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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Exp Eye Res ; 226: 109351, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36539052

RESUMEN

α-Synuclein (α-Syn) is implicated in Parkinson's disease (PD), a neuromotor disorder with prominent visual symptoms. The underlying cause of motor dysfunction has been studied extensively, and is attributed to the death of dopaminergic neurons mediated in part by intracellular aggregation of α-Syn. The cause of visual symptoms, however, is less clear. Neuroretinal degeneration due to the presence of aggregated α-Syn has been reported, but the evidence is controversial. Other symptoms including those arising from primary open angle glaucoma (POAG) are believed to be the side-effects of medications prescribed for PD. Here, we explored the alternative hypothesis that dysfunction of α-Syn in the anterior eye alters the interaction between the actin cytoskeleton of trabecular meshwork (TM) cells with the extracellular matrix (ECM), impairing their ability to respond to physiological changes in intraocular pressure (IOP). A similar dysfunction in neurons is responsible for impaired neuritogenesis, a characteristic feature of PD. Using cadaveric human and bovine TM tissue and primary human TM cells as models, we report two main observations: 1) α-Syn is expressed in human and bovine TM cells, and significant amounts of monomeric and oligomeric α-Syn are present in the AH, and 2) primary human TM cells and human and bovine TM tissue endocytose extracellular recombinant monomeric and oligomeric α-Syn via the prion protein (PrPC), and upregulate fibronectin (FN) and α-smooth muscle actin (α-SMA), fibrogenic proteins implicated in POAG. Transforming growth factor ß2 (TGFß2), a fibrogenic cytokine implicated in ∼50% cases of POAG, is also increased, and so is RhoA-associated coiled-coil-containing protein kinase 1 (ROCK-1). However, silencing of α-Syn in primary human TM cells reduces FN, α-SMA, and ROCK-1 in the absence or presence of over-expressed active TGFß2, suggesting modulation of FN and ROCK-1 independent of, or upstream of TGFß2. These observations suggest that extracellular α-Syn modulates ECM proteins in the TM independently or via PrPC by activating the RhoA-ROCK pathway. These observations reveal a novel function of α-Syn in the anterior eye, and offer new therapeutic options.


Asunto(s)
Fibronectinas , Glaucoma de Ángulo Abierto , Animales , Bovinos , Humanos , alfa-Sinucleína/metabolismo , alfa-Sinucleína/farmacología , Células Cultivadas , Fibronectinas/metabolismo , Glaucoma de Ángulo Abierto/genética , Glaucoma de Ángulo Abierto/metabolismo , Presión Intraocular , Malla Trabecular/metabolismo , Factor de Crecimiento Transformador beta2/farmacología , Factor de Crecimiento Transformador beta2/metabolismo
2.
Curr Neurol Neurosci Rep ; 23(3): 33-48, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36869185

RESUMEN

PURPOSE OF REVIEW: There continue to be a plethora of approaches to the rehabilitation of hemispatial inattention, from different forms of sensory stimulation (visual, auditory and somatosensory feedback), through all major modes of non-invasive brain stimulation to drug therapies. Here we summarise trials published in the years 2017-2022 and tabulate their effect sizes, with the aim of drawing on common themes that may serve to inform future rehabilitative studies. RECENT FINDINGS: Immersive virtual reality approaches to visual stimulation seem well tolerated, although they have yet to yield any clinically relevant improvements. Dynamic auditory stimulation looks very promising and has high potential for implementation. Robotic interventions are limited by their cost and are perhaps best suited to patients with a co-occurring hemiparesis. Regarding brain stimulation, rTMS continues to demonstrate moderate effects but tDCS studies have yielded disappointing results so far. Drugs, primarily aimed at the dopaminergic system, often demonstrate beneficial effects of a medium size, but as with many of the approaches, it seems difficult to predict responders and non-responders. Our main recommendation is that researchers consider incorporating single-case experimental designs into their studies as rehabilitation trials are likely to remain small in terms of patient numbers, and this is the best way to deal with all the factors that cause large between-subject heterogeneity.


Asunto(s)
Trastornos Mentales , Terapia de Exposición Mediante Realidad Virtual , Humanos , Trastornos Mentales/rehabilitación , Estimulación Transcraneal de Corriente Directa/métodos , Estimulación Magnética Transcraneal/métodos
3.
Exp Eye Res ; 212: 108787, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34624335

RESUMEN

Recently, we reported ß-cleavage of the prion protein (PrPC) in human ocular tissues. Here, we explored whether this is unique to the human eye, and its functional implications. A comparison of the cleavage pattern of PrPC in human ocular tissues with common nocturnal and diurnal animals revealed mainly ß-cleavage in humans, and mostly full-length PrPC in animal retinas. Soluble FL PrPC and N-terminal fragment (N2) released from ß-cleavage was observed in the aqueous and vitreous humor (AH & VH). Expression of human PrPC in ARPE-19 cells, a human retinal pigmented epithelial cell line, also showed ß-cleaved PrPC. Surprisingly, ß-cleavage was not altered by a variety of insults, including oxidative stress, suggesting a unique role of this cleavage in the human eye. It is likely that ß-cleaved C- or N-terminal fragments of PrPC protect from various insults unique to the human eye. On the contrary, ß-cleaved C-terminus of PrPC is susceptible to conversion to the pathological PrP-scrapie form, and includes the binding sites for ß1-integrin and amyloid-ß, molecules implicated in several ocular disorders. Considering the species and tissue-specific cleavage of PrPC, our data suggest re-evaluation of prion infectivity and other ocular disorders of the human eye conducted in mouse models.


Asunto(s)
Oftalmopatías/metabolismo , Proteínas PrPC/metabolismo , División del ARN/fisiología , Epitelio Pigmentado de la Retina/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Epitelio Pigmentado de la Retina/patología
4.
Exp Eye Res ; 199: 108200, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32858007

RESUMEN

Prion diseases are invariably fatal neurodegenerative disorders that have gained much publicity due to their transmissible nature. Sporadic Creutzfeldt-Jakob disease (sCJD) is the most common human prion disorder, with an incidence of 1 in a million. Inherited prion disorders are relatively rare, and associated with mutations in the prion protein gene. More than 50 different point mutations, deletions, and insertions have been identified so far. Most are autosomal dominant and fully penetrant. Prion disorders also occur in animals, and are of major concern because of the potential for spreading to humans. The principal pathogenic event underlying all prion disorders is a change in the conformation of prion protein (PrPC) from a mainly α-helical to a ß-sheet rich isoform, PrP-scrapie (PrPSc). Accumulation of PrPSc in the brain parenchyma is the major cause of neuronal degeneration. The mechanism by which PrPSc is transmitted, propagates, and causes neurodegenerative changes has been investigated over the years, and several clues have emerged. Efforts are also ongoing for identifying specific and sensitive diagnostic tests for sCJD and animal prion disorders, but success has been limited. The eye is suitable for these evaluations because it shares several anatomical and physiological features with the brain, and can be observed in vivo during disease progression. The retina, considered an extension of the central nervous system, is involved extensively in prion disorders. Accordingly, Optical Coherence Tomography and electroretinogram have shown some promise as pre-mortem diagnostic tests for human and animal prion disorders. However, a complete understanding of the physiology of PrPC and pathobiology of PrPSc in the eye is essential for developing specific and sensitive tests. Below, we summarize recent progress in ocular physiology and pathology in prion disorders, and the eye as an anatomically accessible site to diagnose, monitor disease progression, and test therapeutic options.


Asunto(s)
Segmento Anterior del Ojo/metabolismo , Regulación de la Expresión Génica , Priones/genética , Animales , Segmento Anterior del Ojo/patología , Homeostasis , Humanos , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Priones/biosíntesis , Conformación Proteica
5.
Exp Eye Res ; 190: 107890, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31811823

RESUMEN

PURPOSE: The avascular cornea, trabecular meshwork (TM), and lens obtain iron, an essential biometal, from the aqueous humor (AH). The mechanism by which this exchange is regulated, however, is unclear. Recently we reported that non-pigmented ciliary epithelial cells express ferroportin (Fpn) (Ashok, 2018b), an iron export protein modulated by hepcidin, the master regulator of iron homeostasis secreted mainly by the liver. Here, we explored whether ciliary epithelial and other cells in the anterior segment synthesize hepcidin, suggesting local regulation of iron exchange at this site. METHODS: Human and bovine eyes were dissected to isolate the ciliary body (CB), corneal endothelial (CE), TM, lens epithelial (LE), and outer epithelial cell layer of the iris. Total mRNA and protein lysates were processed to evaluate the synthesis and expression of hepcidin, the iron regulatory peptide hormone, Fpn, the only known iron export protein, ceruloplasmin (Cp), a ferroxidase necessary for iron export, transferrin receptor (TfR), a major iron uptake protein, and ferritin, a major iron storage protein. A combination of techniques including reverse transcription polymerase chain reaction (RT-PCR) of total mRNA, Western blotting of protein lysates, and immunofluorescence of fixed tissue sections were used to accomplish these goals. RESULTS: RT-PCR of isolated tissue samples revealed hepcidin-specific mRNA in the CB, TM, CE, and LE of the bovine eye. Western blotting of protein lysates from these tissues showed reactivity for hepcidin, Fpn, ferritin, and TfR. Western blotting and immunohistochemistry of similar tissues isolated from cadaveric human eyes showed expression of hepcidin, Fpn, and Cp in these samples. Notably, Fpn and Cp were expressed on the basolateral membrane of non-pigmented ciliary epithelial cells, facing the AH. CONCLUSIONS: Synthesis and expression of hepcidin and Fpn in the ciliary epithelium suggests local regulation of iron transport from choroidal plexus in the ciliary body to the AH across the blood-aqueous barrier. Expression of hepcidin and Fpn in CE, TM, and LE cells indicates additional regulation of iron exchange between the AH and cornea, TM, and lens, suggesting autonomous regulation of iron homeostasis in the anterior segment. Physiological and pathological implications of these observations are discussed.


Asunto(s)
Segmento Anterior del Ojo/metabolismo , Antiinfecciosos/metabolismo , Hepcidinas/biosíntesis , Adulto , Anciano , Animales , Western Blotting , Proteínas de Transporte de Catión/metabolismo , Bovinos , Ceruloplasmina/metabolismo , Cuerpo Ciliar/metabolismo , Electroforesis en Gel de Poliacrilamida , Endotelio Corneal/metabolismo , Células Epiteliales/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Hepcidinas/genética , Humanos , Iris/metabolismo , Cristalino/metabolismo , Masculino , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Malla Trabecular/metabolismo
6.
Int J Mol Sci ; 21(19)2020 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-33023198

RESUMEN

Age-related macular degeneration (AMD) and glaucoma are degenerative conditions of the retina and a significant cause of irreversible blindness in developed countries. Alzheimer's disease (AD), the most common dementia of the elderly, is often associated with AMD and glaucoma. The cardinal features of AD include extracellular accumulation of amyloid ß (Aß) and intracellular deposits of hyper-phosphorylated tau (p-tau). Neuroinflammation and brain iron dyshomeostasis accompany Aß and p-tau deposits and, together, lead to progressive neuronal death and dementia. The accumulation of Aß and iron in drusen, the hallmark of AMD, and Aß and p-tau in retinal ganglion cells (RGC), the main retinal cell type implicated in glaucoma, and accompanying inflammation suggest overlapping pathology. Visual abnormalities are prominent in AD and are believed to develop before cognitive decline. Some are caused by degeneration of the visual cortex, while others are due to RGC loss or AMD-associated retinal degeneration. Here, we review recent information on Aß, p-tau, chronic inflammation, and iron dyshomeostasis as common pathogenic mechanisms linking the three degenerative conditions, and iron chelation as a common therapeutic option for these disorders. Additionally discussed is the role of prion protein, infamous for prion disorders, in Aß-mediated toxicity and, paradoxically, in neuroprotection.


Asunto(s)
Enfermedad de Alzheimer/genética , Encéfalo/metabolismo , Degeneración Macular/genética , Agregación Patológica de Proteínas/genética , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Encéfalo/patología , Glaucoma/complicaciones , Glaucoma/genética , Glaucoma/patología , Humanos , Degeneración Macular/complicaciones , Degeneración Macular/patología , Agregación Patológica de Proteínas/patología , Retina/metabolismo , Retina/patología , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Proteínas tau/genética , Proteínas tau/metabolismo
7.
Pract Neurol ; 20(6): 435-445, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32632038

RESUMEN

Vaccination is one of the most effective and cost-efficient methods for protecting people with multiple sclerosis (MS) from infections. However, use of vaccines has often been problematic because of misguided concerns that they may exacerbate the disease and/or that some disease-modifying therapies may influence the immune response to immunisations and/or their safety. People with MS risk higher morbidity and mortality from vaccine-preventable infections. It is, therefore, important to address any patient's reluctance to accept vaccination and to provide clear guidance for clinicians on which vaccinations to consider proactively. We have reviewed the current literature and provide recommendations regarding vaccines in adults with MS, including specific advice regarding vaccination safety in patients receiving-or going to receive-disease-modifying therapies, vaccination during pregnancy, pretravel counselling and patient education.


Asunto(s)
Esclerosis Múltiple , Vacunas , Femenino , Humanos , Esclerosis Múltiple/terapia , Embarazo , Vacunación
8.
Exp Eye Res ; 175: 1-13, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29859760

RESUMEN

Iron is an essential biometal in the aqueous humor, the principal source of nutrients for the avascular cornea and the lens. Here, we explored whether the ciliary body (CB), the source of aqueous humor, transports iron, and if the prion protein (PrPC) facilitates this process as in the outer retina. Using a combination of human, bovine, and mouse eyes as models, we report the expression of iron export proteins ferroportin and ceruloplasmin, and major iron uptake and storage proteins transferrin, transferrin receptor, and ferritin in the ciliary epithelium, indicating active exchange of iron at this site. Ferroportin and transferrin receptor are also expressed in the corneal endothelium. However, the relative expression of iron export and uptake proteins suggests export from the ciliary epithelium and import by corneal endothelium. In addition, abundant expression of PrPC, a ferrireductase that facilitates iron transport, is noted in pigmented and non-pigmented epithelium of the CB, posterior pigmented epithelium of the iris, corneal endothelium and epithelium, and lens epithelium. Notably, majority of PrPC in the ciliary epithelium is cleaved at the ß-site as in retinal pigment epithelial cells, suggesting a role in iron transport. Most of the PrPC in the cornea, however, is full-length, and susceptible to aggregation by intracerebrally inoculated PrP-scrapie, an infectious conformation of PrPC responsible for human and animal prion disorders. Soluble PrPC is present in the aqueous and vitreous humor, a provocative observation with significant implications. Together, these observations suggest independent cycling of iron in the anterior segment, and a prominent role of PrPC in this process. Aggregation of PrPC in the cornea of PrP-scrapie-infected animals raises the alarming possibility of transmission of animal prions through corneal abrasions.


Asunto(s)
Segmento Anterior del Ojo/metabolismo , Proteínas de Unión a Hierro/metabolismo , Proteínas PrPC/fisiología , Enfermedad Debilitante Crónica/metabolismo , Enfermedad Debilitante Crónica/transmisión , Animales , Transporte Biológico , Western Blotting , Proteínas de Transporte de Catión/metabolismo , Bovinos , Ceruloplasmina , Cuerpo Ciliar/metabolismo , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Células Epiteliales/metabolismo , Femenino , Ferritinas/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Homeostasis/fisiología , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Transferrina/metabolismo , Transferrina/metabolismo
9.
Indian J Crit Care Med ; 21(8): 483-487, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28904476

RESUMEN

AIM: This study aims to develop a standard operational protocol (SOP) for central line catheter care for nurses. MATERIALS AND METHODS: A preliminary draft of protocol based on extensive review of the literature was developed. The current practices of the nurses regarding central line catheter care were observed. Focus group discussions (FGD) were conducted with the nurses to identify the problems encountered by them during care of central line. Four rounds of Delphi were conducted to validate the protocol. The protocol was found to be feasible in terms of understanding, clarity and easy implementation after conducting a pilot study. An observation checklist was developed from the final draft of the protocol. The nurses were taught regarding the central line catheter care as per the protocol. 30 nurses were observed during central line catheter care by the researcher. After implementation of the protocol, feedback of the nurses was taken by conducting FGDs. RESULTS: Content validity index of each item in the protocol was acceptable. The overall Cronbach's alpha value of the checklist was 0.75. It was concluded that the checklist is reliable and each item has a contribution in the checklist. CONCLUSION: This protocol addresses interventions to enable staff to provide proper care of the central line catheter to prevent CLABSI.

10.
J Biol Chem ; 290(9): 5512-22, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25572394

RESUMEN

Brain iron-dyshomeostasis is an important cause of neurotoxicity in prion disorders, a group of neurodegenerative conditions associated with the conversion of prion protein (PrP(C)) from its normal conformation to an aggregated, PrP-scrapie (PrP(Sc)) isoform. Alteration of iron homeostasis is believed to result from impaired function of PrP(C) in neuronal iron uptake via its ferrireductase activity. However, unequivocal evidence supporting the ferrireductase activity of PrP(C) is lacking. Kidney provides a relevant model for this evaluation because PrP(C) is expressed in the kidney, and ∼370 µg of iron are reabsorbed daily from the glomerular filtrate by kidney proximal tubule cells (PT), requiring ferrireductase activity. Here, we report that PrP(C) promotes the uptake of transferrin (Tf) and non-Tf-bound iron (NTBI) by the kidney in vivo and mainly NTBI by PT cells in vitro. Thus, uptake of (59)Fe administered by gastric gavage, intravenously, or intraperitoneally was significantly lower in PrP-knock-out (PrP(-/-)) mouse kidney relative to PrP(+/+) controls. Selective in vivo radiolabeling of plasma NTBI with (59)Fe revealed similar results. Expression of exogenous PrP(C) in immortalized PT cells showed localization on the plasma membrane and intracellular vesicles and increased transepithelial transport of (59)Fe-NTBI and to a smaller extent (59)Fe-Tf from the apical to the basolateral domain. Notably, the ferrireductase-deficient mutant of PrP (PrP(Δ51-89)) lacked this activity. Furthermore, excess NTBI and hemin caused aggregation of PrP(C) to a detergent-insoluble form, limiting iron uptake. Together, these observations suggest that PrP(C) promotes retrieval of iron from the glomerular filtrate via its ferrireductase activity and modulates kidney iron metabolism.


Asunto(s)
FMN Reductasa/metabolismo , Hierro/metabolismo , Riñón/metabolismo , Proteínas PrPC/metabolismo , Animales , Western Blotting , Línea Celular Transformada , Membrana Celular/metabolismo , FMN Reductasa/genética , Femenino , Transporte Iónico/genética , Hierro/farmacocinética , Radioisótopos de Hierro , Riñón/citología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Masculino , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Proteínas PrPC/genética , Transferrina/metabolismo , Transferrina/farmacocinética
11.
Br J Ophthalmol ; 107(10): 1432-1437, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-35750478

RESUMEN

BACKGROUND/AIMS: Neurofilament light chain (NfL) levels in cerebrospinal fluid and serum are reliable indicators for neuroaxonal damage in a broad spectrum of neurodegenerative diseases. Herein, we investigate NfL levels in serum and anterior chamber fluid of patients with glaucoma. METHODS: Patients scheduled for routine glaucoma or cataract surgery were recruited for this study. Retinal nerve fibre layer thickness was measured by optical coherence tomography (OCT, Heidelberg Spectralis). NfL levels in serum and in anterior chamber fluid were analysed with Simoa SR-X Analyzer (Quanterix; NFLIGHT, Lexington, Massachusetts, USA). T-test was used for parametric data and Mann-Whitney-U test for nonparametric data. Spearman's rank-order correlation was used to investigate correlations. P values<0.05 were considered as statistically significant. RESULTS: Sixty patients with glaucoma and 58 controls were enrolled. Serum NfL concentration of patients with glaucoma was similar to serum NfL concentration in controls (median (IQR); 22.7 (18.9) pg/mL vs 22.5 (24.0) pg/mL; p=0.763). A positive correlation of serum NfL with age was observed in both patients with glaucoma (r=0.77; p<0.001) and in the control group (r=0.82, p<0.001). In the anterior chamber fluid, the NfL concentration was substantially increased in patients with glaucoma compared with controls (20.7 (101.3) pg/mL vs 3.1 (2.9) pg/mL; p<0.001). Furthermore, we found a positive correlation of anterior chamber fluid NfL with preoperative intraocular pressure (r=0.39, p=0.003) and with retinal nerve fibre layer thickness (r=0.58, p<0.001). CONCLUSION: NfL levels in anterior chamber fluid are elevated in patients with glaucoma and correlate with intraocular pressure and retinal nerve fibre layer thickness. The presented data strongly support anterior chamber fluid NfL as a new marker for glaucoma.


Asunto(s)
Cámara Anterior , Glaucoma , Filamentos Intermedios , Neuronas Retinianas , Humanos , Cámara Anterior/patología , Glaucoma/patología , Presión Intraocular , Neuronas Retinianas/patología
12.
Neurobiol Dis ; 45(3): 930-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22182691

RESUMEN

Prion disease associated neurotoxicity is mainly attributed to PrP-scrapie (PrP(Sc)), the disease associated isoform of a normal protein, the prion protein (PrP(C)). Participation of other proteins and processes is suspected, but their identity and contribution to the pathogenic process is unclear. Emerging evidence implicates imbalance of brain iron homeostasis as a significant cause of prion disease-associated neurotoxicity. The underlying cause of this change, however, remains unclear. We demonstrate that iron is sequestered in heat and SDS-stable protein complexes in sporadic-Creutzfeldt-Jakob-disease (sCJD) brains, creating a phenotype of iron deficiency. The underlying cause is change in the characteristics of ferritin, an iron storage protein that becomes aggregated, detergent-insoluble, and partitions with denatured ferritin using conventional methods of ferritin purification. A similar phenotype of iron deficiency is noted in the lumbar spinal cord (SC) tissue of scrapie infected hamsters, a site unlikely to be affected by massive neuronal death and non-specific iron deposition. As a result, the iron uptake protein transferrin (Tf) is upregulated in scrapie infected SC tissue, and increases with disease progression. A direct correlation between Tf and PrP(Sc) suggests sequestration of iron in dysfunctional ferritin that either co-aggregates with PrP(Sc) or is rendered dysfunctional by PrP(Sc) through an indirect process. Surprisingly, amplification of PrP(Sc)in vitro by the protein-misfolding-cyclic-amplification (PMCA) reaction using normal brain homogenate as substrate does not increase the heat and SDS-stable pool of iron even though both PrP(Sc) and ferritin aggregate by this procedure. These observations highlight important differences between PrP(Sc)-protein complexes generated in vivo during disease progression and in vitro by the PMCA reaction, and the significance of these complexes in PrP(Sc)-associated neurotoxicity.


Asunto(s)
Encéfalo/metabolismo , Síndrome de Creutzfeldt-Jakob/patología , Ferritinas/metabolismo , Hierro/metabolismo , Priones/metabolismo , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Animales , Cricetinae , Demencia/patología , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Calor , Humanos , Isótopos de Hierro/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Médula Espinal/metabolismo , Factores de Tiempo , Transferrina
13.
J Biol Chem ; 285(39): 30328-39, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20650901

RESUMEN

Filamin A (FLNA) is an integrator of cell mechanics and signaling. The spreading and migration observed in FLNA sufficient A7 melanoma cells but not in the parental FLNA deficient M2 cells have been attributed to FLNA. In A7 and M2 cells, the normal prion (PrP) exists as pro-PrP, retaining its glycosylphosphatidyl-inositol (GPI) anchor peptide signal sequence (GPI-PSS). The GPI-PSS of PrP has a FLNA binding motif and binds FLNA. Reducing PrP expression in A7 cells alters the spatial distribution of FLNA and organization of actin and diminishes cell spreading and migration. Integrin ß1 also binds FLNA. In A7 cells, FLNA, PrP, and integrin ß1 exist as two independent, yet functionally linked, complexes; they are FLNA with PrP or FLNA with integrin ß1. Reducing PrP expression in A7 cells decreases the amount of integrin ß1 bound to FLNA. A PrP GPI-PSS synthetic peptide that crosses the cell membrane inhibits A7 cell spreading and migration. Thus, in A7 cells FLNA does not act alone; the binding of pro-PrP enhances association between FLNA and integrin ß1, which then promotes cell spreading and migration. Pro-PrP is detected in melanoma in situ but not in melanocyte. Invasive melanoma has more pro-PrP. The binding of pro-PrP to FLNA, therefore, contributes to melanomagenesis.


Asunto(s)
Movimiento Celular , Proteínas Contráctiles/metabolismo , Regulación Neoplásica de la Expresión Génica , Cadenas beta de Integrinas/metabolismo , Melanoma/metabolismo , Proteínas de Microfilamentos/metabolismo , Priones/metabolismo , Secuencias de Aminoácidos , Línea Celular Tumoral , Proteínas Contráctiles/genética , Filaminas , Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Humanos , Cadenas beta de Integrinas/genética , Melanoma/genética , Proteínas de Microfilamentos/genética , Priones/genética , Unión Proteica
15.
PLoS Pathog ; 5(3): e1000336, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19283067

RESUMEN

Neurotoxicity in all prion disorders is believed to result from the accumulation of PrP-scrapie (PrP(Sc)), a beta-sheet rich isoform of a normal cell-surface glycoprotein, the prion protein (PrP(C)). Limited reports suggest imbalance of brain iron homeostasis as a significant associated cause of neurotoxicity in prion-infected cell and mouse models. However, systematic studies on the generality of this phenomenon and the underlying mechanism(s) leading to iron dyshomeostasis in diseased brains are lacking. In this report, we demonstrate that prion disease-affected human, hamster, and mouse brains show increased total and redox-active Fe (II) iron, and a paradoxical increase in major iron uptake proteins transferrin (Tf) and transferrin receptor (TfR) at the end stage of disease. Furthermore, examination of scrapie-inoculated hamster brains at different timepoints following infection shows increased levels of Tf with time, suggesting increasing iron deficiency with disease progression. Sporadic Creutzfeldt-Jakob disease (sCJD)-affected human brains show a similar increase in total iron and a direct correlation between PrP and Tf levels, implicating PrP(Sc) as the underlying cause of iron deficiency. Increased binding of Tf to the cerebellar Purkinje cell neurons of sCJD brains further indicates upregulation of TfR and a phenotype of neuronal iron deficiency in diseased brains despite increased iron levels. The likely cause of this phenotype is sequestration of iron in brain ferritin that becomes detergent-insoluble in PrP(Sc)-infected cell lines and sCJD brain homogenates. These results suggest that sequestration of iron in PrP(Sc)-ferritin complexes induces a state of iron bio-insufficiency in prion disease-affected brains, resulting in increased uptake and a state of iron dyshomeostasis. An additional unexpected observation is the resistance of Tf to digestion by proteinase-K, providing a reliable marker for iron levels in postmortem human brains. These data implicate redox-iron in prion disease-associated neurotoxicity, a novel observation with significant implications for prion disease pathogenesis.


Asunto(s)
Encéfalo/metabolismo , Homeostasis/fisiología , Hierro/metabolismo , Enfermedades por Prión/metabolismo , Animales , Western Blotting , Encéfalo/patología , Cricetinae , Humanos , Inmunohistoquímica , Ratones , Microscopía Fluorescente , Neuronas/metabolismo , Neuronas/patología , Enfermedades por Prión/patología , Receptores de Transferrina/metabolismo , Transferrina/metabolismo
16.
Prion ; 15(1): 126-137, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34224321

RESUMEN

Accumulation of redox-active iron in human sporadic Creutzfeldt-Jakob disease (sCJD) brain tissue and scrapie-infected mouse brains has been demonstrated previously. Here, we explored whether upregulation of local hepcidin secreted within the brain is the underlying cause of iron accumulation and associated toxicity. Using scrapie-infected mouse brains, we demonstrate transcriptional upregulation of hepcidin relative to controls. As a result, ferroportin (Fpn), the downstream effector of hepcidin and the only known iron export protein was downregulated, and ferritin, an iron storage protein was upregulated, suggesting increased intracellular iron. A similar transcriptional and translational upregulation of hepcidin, and decreased expression of Fpn and an increase in ferritin expression was observed in sCJD brain tissue. Further evaluation in human neuroblastoma cells (M17) exposed to synthetic mini-hepcidin showed downregulation of Fpn, upregulation of ferritin, and an increase in reactive oxygen species (ROS), resulting in cytotoxicity in a dose-dependent manner. Similar effects were noted in primary neurons isolated from mouse brain. As in M17 cells, primary neurons accumulated ferritin and ROS, and showed toxicity at five times lower concentration of mini-hepcidin. These observations suggest that upregulation of brain hepcidin plays a significant role in iron accumulation and associated neurotoxicity in human and animal prion disorders.


Asunto(s)
Hepcidinas , Enfermedades por Prión , Animales , Encéfalo/metabolismo , Ferritinas/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Ratones , Enfermedades por Prión/genética , Regulación hacia Arriba
17.
Antioxidants (Basel) ; 10(8)2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34439501

RESUMEN

To evaluate the role of iron in sodium iodate (NaIO3)-induced model of age-related macular degeneration (AMD) in ARPE-19 cells in-vitro and in mouse models in-vivo. ARPE-19 cells, a human retinal pigment epithelial cell line, was exposed to 10 mM NaIO3 for 24 h, and the expression and localization of major iron modulating proteins was evaluated by Western blotting (WB) and immunostaining. Synthesis and maturation of cathepsin-D (cat-D), a lysosomal enzyme, was evaluated by quantitative reverse-transcriptase polymerase chain reaction (RT-qPCR) and WB, respectively. For in-vivo studies, C57BL/6 mice were injected with 40 mg/kg mouse body weight of NaIO3 intraperitoneally, and their retina was evaluated after 3 weeks as above. NaIO3 induced a 10-fold increase in ferritin in ARPE-19 cells, which co-localized with LC3II, an autophagosomal marker, and LAMP-1, a lysosomal marker. A similar increase in ferritin was noted in retinal lysates and retinal sections of NaIO3-injected mice by WB and immunostaining. Impaired synthesis and maturation of cat-D was also noted. Accumulated ferritin was loaded with iron, and released from retinal pigmented epithelial (RPE) cells in Perls' and LAMP-1 positive vesicles. NaIO3 impairs lysosomal degradation of ferritin by decreasing the transcription and maturation of cat-D in RPE cells. Iron-loaded ferritin accumulates in lysosomes and is released in lysosomal membrane-enclosed vesicles to the extracellular milieu. Accumulation of ferritin in RPE cells and fusion of ferritin-containing vesicles with adjacent photoreceptor cells is likely to create an iron overload, compromising their viability. Moreover, reduced activity of cat-D is likely to promote accumulation of other cellular debris in lysosomal vesicles, contributing to AMD-like pathology.

18.
J Alzheimers Dis ; 82(4): 1487-1497, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34180415

RESUMEN

BACKGROUND: Accumulation of iron is a consistent feature of Alzheimer's disease (AD) brains. The underlying cause, however, remains debatable. OBJECTIVE: To explore whether local hepcidin synthesized by brain cells contributes to iron accumulation in AD brains. METHODS: Brain tissue from the cingulate cortex of 33 cases of AD pre-assigned to Braak stage I-VI, 6 cases of non-dementia, and 15 cases of non-AD dementia were analyzed for transcriptional upregulation of hepcidin by RT-qPCR and RT-PCR. Change in the expression of ferritin, ferroportin (Fpn), microglial activation marker Iba1, IL-6, and TGFß2 was determined by western blotting. Total tissue iron was determined by colorimetry. RESULTS: Significant transcriptional upregulation of hepcidin was observed in Braak stage III-VI relative to Braak stage I and II, non-AD dementia, and non-dementia samples. Ferritin was increased in Braak stage V, and a significant increase in tissue iron was evident in Braak stage III-VI. The expression of Iba1 and IL-6 was also increased in Braak stage III-VI relative to Braak stage I and II and non-AD dementia samples. Amyloid-ß plaques were absent in most Braak stage I and II samples, and present in Braak stage III-VI samples with few exceptions. CONCLUSION: These observations suggest that upregulation of brain hepcidin is mediated by IL-6, a known transcriptional activator of hepcidin. The consequent downregulation of Fpn on neuronal and other cells results in accumulation of iron in AD brains. The increase in hepcidin is disease-specific, and increases with disease progression, implicating AD-specific pathology in the accumulation of iron.


Asunto(s)
Enfermedad de Alzheimer/patología , Antiinfecciosos/metabolismo , Ferritinas/metabolismo , Hepcidinas/metabolismo , Regulación hacia Arriba , Anciano , Autopsia , Encéfalo/patología , Femenino , Humanos , Interleucina-6/metabolismo , Masculino , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Cureus ; 13(8): e17369, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34567908

RESUMEN

Background Various Indian registries have documented a delay of more than five hours for acute coronary syndrome patients from onset of symptoms to reaching thrombolysis-enabled centres. We conducted this study to evaluate the factors responsible for pre-hospital delay in acute coronary syndrome patients. Methods This was a descriptive cross-sectional study conducted in consecutive acute coronary syndrome patients who reported to the tertiary care medical centre in North India. A standardized tool was used to record the demographic data, socioeconomic status and clinical presentation of patients. All factors which led to pre-hospital delay were noted and the appropriate statistical tests were used for analysis. Results A total of 130 patients (males=93, females=37) were included in the study. The median time at which the acute coronary syndrome patients presented to the thrombolysis and percutaneous coronary intervention enabled centre was 490 minutes (range: 20 - 810 minutes) and 710 minutes (range: 45 - 940 minutes) respectively. The various factors responsible for prehospital delay were rural residence (p-value <0.0001), visit to local dispensary (p-value=0.0023), delay in getting transport (p-value=0.03) and misinterpretation of cardiac symptoms (p-value=0.0004). A significant but weak negative correlation was found between per capita income, decision making time and time taken to receive thrombolytic therapy. Out of a total of 83 ST-elevation myocardial infarction patients, only 46 (51.80%) were thrombolysed. Though 69/83 (83.13%) ST-elevation myocardial infarction patients reached thrombolysis enabled centre directly, only nine (10.84%) were thrombolysed at first medical contact; the rest were transferred to the percutaneous coronary intervention-enabled centre without any prior information.  Conclusion Our study concludes that besides socioeconomic and demographic variables, lack of public awareness, well established public transport & health insurance system lead to significant pre-hospital delays and increase the time to revascularization. Besides, judgemental error on the part of medical practitioners in the peripheries also significantly delays thrombolysis in ST-elevation myocardial infarction patients.

20.
Acta Neuropathol Commun ; 9(1): 62, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33827706

RESUMEN

Definitive diagnosis of Parkinson's disease (PD) and dementia with Lewy bodies (DLB) relies on postmortem finding of disease-associated alpha-synuclein (αSynD) as misfolded protein aggregates in the central nervous system (CNS). The recent development of the real-time quaking induced conversion (RT-QuIC) assay for ultrasensitive detection of αSynD aggregates has revitalized the diagnostic values of clinically accessible biospecimens, including cerebrospinal fluid (CSF) and peripheral tissues. However, the current αSyn RT-QuIC assay platforms vary widely and are thus challenging to implement and standardize the measurements of αSynD across a wide range of biospecimens and in different laboratories. We have streamlined αSyn RT-QuIC assay based on a second generation assay platform that was assembled entirely with commercial reagents. The streamlined RT-QuIC method consisted of a simplified protocol requiring minimal hands-on time, and allowing for a uniform analysis of αSynD in different types of biospecimens from PD and DLB. Ultrasensitive and specific RT-QuIC detection of αSynD aggregates was achieved in million-fold diluted brain homogenates and in nanoliters of CSF from PD and DLB cases but not from controls. Comparative analysis revealed higher seeding activity of αSynD in DLB than PD in both brain homogenates and CSF. Our assay was further validated with CSF samples of 214 neuropathologically confirmed cases from tissue repositories (88 PD, 58 DLB, and 68 controls), yielding a sensitivity of 98% and a specificity of 100%. Finally, a single RT-QuIC assay protocol was employed uniformly to detect seeding activity of αSynD in PD samples across different types of tissues including the brain, skin, salivary gland, and colon. We anticipate that our streamlined protocol will enable interested laboratories to easily and rapidly implement the αSyn RT-QuIC assay for various clinical specimens from PD and DLB. The utilization of commercial products for all assay components will improve the robustness and standardization of the RT-QuIC assay for diagnostic applications across different sites. Due to ultralow sample consumption, the ultrasensitive RT-QuIC assay will facilitate efficient use and sharing of scarce resources of biospecimens. Our streamlined RT-QuIC assay is suitable to track the distribution of αSynD in CNS and peripheral tissues of affected patients. The ongoing evaluation of RT-QuIC assay of αSynD as a potential biomarker for PD and DLB in clinically accessible biospecimens has broad implications for understanding disease pathogenesis, improving early and differential diagnosis, and monitoring therapeutic efficacies in clinical trials.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Enfermedad por Cuerpos de Lewy/diagnóstico , Enfermedad de Parkinson/diagnóstico , alfa-Sinucleína/análisis , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA