminación , Humanos , Recuento de Colonia Microbiana , Tecnología , Esporas Bacterianas , Rayos Ultravioleta , Desinfección
16.
Aging Dis ; 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38421833

RESUMEN

Geographic atrophy (GA) is an advanced form of age-related macular degeneration (AMD), that starts with atrophic lesions in the outer retina that expand to cover the macula and fovea, leading to severe vision loss over time. Pigment Epithelium-Derived Factor (PEDF) has a diverse-range of properties, including its ability to promote cell survival, reduce inflammation, inhibit angiogenesis, combat oxidative stress, regulate autophagy, and stimulate anti-apoptotic pathways, making it a promising therapeutic candidate for GA. However, the relatively short half-life of PEDF protein has precluded its potential as a clinical therapy for GA since it would require frequent injections. Therefore, we describe administration of a PEDF gene, comparing and contrasting delivery routes, viral and non-viral vectors, and consider the critical challenges for PEDF as a neuroprotectant for GA.

17.
Cell Death Dis ; 15(6): 387, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824145

RESUMEN

Obesity exacerbates tissue degeneration and compromises the integrity and reparative potential of mesenchymal stem/stromal cells (MSCs), but the underlying mechanisms have not been sufficiently elucidated. Mitochondria modulate the viability, plasticity, proliferative capacity, and differentiation potential of MSCs. We hypothesized that alterations in the 5-hydroxymethylcytosine (5hmC) profile of mitochondria-related genes may mediate obesity-driven dysfunction of human adipose-derived MSCs. MSCs were harvested from abdominal subcutaneous fat of obese and age/sex-matched non-obese subjects (n = 5 each). The 5hmC profile and expression of nuclear-encoded mitochondrial genes were examined by hydroxymethylated DNA immunoprecipitation sequencing (h MeDIP-seq) and mRNA-seq, respectively. MSC mitochondrial structure (electron microscopy) and function, metabolomics, proliferation, and neurogenic differentiation were evaluated in vitro, before and after epigenetic modulation. hMeDIP-seq identified 99 peaks of hyper-hydroxymethylation and 150 peaks of hypo-hydroxymethylation in nuclear-encoded mitochondrial genes from Obese- versus Non-obese-MSCs. Integrated hMeDIP-seq/mRNA-seq analysis identified a select group of overlapping (altered levels of both 5hmC and mRNA) nuclear-encoded mitochondrial genes involved in ATP production, redox activity, cell proliferation, migration, fatty acid metabolism, and neuronal development. Furthermore, Obese-MSCs exhibited decreased mitochondrial matrix density, membrane potential, and levels of fatty acid metabolites, increased superoxide production, and impaired neuronal differentiation, which improved with epigenetic modulation. Obesity elicits epigenetic changes in mitochondria-related genes in human adipose-derived MSCs, accompanied by structural and functional changes in their mitochondria and impaired fatty acid metabolism and neurogenic differentiation capacity. These observations may assist in developing novel therapies to preserve the potential of MSCs for tissue repair and regeneration in obese individuals.


Asunto(s)
Tejido Adiposo , Diferenciación Celular , Epigénesis Genética , Células Madre Mesenquimatosas , Mitocondrias , Obesidad , Humanos , Células Madre Mesenquimatosas/metabolismo , Obesidad/metabolismo , Obesidad/genética , Obesidad/patología , Mitocondrias/metabolismo , Tejido Adiposo/metabolismo , Diferenciación Celular/genética , Femenino , Masculino , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Adulto , Persona de Mediana Edad , Proliferación Celular
18.
bioRxiv ; 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38293184

RESUMEN

Loss-of-function mutations in the genes encoding PINK1 and PRKN result in early-onset Parkinson disease (EOPD). Together the encoded enzymes direct a neuroprotective pathway that ensures the elimination of damaged mitochondria via autophagy. We performed a genome-wide high content imaging miRNA screen for inhibitors of the PINK1-PRKN pathway and identified all three members of the miRNA family 29 (miR-29). Using RNAseq we identified target genes and found that siRNA against ATG9A phenocopied the effects of miR-29 and inhibited the initiation of PINK1-PRKN mitophagy. Furthermore, we discovered two rare, potentially deleterious, missense variants (p.R631W and p.S828L) in our EOPD cohort and tested them experimentally in cells. While expression of wild-type ATG9A was able to rescue the effects of miR-29a, the EOPD-associated variants behaved like loss-of-function mutations. Together, our study validates miR-29 and its target gene ATG9A as novel regulators of mitophagy initiation. It further serves as proof-of-concept of finding novel, potentially disease-causing EOPD-linked variants specifically in mitophagy regulating genes. The nomination of genetic variants and biological pathways is important for the stratification and treatment of patients that suffer from devastating diseases, such as EOPD.

19.
Methods Mol Biol ; 2708: 155-174, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37558970

RESUMEN

The injection of therapies into the eye is common practice, both clinically and pre-clinically. The most straightforward delivery route is via an intravitreal injection, which introduces the treatment into the largest cavity at the posterior of the eye. This technique is frequently used to deliver gene therapies, including those containing recombinant adeno-associated viral vectors (AAVs), to the back of the eye to enable inner retinal targeting. This chapter provides detailed methodology on how to successfully perform an intravitreal injection in mice. The chapter covers vector preparation considerations, advice on how to minimize vector loss in the injection device, and ways to reduce vector reflux from the eye when administering a therapy. Finally, a protocol is provided on common retinal histology processing techniques to assess vector-mediated expression in retinal ganglion cells. It is hoped that this chapter will enable researchers to carry out effective and consistent intravitreal injections that transduce the inner retinal surface while avoiding common pitfalls.


Asunto(s)
Retina , Células Ganglionares de la Retina , Ratones , Animales , Inyecciones Intravítreas , Transducción Genética , Retina/metabolismo , Terapia Genética/métodos , Dependovirus/genética , Vectores Genéticos/genética
20.
Infect Control Hosp Epidemiol ; 44(8): 1361-1364, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36177872

RESUMEN

Contaminated shoes are a potential vector for dissemination of healthcare-associated pathogens. We demonstrated that healthcare personnel walking into patient rooms frequently transferred pathogens from their shoes to the floor. An 8-second treatment of shoes with a UV-C decontamination device significantly reduced the frequency of transfer of vegetative bacterial pathogens.


Asunto(s)
Habitaciones de Pacientes , Rayos Ultravioleta , Humanos , Recuento de Colonia Microbiana , Bacterias , Descontaminación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA