Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Neuroimmune Pharmacol ; 18(3): 495-508, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37661197

RESUMEN

NeuroHIV and other neurologic disorders present with altered iron metabolism in central nervous system neurons. Many people with HIV also use opioids, which can worsen neuroHIV symptoms by further dysregulating neuronal iron metabolism. Our previous work demonstrated that the µ-opioid agonist morphine causes neuronal endolysosomes to release their iron stores, and neurons respond by upregulating ferritin heavy chain (FHC), an iron storage protein associated with cognitive impairment in neuroHIV. Here, we investigated if this process required divalent metal transporter 1 (DMT1), a well-known iron transporter expressed on endolysosomes. We first optimized conditions to detect DMT1 isoforms (DMT1 1B ± iron responsive element) using fluorescently labeled rat DMT1 constructs expressed in HEK-293 cells. We also expressed these constructs in primary rat cortical neurons to compare their expression and subcellular distribution with endogenous DMT1 isoforms. We found endogenous DMT1 isoforms in the cytoplasm that colocalized with lysosomal-associated protein 1 (LAMP1), a marker of endolysosomes. Next, we blocked endogenous DMT1 isoforms using ebselen, a potent pharmacological inhibitor of DMT1 iron transport. Ebselen pre-treatment blocked morphine's ability to upregulate FHC protein, suggesting this pathway requires DMT1 iron transport from endolysosomes. This was further validated using viral-mediated genetic silencing of DMT1±IRE in cortical neurons, which also blocked FHC upregulation in the presence of morphine. Overall, our work demonstrates that the µ-opioid agonist morphine utilizes the endolysosomal iron transporter DMT1 to modulate neuronal cellular iron metabolism, upregulate FHC protein, and contribute to cognitive decline in neuroHIV. Morphine requires DMT1 to upregulate neuronal FHC. Cortical neurons treated with morphine release their endolysosomal iron stores to the cytoplasm and upregulate FHC, an iron storage protein associated with dendritic spine deficits and cognitive impairment in neuroHIV. This pathway requires the endolysosomal iron transporter DMT1, as pharmacological and genetic inhibitors of the transporter completely block morphine's ability to upregulate FHC. Created with BioRender.com .


Asunto(s)
Apoferritinas , Morfina , Animales , Humanos , Ratas , Analgésicos Opioides/farmacología , Analgésicos Opioides/metabolismo , Apoferritinas/metabolismo , Células HEK293 , Hierro/metabolismo , Lisosomas , Morfina/farmacología , Neuronas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
2.
Mol Neurobiol ; 60(4): 1811-1825, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36576708

RESUMEN

Alteration of neuronal protein processing is often associated with neurological disorders and is highly dependent on cellular protein trafficking. A prime example is the amyloidogenic processing of amyloid precursor protein (APP) in intracellular vesicles, which plays a key role in age-related cognitive impairment. Most approaches to correct this altered processing aim to limit enzymatic activities that lead to toxic products, such as protein cleavage by ß-secretase and the resulting amyloid ß production. A viable alternative is to direct APP to cellular compartments where non-amyloidogenic mechanisms are favored. To this end, we exploited the molecular properties of the herpes simplex virus 1 (HSV-1) transport protein US9 to guide APP interaction with preferred endogenous targets. Specifically, we generated a US9 chimeric construct that facilitates APP processing through the non-amyloidogenic pathway and tested it in primary cortical neurons. In addition to reducing amyloid ß production, our approach controls other APP-dependent biochemical steps that lead to neuronal deficits, including phosphorylation of APP and tau proteins. Notably, it also promotes the release of neuroprotective soluble αAPP. In contrast to other neuroprotective strategies, these US9-driven effects rely on the activity of endogenous neuronal proteins, which lends itself well to the study of fundamental mechanisms of APP processing/trafficking. Overall, this work introduces a new method to limit APP misprocessing and its cellular consequences without directly targeting secretase activity, offering a novel tool to reduce cognitive decline in pathologies such as Alzheimer's disease and HIV-associated neurocognitive disorders.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Humanos , Precursor de Proteína beta-Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/metabolismo , Neuronas/metabolismo , Transporte de Proteínas
3.
Br J Pharmacol ; 179(8): 1753-1768, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34825370

RESUMEN

BACKGROUND AND PURPOSE: Airway remodelling is a critical feature of chronic lung diseases. Epithelial-mesenchymal transition (EMT) represents an important source of myofibroblasts, contributing to airway remodelling. Here, we investigated the sphingosine-1-phosphate (S1P) role in EMT and its involvement in asthma-related airway dysfunction. EXPERIMENTAL APPROACH: A549 cells were used to assess the S1P effect on EMT and its interaction with TGF-ß signalling. To assess the S1P role in vivo and its impact on lung function, two experimental models of asthma were used by exposing BALB/c mice to subcutaneous administration of either S1P or ovalbumin (OVA). KEY RESULTS: Following incubation with TGF-ß or S1P, A549 acquire a fibroblast-like morphology associated with an increase of mesenchymal markers and down-regulation of the epithelial. These effects are reversed by treatment with the TGF-ß receptor antagonist LY2109761. Systemic administration of S1P to BALB/c mice induces asthma-like disease characterized by mucous cell metaplasia and increased levels of TGF-ß, IL-33 and FGF-2 within the lung. The bronchi harvested from S1P-treated mice display bronchial hyperresponsiveness associated with overexpression of the mesenchymal and fibrosis markers and reduction of the epithelial.The S1P-induced switch from the epithelial toward the mesenchymal pattern correlates to a significant increase of lung resistance and fibroblast activation. TGF-ß blockade, in S1P-treated mice, abrogates these effects. Finally, inhibition of sphingosine kinases by SK1-II in OVA-sensitized mice, abrogates EMT, pulmonary TGF-ß up-regulation, fibroblasts recruitment and airway hyperresponsiveness. CONCLUSION AND IMPLICATIONS: Targeting S1P/TGF-ß axis may hold promise as a feasible therapeutic target to control airway dysfunction in asthma.


Asunto(s)
Asma , Transición Epitelial-Mesenquimal , Esfingosina , Factor de Crecimiento Transformador beta , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Asma/metabolismo , Asma/patología , Células Epiteliales , Lisofosfolípidos , Ratones , Ratones Endogámicos BALB C , Ovalbúmina , Esfingosina/análogos & derivados , Esfingosina/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1
4.
Cell Mol Life Sci ; 78(9): 4283-4303, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33585975

RESUMEN

HIV-associated neurocognitive disorder (HAND) is characterized by cognitive and behavioral deficits in people living with HIV. HAND is still common in patients that take antiretroviral therapies, although they tend to present with less severe symptoms. The continued prevalence of HAND in treated patients is a major therapeutic challenge, as even minor cognitive impairment decreases patient's quality of life. Therefore, modern HAND research aims to broaden our understanding of the mechanisms that drive cognitive impairment in people with HIV and identify promising molecular pathways and targets that could be exploited therapeutically. Recent studies suggest that HAND in treated patients is at least partially induced by subtle synaptodendritic damage and disruption of neuronal networks in brain areas that mediate learning, memory, and executive functions. Although the causes of subtle neuronal dysfunction are varied, reversing synaptodendritic damage in animal models restores cognitive function and thus highlights a promising therapeutic approach. In this review, we examine evidence of synaptodendritic damage and disrupted neuronal connectivity in HAND from clinical neuroimaging and neuropathology studies and discuss studies in HAND models that define structural and functional impairment of neurotransmission. Then, we report molecular pathways, mechanisms, and comorbidities involved in this neuronal dysfunction, discuss new approaches to reverse neuronal damage, and highlight current gaps in knowledge. Continued research on the manifestation and mechanisms of synaptic injury and network dysfunction in HAND patients and experimental models will be critical if we are to develop safe and effective therapies that reverse subtle neuropathology and cognitive impairment.


Asunto(s)
Infecciones por VIH/patología , Trastornos Neurocognitivos/patología , Neuronas/metabolismo , Citoesqueleto de Actina , Animales , Astrocitos/metabolismo , Espinas Dendríticas/metabolismo , Infecciones por VIH/complicaciones , Humanos , Trastornos Neurocognitivos/etiología , Neuronas/patología , Receptores Ionotrópicos de Glutamato/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/patología
5.
Methods Mol Biol ; 2201: 139-162, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32975796

RESUMEN

Opioid use has substantially increased over recent years and remains a major driver of new HIV infections worldwide. Clinical studies indicate that opioids may exacerbate the symptoms of HIV-associated neurocognitive disorders (HAND), but the mechanisms underlying opioid-induced cognitive decline remain obscure. We recently reported that the µ-opioid agonist morphine increased neuronal iron levels and levels of ferritin proteins that store iron, suggesting that opioids modulate neuronal iron homeostasis. Additionally, increased iron and ferritin heavy chain protein were necessary for morphine's ability to reduce the density of thin and mushroom dendritic spines in cortical neurons, which are considered critical mediators of learning and memory, respectively. As altered iron homeostasis has been reported in HAND and related neurocognitive disorders like Alzheimer's, Parkinson's, and Huntington's disease, understanding how opioids regulate neuronal iron metabolism may help identify novel drug targets in HAND with potential relevance to these other neurocognitive disorders. Here, we review the known mechanisms of opioid-mediated regulation of neuronal iron and corresponding cellular responses and discuss the implications of these findings for patients with HAND. Furthermore, we discuss a new molecular approach that can be used to understand if opioid modulation of iron affects the expression and processing of amyloid precursor protein and the contributions of this pathway to HAND.


Asunto(s)
Disfunción Cognitiva/metabolismo , Hierro/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Animales , Disfunción Cognitiva/fisiopatología , Espinas Dendríticas/metabolismo , Ferritinas/metabolismo , Infecciones por VIH/complicaciones , Humanos , Morfina/farmacología , Trastornos Neurocognitivos/metabolismo , Neuronas/metabolismo , Receptores Opioides/metabolismo
6.
Elife ; 92020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31971513

RESUMEN

Synaptodendritic pruning is a common cause of cognitive decline in neurological disorders, including HIV-associated neurocognitive disorders (HAND). HAND persists in treated patients as a result of chronic inflammation and low-level expression of viral proteins, though the mechanisms involved in synaptic damage are unclear. Here, we report that the chemokine CXCL12 recoups both cognitive performance and synaptodendritic health in a rodent model of HAND, which recapitulates the neuroinflammatory state of virally controlled individuals and the associated structural/functional deficiencies. CXCL12 preferentially regulates plastic thin spines on layer II/III pyramidal neurons of the medial prefrontal cortex via CXCR4-dependent stimulation of the Rac1/PAK actin polymerization pathway, leading to increased spine density and improved flexible behavior. Our studies unveil a critical role of CXCL12/CXCR4 signaling in spine dynamics and cognitive flexibility, suggesting that HAND - or other diseases driven by spine loss - may be reversible and upturned by targeting Rac1-dependent processes in cortical neurons.


Asunto(s)
Quimiocina CXCL12/metabolismo , Cognición/fisiología , Espinas Dendríticas/metabolismo , Corteza Prefrontal/fisiología , Complejo SIDA Demencia , Animales , Células Cultivadas , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Masculino , Corteza Prefrontal/citología , Células Piramidales/citología , Células Piramidales/metabolismo , Ratas , Ratas Transgénicas , Receptores CXCR4/metabolismo , Proteína de Unión al GTP rac1/metabolismo
7.
eNeuro ; 6(4)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31300544

RESUMEN

HIV-associated neurocognitive disorders (HAND) remain prevalent and are aggravated by µ-opioid use. We have previously shown that morphine and other µ-opioids may contribute to HAND by inhibiting the homeostatic and neuroprotective chemokine receptor CXCR4 in cortical neurons, and this novel mechanism depends on upregulation of the protein ferritin heavy chain (FHC). Here, we examined the cellular events and potential mechanisms involved in morphine-mediated FHC upregulation using rat cortical neurons of either sex in vitro and in vivo. Morphine dose dependently increased FHC protein levels in primary neurons through µ-opioid receptor (µOR) and Gαi-protein signaling. Cytoplasmic FHC levels were significantly elevated, but nuclear FHC levels and FHC gene expression were unchanged. Morphine-treated rats also displayed increased FHC levels in layer 2/3 neurons of the prefrontal cortex. Importantly, both in vitro and in vivo FHC upregulation was accompanied by loss of mature dendritic spines, which was also dependent on µOR and Gαi-protein signaling. Moreover, morphine upregulated ferritin light chain (FLC), a component of the ferritin iron storage complex, suggesting that morphine altered neuronal iron metabolism. Indeed, prior to FHC upregulation, morphine increased cytoplasmic labile iron levels as a function of decreased endolysosomal iron. In line with this, chelation of endolysosomal iron (but not extracellular iron) blocked morphine-induced FHC upregulation and dendritic spine reduction, whereas iron overloading mimicked the effect of morphine on FHC and dendritic spines. Overall, these data demonstrate that iron mediates morphine-induced FHC upregulation and consequent dendritic spine deficits and implicate endolysosomal iron efflux to the cytoplasm in these effects.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Apoferritinas/metabolismo , Corteza Cerebral/efectos de los fármacos , Endosomas/metabolismo , Hierro/metabolismo , Lisosomas/metabolismo , Morfina/administración & dosificación , Neuronas/efectos de los fármacos , Animales , Corteza Cerebral/metabolismo , Espinas Dendríticas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Neuronas/citología , Neuronas/metabolismo , Cultivo Primario de Células , Ratas Sprague-Dawley , Receptores Opioides mu/metabolismo , Regulación hacia Arriba
8.
Methods Mol Biol ; 1882: 183-195, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30378055

RESUMEN

Stress granules are nonmembranous organelles that function as a stress-adaptation mechanism. We have recently shown that stress granules are mobilized by mutant KRAS pancreatic cancer cells under stress to enhance tumor fitness and survival. In this chapter, we outline a method for inducing, detecting, and quantifying stress granules in pancreatic cancer cells in vitro and in vivo. This method can be utilized to better understand the mechanisms driving stress granule formation and their role in pancreatic tumorigenesis.


Asunto(s)
Carcinogénesis/patología , Técnicas de Cultivo de Célula/métodos , Gránulos Citoplasmáticos/patología , Neoplasias Pancreáticas/patología , Estrés Fisiológico , Animales , Técnicas de Cultivo de Célula/instrumentación , Línea Celular Tumoral , Gránulos Citoplasmáticos/genética , Humanos , Procesamiento de Imagen Asistido por Computador/instrumentación , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Microscopía Fluorescente/instrumentación , Microscopía Fluorescente/métodos , Páncreas/patología , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética
10.
Mar Drugs ; 11(11): 4451-63, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24217287

RESUMEN

An in-depth study of the secondary metabolites contained in the Caribbean sponge Smenospongia aurea led to the isolation of smenamide A (1) and B (2), hybrid peptide/polyketide compounds containing a dolapyrrolidinone unit. Their structures were elucidated using high-resolution ESI-MS/MS and homo- and heteronuclear 2D NMR experiments. Structures of smenamides suggested that they are products of the cyanobacterial metabolism, and 16S rRNA metagenomic analysis detected Synechococcus spongiarum as the only cyanobacterium present in S. aurea. Smenamides showed potent cytotoxic activity at nanomolar levels on lung cancer Calu-1 cells, which for compound 1 is exerted through a clear pro-apoptotic mechanism. This makes smenamides promising leads for antitumor drug design.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Péptidos/química , Péptidos/farmacología , Policétidos/química , Policétidos/farmacología , Poríferos/química , Animales , Secuencia de Bases , Región del Caribe , Línea Celular Tumoral , Cianobacterias/genética , Cianobacterias/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Halogenación , Humanos , Datos de Secuencia Molecular , Poríferos/microbiología , ARN Ribosómico 16S/genética
11.
Am J Transl Res ; 5(6): 563-81, 2013 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-24093054

RESUMEN

CD133 (promini-1) is a member of the transmembrane glycoprotein family, was initially described as a specific marker to select human hematopoietic progenitor cells. Then, it was recognised as important marker to identify and isolate the specific cell subpopulation termed "cancer stem cells". Many studies showed that CD133(+) cells have stemness properties such as self-renewal, differentiation ability, high proliferation and they are able also to form tumours in xenografts. Moreover it has been demonstrated that CD133(+) cells more resistant to radiation and standard chemotherapy than CD133(-) cells. Although this, others investigations demonstrated that also CD133(-) cells can show the same characteristics of those positive for CD133(+). Hence, some inconsistencies among published data on CD133 function can be ascribed to different causes questioning the main role as specific marker of cancer stem cells. In fact, many authors indicate that CD133 is expressed both in differentiated and undifferentiated cells, and CD133(-) cancer cells can also initiate tumours. Indeed, it is still a matter of debate whether CD133(+) cells truly represent the ultimate tumourigenic population. However, the belief that CD133 may act as a universal marker of CSCs has been met with a high degree of controversy in the research community. In this review there is an attempt to highlight: i) the role and function of CD133, with an overview on the current stage of knowledge about this molecule, ii) the difficulty often encountered in its identification iii) the utility of CD133 expression as a prognostic marker.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA