Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Neurosci ; 44(14)2024 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-38383499

RESUMO

Human endogenous retroviruses (HERVs) are associated with the pathogenesis of amyotrophic lateral sclerosis (ALS); a disease characterized by motor neuron degeneration and cell death. The HERV-K subtype HML-2 envelope protein (HERV-K Env) is expressed in the brain, spinal cord, and cerebrospinal fluid of people living with ALS and through CD98 receptor-linked interactions causes neurodegeneration. HERV-K Env-induced increases in oxidative stress are implicated in the pathogenesis of ALS, and ferrous iron (Fe2+) generates reactive oxygen species (ROS). Endolysosome stores of Fe2+ are central to iron trafficking and endolysosome deacidification releases Fe2+ into the cytoplasm. Because HERV-K Env is an arginine-rich protein that is likely endocytosed and arginine is a pH-elevating amino acid, it is important to determine HERV-K Env effects on endolysosome pH and whether HERV-K Env-induced neurotoxicity is downstream of Fe2+ released from endolysosomes. Here, we showed using SH-SY5Y human neuroblastoma cells and primary cultures of human cortical neurons (HCNs, information on age and sex was not available) that HERV-K Env (1) is endocytosed via CD98 receptors, (2) concentration dependently deacidified endolysosomes, (3) decreased endolysosome Fe2+ concentrations, (4) increased cytosolic and mitochondrial Fe2+ and ROS levels, (5) depolarized mitochondrial membrane potential, and (6) induced cell death, effects blocked by an antibody against the CD98 receptor and by the endolysosome iron chelator deferoxamine. Thus, HERV-K Env-induced increases in cytosolic and mitochondrial Fe2+ and ROS as well as cell death appear to be mechanistically caused by HERV-K Env endocytosis, endolysosome deacidification, and endolysosome Fe2+ efflux into the cytoplasm.


Assuntos
Esclerose Lateral Amiotrófica , Retrovirus Endógenos , Neuroblastoma , Síndromes Neurotóxicas , Humanos , Esclerose Lateral Amiotrófica/patologia , Ferro , Espécies Reativas de Oxigênio , Arginina
2.
Ann Neurol ; 92(4): 545-561, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35801347

RESUMO

OBJECTIVE: Human endogenous retroviruses have been implicated in neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). Expression of human endogenous retrovirus K (HERV-K) subtype HML-2 envelope (Env) in human neuronal cultures and in transgenic mice results in neurotoxicity and neurodegeneration, and mice expressing HML-2 Env display behavioral and neuromuscular characteristics resembling ALS. This study aims to characterize the neurotoxic properties of HML-2 Env. METHODS: Env neurotoxicity was detected in ALS cerebrospinal fluid and confirmed using recombinant Env protein in a cell-based assay and a mouse model. The mechanism of neurotoxicity was assessed with immunoprecipitation followed by mass spectrometry and Western blot, and by screening a panel of inhibitors. RESULTS: We observed that recombinant HML-2 Env protein caused neurotoxicity resulting in neuronal cell death, retraction of neurites, and decreased neuronal electrical activity. Injection of the Env protein into the brains of mice also resulted in neuronal cell death. HML-2 Env protein was also found in the cerebrospinal fluid of patients with sporadic ALS. The neurotoxic properties of the Env and the cerebrospinal fluid could be rescued with the anti-Env antibody. The Env was found to bind to CD98HC complexed to ß1 integrin on the neuronal cell surface. Using a panel of compounds to screen for their ability to block Env-induced neurotoxicity, we found that several compounds were protective and are linked to the ß1 integrin pathway. INTERPRETATION: HERV-K Env is released extracellularly in ALS and causes neurotoxicity via a novel mechanism. Present results pave the way for new treatment strategies in sporadic ALS. ANN NEUROL 2022;92:545-561.


Assuntos
Esclerose Lateral Amiotrófica , Retrovirus Endógenos , Esclerose Lateral Amiotrófica/genética , Animais , Produtos do Gene env , Humanos , Integrina beta1 , Camundongos , Camundongos Transgênicos
3.
Brain ; 145(7): 2555-2568, 2022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35788639

RESUMO

The underlying mechanisms by which severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) leads to acute and long-term neurological manifestations remains obscure. We aimed to characterize the neuropathological changes in patients with coronavirus disease 2019 and determine the underlying pathophysiological mechanisms. In this autopsy study of the brain, we characterized the vascular pathology, the neuroinflammatory changes and cellular and humoral immune responses by immunohistochemistry. All patients died during the first wave of the pandemic from March to July 2020. All patients were adults who died after a short duration of the infection, some had died suddenly with minimal respiratory involvement. Infection with SARS-CoV-2 was confirmed on ante-mortem or post-mortem testing. Descriptive analysis of the pathological changes and quantitative analyses of the infiltrates and vascular changes were performed. All patients had multifocal vascular damage as determined by leakage of serum proteins into the brain parenchyma. This was accompanied by widespread endothelial cell activation. Platelet aggregates and microthrombi were found adherent to the endothelial cells along vascular lumina. Immune complexes with activation of the classical complement pathway were found on the endothelial cells and platelets. Perivascular infiltrates consisted of predominantly macrophages and some CD8+ T cells. Only rare CD4+ T cells and CD20+ B cells were present. Astrogliosis was also prominent in the perivascular regions. Microglial nodules were predominant in the hindbrain, which were associated with focal neuronal loss and neuronophagia. Antibody-mediated cytotoxicity directed against the endothelial cells is the most likely initiating event that leads to vascular leakage, platelet aggregation, neuroinflammation and neuronal injury. Therapeutic modalities directed against immune complexes should be considered.


Assuntos
COVID-19 , Doenças do Sistema Nervoso , Adulto , Complexo Antígeno-Anticorpo , Ativação do Complemento , Células Endoteliais , Humanos , Inflamação , SARS-CoV-2
4.
Proc Natl Acad Sci U S A ; 117(30): 17842-17853, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32669437

RESUMO

Stem cells are capable of unlimited proliferation but can be induced to form brain cells. Factors that specifically regulate human development are poorly understood. We found that human stem cells expressed high levels of the envelope protein of an endogenized human-specific retrovirus (HERV-K, HML-2) from loci in chromosomes 12 and 19. The envelope protein was expressed on the cell membrane of the stem cells and was critical in maintaining the stemness via interactions with CD98HC, leading to triggering of human-specific signaling pathways involving mammalian target of rapamycin (mTOR) and lysophosphatidylcholine acyltransferase (LPCAT1)-mediated epigenetic changes. Down-regulation or epigenetic silencing of HML-2 env resulted in dissociation of the stem cell colonies and enhanced differentiation along neuronal pathways. Thus HML-2 regulation is critical for human embryonic and neurodevelopment, while it's dysregulation may play a role in tumorigenesis and neurodegeneration.


Assuntos
Diferenciação Celular , Retrovirus Endógenos/fisiologia , Neurônios/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Biomarcadores , Diferenciação Celular/genética , Autorrenovação Celular/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Regulação Viral da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Ligação Proteica , Células-Tronco/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas do Envelope Viral/genética
5.
Proc Natl Acad Sci U S A ; 117(49): 31365-31375, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33229545

RESUMO

When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection.


Assuntos
Antivirais/análise , Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Inibidores de Proteases/análise , Inibidores de Proteases/farmacologia , Zika virus/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Inteligência Artificial , Chlorocebus aethiops , Modelos Animais de Doenças , Imunocompetência , Concentração Inibidora 50 , Metaciclina/farmacologia , Camundongos Endogâmicos C57BL , Inibidores de Proteases/uso terapêutico , Relação Quantitativa Estrutura-Atividade , Bibliotecas de Moléculas Pequenas , Células Vero , Infecção por Zika virus/tratamento farmacológico , Infecção por Zika virus/virologia
6.
Proc Natl Acad Sci U S A ; 115(51): E11924-E11932, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30509980

RESUMO

The human chaperonin Hsp60 is thought to play a role in the progression of Alzheimer's disease by mitigating against intracellular ß-amyloid stress. Here, we show that the bacterial homolog GroEL (51% sequence identity) reduces the neurotoxic effects of amyloid-ß(1-42) (Aß42) on human neural stem cell-derived neuronal cultures. To understand the mechanism of GroEL-mediated abrogation of neurotoxicity, we studied the interaction of Aß42 with GroEL using a variety of biophysical techniques. Aß42 binds to GroEL as a monomer with a lifetime of ∼1 ms, as determined from global analysis of multiple relaxation-based NMR experiments. Dynamic light scattering demonstrates that GroEL dissolves small amounts of high-molecular-weight polydisperse aggregates present in fresh soluble Aß42 preparations. The residue-specific transverse relaxation rate profile for GroEL-bound Aß42 reveals the presence of three anchor-binding regions (residues 16-21, 31-34, and 40-41) located within the hydrophobic GroEL-consensus binding sequences. Single-molecule FRET analysis of Aß42 binding to GroEL results in no significant change in the FRET efficiency of a doubly labeled Aß42 construct, indicating that Aß42 samples a random coil ensemble when bound to GroEL. Finally, GroEL substantially slows down the disappearance of NMR visible Aß42 species and the appearance of Aß42 protofibrils and fibrils as monitored by electron and atomic force microscopies. The latter observations correlate with the effect of GroEL on the time course of Aß42-induced neurotoxicity. These data provide a physical basis for understanding how Hsp60 may serve to slow down the progression of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/efeitos dos fármacos , Chaperonina 60/antagonistas & inibidores , Chaperonina 60/metabolismo , Síndromes Neurotóxicas/metabolismo , Fragmentos de Peptídeos/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Chaperonina 60/uso terapêutico , Escherichia coli/genética , Escherichia coli/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Humanos , Cinética , Espectroscopia de Ressonância Magnética , Microscopia de Força Atômica , Microscopia Eletrônica , Modelos Moleculares , Células-Tronco Neurais/efeitos dos fármacos , Síndromes Neurotóxicas/tratamento farmacológico , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Coloração e Rotulagem
7.
Mol Ther ; 27(12): 2067-2079, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31653397

RESUMO

Zika virus (ZIKV) infection is associated with microcephaly in neonates and Guillain-Barré syndrome in adults. ZIKV produces a class of nonstructural (NS) regulatory proteins that play a critical role in viral transcription and replication, including NS5, which possesses RNA-dependent RNA polymerase (RdRp) activity. Here we demonstrate that rilpivirine (RPV), a non-nucleoside reverse transcriptase inhibitor (NNRTI) used in the treatment of HIV-1 infection, inhibits the enzymatic activity of NS5 and suppresses ZIKV infection and replication in primary human astrocytes. Similarly, other members of the NNRTI family, including etravirine and efavirenz, showed inhibitory effects on viral infection of brain cells. Site-directed mutagenesis identified 14 amino acid residues within the NS5 RdRp domain (AA265-903), which are important for the RPV interaction and the inhibition of NS5 polymerase activity. Administration of RPV to ZIKV-infected interferon-alpha/beta receptor (IFN-A/R) knockout mice improved the clinical outcome and prevented ZIKV-induced mortality. Histopathological examination of the brains from infected animals revealed that RPV reduced ZIKV RNA levels in the hippocampus, frontal cortex, thalamus, and cerebellum. Repurposing of NNRTIs, such as RPV, for the inhibition of ZIKV replication offers a possible therapeutic strategy for the prevention and treatment of ZIKV-associated disease.


Assuntos
Fármacos Anti-HIV/farmacologia , Encéfalo/efeitos dos fármacos , Receptor de Interferon alfa e beta/fisiologia , Rilpivirina/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Infecção por Zika virus/tratamento farmacológico , Zika virus/efeitos dos fármacos , Animais , Encéfalo/virologia , Humanos , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Conformação Proteica , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
8.
J Neurovirol ; 24(1): 16-27, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29063516

RESUMO

Paroxetine and fluconazole have neuroprotective effects in an in vitro model of HIV protein-mediated neuronal injury. This study evaluated the safety, tolerability, and efficacy of both paroxetine and fluconazole for the treatment of HIV-associated neurocognitive disorder (HAND). A 24-week randomized double-blind, placebo-controlled 2 × 2 factorial design study was used. HIV+ individuals with cognitive impairment were enrolled in the 24-week trial. Participants were randomly assigned to one of four groups: (1) paroxetine 20 mg/day, (2) fluconazole 100 mg every 12 h, (3) paroxetine and fluconazole, or (4) placebo. Safety, tolerability, and efficacy were evaluated. Forty-five HIV+ individuals were enrolled. Medications were well tolerated. Compared to no paroxetine arms, HIV+ individuals receiving paroxetine showed improved NPZ8 summary scores, (mean change = 0.25 vs - 0.19, p = 0.049), CalCAP sequential test reaction time (mean change = 0.34 vs -0.23, p = 0.014), Trail Making Part B test performance (mean change = 0.49 vs - 0.33, p = 0.041), and FAS verbal fluency (mean change = 0.25 vs 0.02, p = 0.020) but a decline in the Letter number sequencing test (mean change = - 0.40 vs 0.26, p = 0.023). Biomarkers of cellular stress, inflammation, and neuronal damage were not affected by paroxetine. HIV+ individuals receiving fluconazole did not show a benefit in cognition and showed an increase in multiple markers of cellular stress compared to the no fluconazole arms. In conclusion, paroxetine was associated with improvement in a summary neuropsychological test measure and in several neuropsychological tests but worse performance in one neuropsychological test. Further studies of paroxetine for the treatment of HAND and to define its precise neuroprotective properties are warranted.


Assuntos
Complexo AIDS Demência/tratamento farmacológico , Fármacos Anti-HIV/uso terapêutico , Fluconazol/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Paroxetina/uso terapêutico , Complexo AIDS Demência/fisiopatologia , Complexo AIDS Demência/psicologia , Complexo AIDS Demência/virologia , Adulto , Antidepressivos de Segunda Geração/uso terapêutico , Antifúngicos/uso terapêutico , Método Duplo-Cego , Esquema de Medicação , Reposicionamento de Medicamentos , Análise Fatorial , Feminino , HIV-1/patogenicidade , Humanos , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Segurança do Paciente , Resultado do Tratamento
10.
J Neuroinflammation ; 14(1): 131, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28655310

RESUMO

BACKGROUND: The cause of neurodegeneration in progressive forms of multiple sclerosis is unknown. We investigated the impact of specific neuroinflammatory markers on human neurons to identify potential therapeutic targets for neuroprotection against chronic inflammation. METHODS: Surface immunocytochemistry directly visualized protease-activated receptor-1 (PAR1) and interleukin-1 (IL-1) receptors on neurons in human postmortem cortex in patients with and without neuroinflammatory lesions. Viability of cultured neurons was determined after exposure to cerebrospinal fluid from patients with progressive multiple sclerosis or purified granzyme B and IL-1ß. Inhibitors of PAR1 activation and of PAR1-associated second messenger signaling were used to elucidate a mechanism of neurotoxicity. RESULTS: Immunohistochemistry of human post-mortem brain tissue demonstrated cells expressing higher amounts of PAR1 near and within subcortical lesions in patients with multiple sclerosis compared to control tissue. Human cerebrospinal fluid samples containing granzyme B and IL-1ß were toxic to human neuronal cultures. Granzyme B was neurotoxic through activation of PAR1 and subsequently the phospholipase Cß-IP3 second messenger system. Inhibition of PAR1 or IP3 prevented granzyme B toxicity. IL-1ß enhanced granzyme B-mediated neurotoxicity by increasing PAR1 expression. CONCLUSIONS: Neurons within the inflamed central nervous system are imperiled because they express more PAR1 and are exposed to a neurotoxic combination of both granzyme B and IL-1ß. The effects of these inflammatory mediators may be a contributing factor in the progressive brain atrophy associated with neuroinflammatory diseases. Knowledge of how exposure to IL-1ß and granzyme B act synergistically to cause neuronal death yields potential novel neuroprotective treatments for neuroinflammatory diseases.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Granzimas/toxicidade , Interleucina-1beta/toxicidade , Receptor PAR-1/agonistas , Receptor PAR-1/metabolismo , Idoso , Idoso de 80 Anos ou mais , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/líquido cefalorraquidiano , Esclerose Múltipla/patologia
11.
J Neurovirol ; 20(6): 591-602, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25227932

RESUMO

Effective combined antiretroviral therapy (cART) in HIV-infected patients has made HIV a treatable infection; however, debilitating HIV-associated neurocognitive disorders (HAND) can still affect approximately 50% of HIV-infected individuals even under cART. While cART has greatly reduced the prevalence of the most severe form of HAND, milder forms have increased in prevalence, leaving the total proportion of HIV-infected individuals suffering from HAND relatively unchanged. In this study, an in vitro drug screen identified fluconazole and paroxetine as protective compounds against HIV gp120 and Tat neurotoxicity. Using an accelerated, consistent SIV/macaque model of HIV-associated CNS disease, we tested the in vivo neuroprotective capabilities of combination fluconazole/paroxetine (FluPar) treatment. FluPar treatment protected macaques from SIV-induced neurodegeneration, as measured by neurofilament light chain in the CSF, APP accumulation in axons, and CaMKIIα in the frontal cortex, but did not significantly reduce markers of neuroinflammation or plasma or CNS viral loads. Since HIV and SIV neurodegeneration is often attributed to accompanying neuroinflammation, this study provides proof of concept that neuroprotection can be achieved even in the face of ongoing neuroinflammation and viral replication.


Assuntos
Fluconazol/farmacologia , Neurônios/efeitos dos fármacos , Nootrópicos/farmacologia , Paroxetina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Complexo AIDS Demência/tratamento farmacológico , Complexo AIDS Demência/fisiopatologia , Complexo AIDS Demência/virologia , Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Síndrome da Imunodeficiência Adquirida/fisiopatologia , Síndrome da Imunodeficiência Adquirida/virologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Humanos , Macaca nemestrina , Proteínas de Neurofilamentos/líquido cefalorraquidiano , Proteínas de Neurofilamentos/genética , Neurônios/patologia , Neurônios/virologia , Cultura Primária de Células , Ratos , Síndrome de Imunodeficiência Adquirida dos Símios/líquido cefalorraquidiano , Síndrome de Imunodeficiência Adquirida dos Símios/fisiopatologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Vírus da Imunodeficiência Símia/fisiologia , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
12.
FASEB J ; 27(1): 174-86, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23038754

RESUMO

Alzheimer's disease (AD), one of the leading neurodegenerative disorders of older adults, which causes major socioeconomic burdens globally, lacks effective therapeutics without significant side effects. Besides the hallmark pathology of amyloid plaques and neurofibrillary tangles (NFTs), it has been reported that cyclin-dependent kinase 5 (Cdk5), a critical neuronal kinase, is hyperactivated in AD brains and is, in part, responsible for the above pathology. Here we show that a modified truncated 24-aa peptide (TFP5), derived from the Cdk5 activator p35, penetrates the blood-brain barrier after intraperitoneal injections, inhibits abnormal Cdk5 hyperactivity, and significantly rescues AD pathology (up to 70-80%) in 5XFAD AD model mice. The mutant mice, injected with TFP5 exhibit behavioral rescue, whereas no rescue was observed in mutant mice injected with either saline or scrambled peptide. However, TFP5 does not inhibit cell cycle Cdks or normal Cdk5/p35 activity, and thereby has no toxic side effects (even at 200 mg/kg), a common problem in most current therapeutics for AD. In addition, treated mice displayed decreased inflammation, amyloid plaques, NFTs, cell death, and an extended life by 2 mo. These results suggest TFP5 as a potential therapeutic, toxicity-free candidate for AD.


Assuntos
Doença de Alzheimer/prevenção & controle , Ativadores de Enzimas/farmacologia , Proteínas do Tecido Nervoso/farmacologia , Sequência de Aminoácidos , Animais , Apoptose , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Fosforilação
13.
Biomed Phys Eng Express ; 10(4)2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38608316

RESUMO

Objectives: The aim of this study was to evaluate Cu-64 PET phantom image quality using Bayesian Penalized Likelihood (BPL) and Ordered Subset Expectation Maximum with point-spread function modeling (OSEM-PSF) reconstruction algorithms. In the BPL, the regularization parameterßwas varied to identify the optimum value for image quality. In the OSEM-PSF, the effect of acquisition time was evaluated to assess the feasibility of shortened scan duration.Methods: A NEMA IEC PET body phantom was filled with known activities of water soluble Cu-64. The phantom was imaged on a PET/CT scanner and was reconstructed using BPL and OSEM-PSF algorithms. For the BPL reconstruction, variousßvalues (150, 250, 350, 450, and 550) were evaluated. For the OSEM-PSF algorithm, reconstructions were performed using list-mode data intervals ranging from 7.5 to 240 s. Image quality was assessed by evaluating the signal to noise ratio (SNR), contrast to noise ratio (CNR), and background variability (BV).Results: The SNR and CNR were higher in images reconstructed with BPL compared to OSEM-PSF. Both the SNR and CNR increased with increasingß, peaking atß= 550. The CNR for allß, sphere sizes and tumor-to-background ratios (TBRs) satisfied the Rose criterion for image detectability (CNR > 5). BPL reconstructed images withß= 550 demonstrated the highest improvement in image quality. For OSEM-PSF reconstructed images with list-mode data duration ≥ 120 s, the noise level and CNR were not significantly different from the baseline 240 s list-mode data duration.Conclusions: BPL reconstruction improved Cu-64 PET phantom image quality by increasing SNR and CNR relative to OSEM-PSF reconstruction. Additionally, this study demonstrated scan time can be reduced from 240 to 120 s when using OSEM-PSF reconstruction while maintaining similar image quality. This study provides baseline data that may guide future studies aimed to improve clinical Cu-64 imaging.


Assuntos
Algoritmos , Teorema de Bayes , Radioisótopos de Cobre , Processamento de Imagem Assistida por Computador , Imagens de Fantasmas , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Razão Sinal-Ruído , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Processamento de Imagem Assistida por Computador/métodos , Funções Verossimilhança , Humanos
14.
Viruses ; 16(4)2024 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-38675983

RESUMO

Human immunodeficiency virus type 1 (HIV-1) infection can result in HIV-associated neurocognitive disorder (HAND), a spectrum of disorders characterized by neurological impairment and chronic inflammation. Combined antiretroviral therapy (cART) has elicited a marked reduction in the number of individuals diagnosed with HAND. However, there is continual, low-level viral transcription due to the lack of a transcription inhibitor in cART regimens, which results in the accumulation of viral products within infected cells. To alleviate stress, infected cells can release accumulated products, such as TAR RNA, in extracellular vesicles (EVs), which can contribute to pathogenesis in neighboring cells. Here, we demonstrate that cART can contribute to autophagy deregulation in infected cells and increased EV release. The impact of EVs released from HIV-1 infected myeloid cells was found to contribute to CNS pathogenesis, potentially through EV-mediated TLR3 (Toll-like receptor 3) activation, suggesting the need for therapeutics to target this mechanism. Three HIV-1 TAR-binding compounds, 103FA, 111FA, and Ral HCl, were identified that recognize TAR RNA and reduce TLR activation. These data indicate that packaging of viral products into EVs, potentially exacerbated by antiretroviral therapeutics, may induce chronic inflammation of the CNS observed in cART-treated patients, and novel therapeutic strategies may be exploited to mitigate morbidity.


Assuntos
Autofagia , Vesículas Extracelulares , Infecções por HIV , HIV-1 , Receptor 3 Toll-Like , Vesículas Extracelulares/metabolismo , Humanos , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/genética , HIV-1/fisiologia , Infecções por HIV/virologia , Infecções por HIV/metabolismo , Infecções por HIV/tratamento farmacológico , Autofagia/efeitos dos fármacos , RNA Viral/metabolismo , RNA Viral/genética
15.
Nat Commun ; 15(1): 4163, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755145

RESUMO

TAR DNA-binding protein 43 (TDP-43) proteinopathy in brain cells is the hallmark of amyotrophic lateral sclerosis (ALS) but its cause remains elusive. Asparaginase-like-1 protein (ASRGL1) cleaves isoaspartates, which alter protein folding and susceptibility to proteolysis. ASRGL1 gene harbors a copy of the human endogenous retrovirus HML-2, whose overexpression contributes to ALS pathogenesis. Here we show that ASRGL1 expression was diminished in ALS brain samples by RNA sequencing, immunohistochemistry, and western blotting. TDP-43 and ASRGL1 colocalized in neurons but, in the absence of ASRGL1, TDP-43 aggregated in the cytoplasm. TDP-43 was found to be prone to isoaspartate formation and a substrate for ASRGL1. ASRGL1 silencing triggered accumulation of misfolded, fragmented, phosphorylated and mislocalized TDP-43 in cultured neurons and motor cortex of female mice. Overexpression of ASRGL1 restored neuronal viability. Overexpression of HML-2 led to ASRGL1 silencing. Loss of ASRGL1 leading to TDP-43 aggregation may be a critical mechanism in ALS pathophysiology.


Assuntos
Esclerose Lateral Amiotrófica , Proteínas de Ligação a DNA , Neurônios , Proteinopatias TDP-43 , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Humanos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos , Feminino , Proteinopatias TDP-43/metabolismo , Proteinopatias TDP-43/patologia , Proteinopatias TDP-43/genética , Neurônios/metabolismo , Neurônios/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Masculino , Córtex Motor/metabolismo , Córtex Motor/patologia
16.
Neurology ; 100(13): 624-628, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36526429

RESUMO

BACKGROUND AND OBJECTIVES: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can cause a wide range of neurologic complications; however, its neuropenetrance during the acute phase of the illness is unknown. METHODS: Extracellular vesicles were isolated from brain biopsy tissue from a patient undergoing epilepsy surgery using ultracentrifugation and analyzed by Western blot and qPCR for the presence of virus protein and RNA, respectively. Biopsy tissue was assessed by immunohistochemistry for the presence of microvascular damage and compared with 3 other non-COVID surgical epilepsy brain tissues. RESULTS: We demonstrate the presence of viral nucleocapsid protein in extracellular vesicles and microvascular disease in the brain of a patient undergoing epilepsy surgery shortly after SARS-CoV-2 infection. Endothelial cell activation was indicated by increased levels of platelet endothelial cell adhesion molecule-1 and was associated with fibrinogen leakage and immune cell infiltration in the biopsy tissue as compared with control non-COVID surgical epilepsy brain tissues. DISCUSSION: Despite the lack of evidence of viral replication within the brain, the presence of the nucleocapsid protein was associated with disease-specific endothelial cell activation, fibrinogen leakage, and immune cell infiltration.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , Nucleocapsídeo/metabolismo , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/metabolismo , Encéfalo/metabolismo
17.
Brain Commun ; 5(1): fcac298, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36655171

RESUMO

Recent studies suggest that changes in neuronal metabolism are associated with epilepsy. High rates of ATP depletion, lactate dehydrogenase A and lactate production have all been found in epilepsy patients, animal and tissue culture models. As such, it can be hypothesized that chronic seizures lead to continuing elevations in neuronal energy demand which may lead to an adapted metabolic response and elevations of lactate dehydrogenase A. In this study, we examine elevations in the lactate dehydrogenase A protein as a long-term cellular adaptation to elevated metabolic demand from chronic neuronal activation. We investigate this cellular adaptation in human tissue samples and explore the mechanisms of lactate dehydrogenase A upregulation using cultured neurones treated with low Mg2+, a manipulation that leads to NMDA-mediated neuronal activation. We demonstrate that human epileptic tissue preferentially upregulates neuronal lactate dehydrogenase A, and that in neuronal cultures chronic and repeated elevations in neural activity lead to upregulation of neuronal lactate dehydrogenase A. Similar to states of hypoxia, this metabolic change occurs through the AMP-activated protein kinase/hypoxia-inducible factor-1α pathway. Our data therefore reveal a novel long-term bioenergetic adaptation that occurs in chronically activated neurones and provide a basis for understanding the interplay between metabolism and neural activity during epilepsy.

18.
Exp Neurol ; 367: 114469, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37327963

RESUMO

Prenatal Zika virus (ZIKV) infection is a serious global concern as it can lead to brain injury and many serious birth defects, collectively known as congenital Zika syndrome. Brain injury likely results from viral mediated toxicity in neural progenitor cells. Additionally, postnatal ZIKV infections have been linked to neurological complications, yet the mechanisms driving these manifestations are not well understood. Existing data suggest that the ZIKV envelope protein can persist in the central nervous system for extended periods of time, but it is unknown if this protein can independently contribute to neuronal toxicity. Here we find that the ZIKV envelope protein is neurotoxic, leading to overexpression of poly adenosine diphosphate -ribose polymerase 1, which can induce parthanatos. Together, these data suggest that neuronal toxicity resulting from the envelope protein may contribute to the pathogenesis of post-natal ZIKV-related neurologic complications.


Assuntos
Lesões Encefálicas , Doenças do Sistema Nervoso , Síndromes Neurotóxicas , Infecção por Zika virus , Zika virus , Gravidez , Feminino , Humanos , Zika virus/metabolismo , Infecção por Zika virus/complicações , Infecção por Zika virus/patologia , Proteínas do Envelope Viral/metabolismo , Neurônios/patologia
19.
J Clin Invest ; 133(13)2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37395282

RESUMO

Human endogenous retroviruses (HERVs) are ancestral viral relics that constitute nearly 8% of the human genome. Although normally silenced, the most recently integrated provirus HERV-K (HML-2) can be reactivated in certain cancers. Here, we report pathological expression of HML-2 in malignant gliomas in both cerebrospinal fluid and tumor tissue that was associated with a cancer stem cell phenotype and poor outcomes. Using single-cell RNA-Seq, we identified glioblastoma cellular populations with elevated HML-2 transcripts in neural progenitor-like cells (NPC-like) that drive cellular plasticity. Using CRISPR interference, we demonstrate that HML-2 critically maintained glioblastoma stemness and tumorigenesis in both glioblastoma neurospheres and intracranial orthotopic murine models. Additionally, we demonstrate that HML-2 critically regulated embryonic stem cell programs in NPC-derived astroglia and altered their 3D cellular morphology by activating the nuclear transcription factor OCT4, which binds to an HML-2-specific long-terminal repeat (LTR5Hs). Moreover, we discovered that some glioblastoma cells formed immature retroviral virions, and inhibiting HML-2 expression with antiretroviral drugs reduced reverse transcriptase activity in the extracellular compartment, tumor viability, and pluripotency. Our results suggest that HML-2 fundamentally contributes to the glioblastoma stem cell niche. Because persistence of glioblastoma stem cells is considered responsible for treatment resistance and recurrence, HML-2 may serve as a unique therapeutic target.


Assuntos
Retrovirus Endógenos , Glioblastoma , Humanos , Animais , Camundongos , Retrovirus Endógenos/genética , Glioblastoma/genética , Nicho de Células-Tronco , Provírus/genética
20.
J Neurosci ; 31(47): 17074-90, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22114277

RESUMO

Infection by the human immunodeficiency virus (HIV) can result in debilitating neurological syndromes collectively known as HIV-associated neurocognitive disorders. Although the HIV coat protein gp120 has been identified as a potent neurotoxin that enhances NMDA receptor function, the exact mechanisms for this effect are not known. Here we provide evidence that gp120 activates two separate signaling pathways that converge to enhance NMDA-evoked calcium flux by clustering NMDA receptors in modified membrane microdomains. gp120 enlarged and stabilized the structure of lipid microdomains on dendrites by mechanisms that involved a redox-regulated translocation of a sphingomyelin hydrolase (neutral sphingomyelinase-2) to the plasma membrane. A concurrent pathway was activated that accelerated the forward traffic of NMDA receptors by a PKA-dependent phosphorylation of the NR1 C-terminal serine 897 (masks an ER retention signal), followed by a PKC-dependent phosphorylation of serine 896 (important for surface expression). NMDA receptors were preferentially targeted to synapses and clustered in modified membrane microdomains. In these conditions, NMDA receptors were unable to laterally disperse and did not internalize, even in response to strong agonist induction. Focal NMDA-evoked calcium bursts were enhanced by threefold in these regions. Inhibiting membrane modification or NR1 phosphorylation prevented gp120 from accelerating the surface localization of NMDA receptors. Disrupting the structure of membrane microdomains after gp120 treatments restored the ability of NMDA receptors to disperse and internalize. These findings demonstrate that gp120 contributes to synaptic dysfunction in the setting of HIV infection by interfering with NMDA receptor trafficking.


Assuntos
Proteína gp120 do Envelope de HIV/fisiologia , Microdomínios da Membrana/metabolismo , Agregação de Receptores/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Células Cultivadas , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/virologia , Humanos , Microdomínios da Membrana/virologia , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA