Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 78(9): 4283-4303, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33585975

RESUMO

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.


Assuntos
Infecções por HIV/patologia , Transtornos Neurocognitivos/patologia , Neurônios/metabolismo , Citoesqueleto de Actina , Animais , Astrócitos/metabolismo , Espinhas Dendríticas/metabolismo , Infecções por HIV/complicações , Humanos , Transtornos Neurocognitivos/etiologia , Neurônios/patologia , Receptores Ionotrópicos de Glutamato/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/patologia
2.
J Neuroimmune Pharmacol ; 18(3): 495-508, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37661197

RESUMO

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 .


Assuntos
Apoferritinas , Morfina , Animais , Humanos , Ratos , Analgésicos Opioides/farmacologia , Analgésicos Opioides/metabolismo , Apoferritinas/metabolismo , Células HEK293 , Ferro/metabolismo , Lisossomos , Morfina/farmacologia , Neurônios/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
3.
ACS Omega ; 5(31): 19469-19477, 2020 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-32803041

RESUMO

Fluorescence sensing of oxalate has garnered some attention in the past two decades as a result of this anion's prominence and impact on society. Previous work on oxalate sensors and other divalent anion sensors has led to the conclusion that the sensors are selective for the anion under investigation. However, sensor selectivity is often determined by testing against a relatively small array of "guest" molecules or analytes and studies often exclude potentially interfering compounds. For example, studies on oxalate sensors have excluded compounds such as citrate and urate, which are anions in the biological matrices where oxalate is measured (e.g., urine, blood, and bacterial lysate). In the present study, we reassessed the selectivity of a dinuclear copper(II) macrocycle (Cu2L) in an eosin Y displacement assay using biologically relevant anions. Although previously reported as selective for oxalate, we found greater indicator displacement (fluorescence response) for urate and oxaloacetate and a significant response to citrate. These anions are larger than oxalate and do not appear to fit into the putative binding pocket of Cu2L. Consistent with previous reports, Cu2L did not release eosin Y in the presence of several other dicarboxylates, including adipate, glutarate, malate (except at 10 mM), fumarate, succinate, or malonate (except at 10 mM), and the monocarboxylate acetate. This was demonstrated by the failure of the anions to reverse eosin Y quenching by Cu2L. We also assessed, for the first time, other monocarboxylates, including butyrate, pyruvate, lactate, propionate, and formate. None of these anions were able to displace eosin Y, indicating no interaction with Cu2L that interfered with the eosin Y binding site. Single-crystal X-ray crystallography revealed that nonselective binding of the anions is likely partly caused by readily accessible copper(II) ions on the external surface of Cu2L. In addition, π-π stacking of urate with the aromatic groups of Cu2L cannot be ruled out as a contributor to binding. We conclude that Cu2L is not suitable for oxalate sensing in a biological matrix unless interfering compounds are selectively removed or masked.

4.
eNeuro ; 6(4)2019.
Artigo em Inglês | MEDLINE | ID: mdl-31300544

RESUMO

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.


Assuntos
Analgésicos Opioides/administração & dosagem , Apoferritinas/metabolismo , Córtex Cerebral/efeitos dos fármacos , Endossomos/metabolismo , Ferro/metabolismo , Lisossomos/metabolismo , Morfina/administração & dosagem , Neurônios/efeitos dos fármacos , Animais , Córtex Cerebral/metabolismo , Espinhas Dendríticas/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Masculino , Neurônios/citologia , Neurônios/metabolismo , Cultura Primária de Células , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa