Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 212
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 300(1): 105526, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38043797

RESUMO

Despite antiretroviral therapy (ART), chronic forms of HIV-associated neurocognitive disorders (HAND) affect an estimated 50% of individuals living with HIV, greatly impacting their quality of life. The prevailing theory of HAND progression posits that chronic inflammation arising from the activation of latent viral reservoirs leads to progressive damage in the central nervous system (CNS). Recent evidence indicates that blood-brain barrier (BBB) pericytes are capable of active HIV-1 infection; however, their latent infection has not been defined. Given their location and function, BBB pericytes are poised to be a key viral reservoir in the development of HAND. We present the first transcriptional analysis of uninfected, active, and latent human BBB pericytes, revealing distinct transcriptional phenotypes. In addition, we demonstrate that latent infection of BBB pericytes relies on AKT signaling for reservoir survival. These findings provide insight into the state of reservoir maintenance in the CNS during HIV-1 infection and provide novel targets for reservoir clearance.


Assuntos
Barreira Hematoencefálica , Reservatórios de Doenças , Infecções por HIV , HIV-1 , Infecção Latente , Pericitos , Humanos , Barreira Hematoencefálica/virologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/transmissão , Infecções por HIV/virologia , Infecção Latente/virologia , Pericitos/virologia , Proteínas Proto-Oncogênicas c-akt/genética , Qualidade de Vida , Latência Viral , Reservatórios de Doenças/virologia
2.
Am J Physiol Cell Physiol ; 326(2): C487-C504, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38145295

RESUMO

Blood-brain barrier (BBB) breakdown is one of the pathophysiological characteristics of ischemic stroke, which may contribute to the progression of brain tissue damage and subsequent neurological impairment. Human immunodeficiency virus (HIV)-infected individuals are at greater risk for ischemic stroke due to diminished immune function and HIV-associated vasculopathy. Studies have shown that astrocytes are involved in maintaining BBB integrity and facilitating HIV-1 infection in the brain. The present study investigated whether targeting astrocyte-endothelial cell signaling with cenicriviroc (CVC), a dual chemokine receptor (CCR)2 and CCR5 antagonist, may protect against dysregulation of cross talk between these cells after oxygen-glucose deprivation/reoxygenation (OGD/R) combined with HIV-1 infection. Permeability assay with 10 kDa fluorescein isothiocyanate (FITC)-dextran demonstrated that CVC alleviated endothelial barrier disruption in noncontact coculture of human brain microvascular endothelial cells (HBMECs) with HIV-1-infected human astrocytes, and reversed downregulation of tight junction protein claudin-5 induced by OGD/R- and HIV-1. Moreover, CVC attenuated OGD/R- and HIV-1-triggered upregulation of the NOD-like receptor protein-3 (NLRP3) inflammasome and IL-1ß secretion. Treatment with CVC also suppressed astrocyte pyroptosis by attenuating cleaved caspase-1 levels and the formation of cleaved N-terminal GSDMD (N-GSDMD). Secretome profiling revealed that CVC ameliorated secretion levels of chemokine CC chemokine ligand 17 (CCL17), adhesion molecule intercellular adhesion molecule-1 (ICAM-1), and T cell activation modulator T cell immunoglobulin and mucin domain 3 (TIM-3) by astrocytes synergistically induced by OGD/R and HIV-1. Overall, these results suggest that CVC contributes to restoring astrocyte-endothelial cross interactions in an astrocyte-dependent manner via protection against NLRP3 activation and pyroptosis.NEW & NOTEWORTHY The present study reveals the role of astrocytic NOD-like receptor protein-3 (NLRP3) inflammasome in dysfunctional astrocyte-endothelial cross interactions triggered in response to oxygen/glucose deprivation injury associated with human immunodeficiency virus type 1 (HIV-1) infection. Our results suggest that blocking NLRP3 inflammasome activation and pyroptosis-mediated inflammation with cenicriviroc (CVC) may constitute a potentially effective therapeutic strategy for blood-brain barrier (BBB) protection during HIV-1-associated ischemic stroke.


Assuntos
Infecções por HIV , HIV-1 , Imidazóis , AVC Isquêmico , Sulfóxidos , Humanos , Astrócitos/metabolismo , Inflamassomos/metabolismo , Inflamassomos/farmacologia , HIV-1/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Células Endoteliais/metabolismo , Piroptose , Proteínas NLR/metabolismo , Oxigênio/metabolismo , Isquemia/metabolismo , AVC Isquêmico/metabolismo , Glucose/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo
3.
Anesth Analg ; 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38446700

RESUMO

BACKGROUND: Clinical data demonstrate that chronic use of opioid analgesics increases neuropathic pain in people living with human immunodeficiency virus (HIV). Therefore, it is important to elucidate the molecular mechanisms of HIV-related chronic pain. In this study, we investigated the role of the transcription factor cMyc, epigenetic writer enhancer of zeste homology 2 (EZH2), and sirtuin 3 (Sirt3) pathway in HIV glycoprotein gp120 with morphine (gp120M)-induced neuropathic pain in rats. METHODS: Neuropathic pain was induced by intrathecal administration of recombinant gp120 with morphine. Mechanical withdrawal threshold was measured using von Frey filaments, and thermal latency using the hotplate test. Spinal expression of cMyc, EZH2, and Sirt3 were measured using Western blots. Antinociceptive effects of intrathecal administration of antisense oligodeoxynucleotide against cMyc, a selective inhibitor of EZH2, or recombinant Sirt3 were tested. RESULTS: In the spinal dorsal horn, gp120M upregulated expression of cMyc (ratio of gp120M versus control, 1.68 ± 0.08 vs 1.00 ± 0.14, P = .0132) and EZH2 (ratio of gp120M versus control, 1.76 ± 0.05 vs 1.00 ± 0.16, P = .006), and downregulated Sirt3 (ratio of control versus gp120M, 1.00 ± 0.13 vs 0.43 ± 0.10, P = .0069) compared to control. Treatment with intrathecal antisense oligodeoxynucleotide against cMyc, GSK126 (EZH2 selective inhibitor), or recombinant Sirt3 reduced mechanical allodynia and thermal hyperalgesia in this gp120M pain model. Knockdown of cMyc reduced spinal EZH2 expression in gp120M treated rats. Chromatin immunoprecipitation (ChIP) assay showed that enrichment of cMyc binding to the ezh2 gene promoter region was increased in the gp120M-treated rat spinal dorsal horn, and that intrathecal administration of antisense ODN against cMyc (AS-cMyc) reversed the increased enrichment of cMyc. Enrichment of trimethylation of histone 3 on lysine residue 27 (H3K27me3; an epigenetic mark associated with the downregulation of gene expression) binding to the sirt3 gene promoter region was upregulated in the gp120M-treated rat spinal dorsal horn; that intrathecal GSK126 reversed the increased enrichment of H3K27me3 in the sirt3 gene promoter. Luciferase reporter assay demonstrated that cMyc mediated ezh2 gene transcription at the ezh2 gene promoter region, and that H3K27me3 silenced sirt3 gene transcription at the gene promoter region. CONCLUSION: These results demonstrated that spinal Sirt3 decrease in gp120M-induced neuropathic pain was mediated by cMyc-EZH2/H3K27me3 activity in an epigenetic manner. This study provided new insight into the mechanisms of neuropathic pain in HIV patients with chronic opioids.

4.
Int J Mol Sci ; 25(3)2024 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-38339172

RESUMO

The study investigated a profile of chemokines and growth factors in the aqueous humor (AH) of eyes with Fuch's endothelial corneal dystrophy (FECD) and cataracts in comparison with cataract patients as a control group. A total of 52 AH samples (26 FECD + cataract and 26 cataract/control) were collected before cataract surgery. None of the patients had any clinically apparent inflammation at the time of AH collection. The AH levels of MCP-1 (CCL2), MIP-1α (CCL3), MIP-1ß(CCL4), RANTES (CCL5), eotaxin (CCL11), IP-10 (CXCL10), FGF basic, G-CSF, GM-CSF, PDGF-bb, and VEGF were compared between the groups. The analyses were performed using the Bio-Plex 200 System from Bio-Rad. Among the studied parameters, the AH levels of RANTES, eotaxin, and IP-10 significantly increased in the FECD + cataract eyes, compared with the cataract controls (p < 0.05). Elevated levels of the RANTES, Eotaxin, and IP-10 indicate more intense inflammation in the eyes of patients in the FECD + cataract group. Moreover, these factors exhibit potential as predictive biomarkers for early detection of FECD in cataract patients. The discovery of elevated concentrations of biochemical markers in a patient, who has not yet received a clinical diagnosis, may suggest the need for heightened observation of the other eye to monitor the potential development of FECD.


Assuntos
Catarata , Distrofias Hereditárias da Córnea , Humanos , Citocinas/metabolismo , Humor Aquoso/metabolismo , Quimiocina CXCL10/metabolismo , Quimiocinas/metabolismo , Catarata/metabolismo , Inflamação/metabolismo , Distrofias Hereditárias da Córnea/metabolismo
5.
Mol Cell Biochem ; 478(3): 581-595, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35976519

RESUMO

We evaluated the mechanistic link between circadian rhythms and gut barrier permeability. Mice were subjected to either constant 24-h light (LL) or 12-h light/dark cycles (LD). Mice housed in LL experienced a significant increase in gut barrier permeability that was associated with dysregulated ß-catenin expression and altered expression of tight junction (TJ) proteins. Silencing of ß-catenin resulted in disruption of barrier function in SW480 cells, with ß-catenin appearing to be an upstream regulator of the core circadian components, such as Bmal1, Clock, and Per1/2. In addition, ß-catenin silencing downregulated ZO-1 and occludin TJ proteins with only limited or no changes at their mRNA levels, suggesting post transcriptional regulation. Indeed, silencing of ß-catenin significantly upregulated expression of matrix metallopeptidase (MMP)-2 and MMP-9, and blocking MMP-2/9 activity attenuated epithelial disruption induced by ß-catenin silencing. These results indicate the regulatory role of circadian disruption on gut barrier integrity and the associations between TJ proteins and circadian rhythms, while demonstrating the regulatory role of ß-catenin in this process.


Assuntos
Cateninas , Ritmo Circadiano , Animais , Camundongos , Cateninas/genética , Regulação da Expressão Gênica
6.
Environ Res ; 220: 115148, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36580985

RESUMO

Assessing health outcomes associated with exposure to polychlorinated biphenyls (PCBs) is important given their persistent and ubiquitous nature. PCBs are classified as a Group 1 carcinogen, but the full range of potential noncancer health effects from exposure to PCBs has not been systematically summarized and evaluated. We used systematic review methods to identify and screen the literature using combined manual review and machine learning approaches. A protocol was developed that describes the literature search strategy and Populations, Exposures, Comparators, and Outcomes (PECO) criteria used to facilitate subsequent screening and categorization of literature into a systematic evidence map of PCB exposure and noncancer health endpoints across 15 organs/systems. A comprehensive literature search yielded 62,599 records. After electronic prioritization steps, 17,037 studies were manually screened at the title and abstract level. An additional 900 studies identified by experts or supplemental searches were also included. After full-text screening of 3889 references, 1586 studies met the PECO criteria. Relevant study details such as the endpoints assessed, exposure duration, and species were extracted into literature summary tables. This review compiles and organizes the human and mammalian studies from these tables into an evidence map for noncancer health endpoints and PCB mixture exposure to identify areas of robust research as well as areas of uncertainty that would benefit from future investigation. Summary data are available online as interactive visuals with downloadable metadata. Sufficient research is available to inform PCB hazard assessments for most organs/systems, but the amount of data to inform associations with specific endpoints differs. Furthermore, despite many years of research, sparse data exist for inhalation and dermal exposures, which are highly relevant human exposure routes. This evidence map provides a foundation for future systematic reviews and noncancer hazard assessments of PCB mixtures and for strategic planning of research to inform areas of greater uncertainty.


Assuntos
Bifenilos Policlorados , Animais , Humanos , Carcinógenos , Mamíferos , Bifenilos Policlorados/toxicidade , Incerteza
7.
Retrovirology ; 19(1): 27, 2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36476484

RESUMO

While HIV-1 is primarily an infection of CD4 + T cells, there is an emerging interest towards understanding how infection of other cell types can contribute to HIV-associated comorbidities. For HIV-1 to cross from the blood stream into tissues, the virus must come in direct contact with the vascular endothelium, including pericytes that envelope vascular endothelial cells. Pericytes are multifunctional cells that have been recognized for their essential role in angiogenesis, vessel maintenance, and blood flow rate. Most importantly, recent evidence has shown that pericytes can be a target of HIV-1 infection and support an active stage of the viral life cycle, with latency also suggested by in vitro data. Pericyte infection by HIV-1 has been confirmed in the postmortem human brains and in lungs from SIV-infected macaques. Moreover, pericyte dysfunction has been implicated in a variety of pathologies ranging from ischemic stroke to diabetes, which are common comorbidities among people with HIV-1. In this review, we discuss the role of pericytes during HIV-1 infection and their contribution to the progression of HIV-associated comorbidities.


Assuntos
Infecções por HIV , HIV-1 , Humanos , Células Endoteliais
8.
Cell Mol Neurobiol ; 42(7): 2131-2146, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34086179

RESUMO

The blood-brain barrier (BBB) is essential for the homeostasis of the central nervous system (CNS). Functions of the BBB are performed by the neurovascular unit (NVU), which consists of endothelial cells, pericytes, astrocytes, microglia, basement membrane, and neurons. NVU cells interact closely and together are responsible for neurovascular coupling, BBB integrity, and transendothelial fluid transport. Studies have shown that NVU dysfunction is implicated in several acute and chronic neurological diseases, including Alzheimer's disease, multiple sclerosis, and stroke. The mechanisms of NVU disruption remain poorly understood, partially due to difficulties in selective targeting of NVU cells. In this review, we discuss the relative merits of available protein markers and drivers of the NVU along with recent advancements that have been made in the field to increase efficiency and specificity of NVU research.


Assuntos
Barreira Hematoencefálica , Células Endoteliais , Astrócitos , Sistema Nervoso Central , Pericitos
9.
Mol Pharm ; 19(7): 2254-2267, 2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35506882

RESUMO

The United States is in the midst of an opioid epidemic that is linked to a number of serious health issues, including an increase in cerebrovascular events, namely, stroke. Chronic prescription opioid use exacerbates the risk and severity of ischemic stroke, contributing to stroke as the fifth overall cause of death in the United States and costing the US health care system over $30 billion annually. Pathologically, opioids challenge the integrity of the blood-brain barrier (BBB), resulting in a dysregulation of tight junction (TJ) proteins that are crucial in maintaining barrier homeostasis. Despite this, treatment options for ischemic stroke are limited, and there are no pharmacological options to attenuate TJ damage, including in incidents that are linked to opioid use. Herein, we have generated carrier-free, pure "nanodrugs" or nanoparticles of naloxone and naltrexone with enhanced therapeutic properties compared to the original (parent) drugs. The generated nanoformulations of both opioid antagonists exhibited successful attenuation of morphine- or oxycodone-induced alterations of TJ protein expression and reduced oxidative stress to a greater extent than the parent drugs (non-nano). As a proof of concept, we then proceeded to evaluate the therapeutic effectiveness of the generated nanodrugs in an ischemic stroke model of mice exposed to morphine or oxycodone. Our results demonstrate that the opioid antagonist nanoformulations reduced stroke severity in mice. Overall, this research implements advances in nanotechnology-based repurposing of FDA-approved therapeutics, and the obtained results also suggest underlying pharmacological mechanisms of opioid antagonists, further supporting these opioid antagonists and their respective nanoformulations as potential therapeutic agents for ischemic stroke.


Assuntos
AVC Isquêmico , Nanopartículas , Transtornos Relacionados ao Uso de Opioides , Acidente Vascular Cerebral , Analgésicos Opioides/uso terapêutico , Animais , AVC Isquêmico/tratamento farmacológico , Camundongos , Morfina/uso terapêutico , Naloxona , Naltrexona , Nanopartículas/uso terapêutico , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Oxicodona , Acidente Vascular Cerebral/tratamento farmacológico , Proteínas de Junções Íntimas
10.
Neurobiol Dis ; 155: 105388, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33962010

RESUMO

Human immunodeficiency virus-1 (HIV-1) has been shown to cross the blood-brain barrier and cause HIV-associated neurocognitive disorders (HAND) through a process that may involve direct or indirect interactions with the central nervous system (CNS) cells and alterations of amyloid ß (Aß) homeostasis. The present study focused on the mechanisms of HIV-1 infecting human neural progenitor cells (hNPCs) and affecting NPC intercellular communications with human brain endothelial cells (HBMEC). Despite the lack of the CD4 receptor, hNPCs were effectively infected by HIV-1 via a mechanism involving the chemokine receptors, CXCR4 and CCR5. HIV-1 infection increased expression of connexin-43 (Cx43), phosphorylated Cx43 (pCx43), and pannexin 2 (Panx2) protein levels in hNPCs, suggesting alterations in gap-junction (GJ) and pannexin channel communication. Indeed, a functional GJ assay indicated an increase in communication between HIV-infected hNPCs and non-infected HBMEC. We next analyzed the impact of HBMEC-derived extracellular vesicles (EVs) and EVs carrying Aß (EV-Aß) on the expression of Cx43, pCx43, and Panx2 in HIV-1 infected and non-infected hNPCs. Exposure to EV-Aß resulted in significant reduction of Cx43 and pCx43 protein expression in non-infected hNPCs when compared to EV controls. Interestingly, EV-Aß treatment significantly increased levels of Cx43, pCx43, and Panx2 in HIV-1-infected hNPCs when compared to non-infected controls. These results were confirmed in a GJ functional assay and an ATP release assay, which is an indicator of connexin hemichannel and/or pannexin channel functions. Overall, the current study demonstrates the importance of hNPCs in HIV-1 infection and indicates that intercellular communications between infected hNPCs and HBMEC can be effectively modulated by EVs carrying Aß as their cargo.


Assuntos
Comunicação Celular/fisiologia , Vesículas Extracelulares/metabolismo , Junções Comunicantes/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Células-Tronco Neurais/metabolismo , Peptídeos beta-Amiloides/metabolismo , Linhagem Celular , Células Cultivadas , Endotélio Vascular/metabolismo , Endotélio Vascular/virologia , Vesículas Extracelulares/virologia , Junções Comunicantes/virologia , Humanos , Células-Tronco Neurais/virologia
11.
J Neuroinflammation ; 18(1): 167, 2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34325716

RESUMO

BACKGROUND: Neurological complications are common in patients affected by COVID-19 due to the ability of SARS-CoV-2 to infect brains. While the mechanisms of this process are not fully understood, it has been proposed that SARS-CoV-2 can infect the cells of the neurovascular unit (NVU), which form the blood-brain barrier (BBB). The aim of the current study was to analyze the expression pattern of the main SARS-CoV-2 receptors in naïve and HIV-1-infected cells of the NVU in order to elucidate a possible pathway of the virus entry into the brain and a potential modulatory impact of HIV-1 in this process. METHODS: The gene and protein expression profile of ACE2, TMPRSS2, ADAM17, BSG, DPP4, AGTR2, ANPEP, cathepsin B, and cathepsin L was assessed by qPCR, immunoblotting, and immunostaining, respectively. In addition, we investigated if brain endothelial cells can be affected by the exposure to the S1 subunit of the S protein, the domain responsible for the direct binding of SARS-CoV-2 to the ACE2 receptors. RESULTS: The receptors involved in SARS-CoV-2 infection are co-expressed in the cells of the NVU, especially in astrocytes and microglial cells. These receptors are functionally active as exposure of endothelial cells to the SARS CoV-2 S1 protein subunit altered the expression pattern of tight junction proteins, such as claudin-5 and ZO-1. Additionally, HIV-1 infection upregulated ACE2 and TMPRSS2 expression in brain astrocytes and microglia cells. CONCLUSIONS: These findings provide key insight into SARS-CoV-2 recognition by cells of the NVU and may help to develop possible treatment of CNS complications of COVID-19.


Assuntos
Vasos Sanguíneos/metabolismo , COVID-19/complicações , Infecções por HIV/metabolismo , HIV-1 , Neurônios/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , SARS-CoV-2 , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Astrócitos/metabolismo , Encefalopatias/etiologia , Células Cultivadas , Endotélio Vascular/metabolismo , Humanos , Microglia/metabolismo , Doenças do Sistema Nervoso/etiologia , Cultura Primária de Células , Receptor Tipo 2 de Angiotensina , Replicação Viral
12.
FASEB J ; 34(12): 16319-16332, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33058236

RESUMO

HIV-1 enters the brain by altering properties of the blood-brain barrier (BBB). Recent evidence indicates that among cells of the BBB, pericytes are prone to HIV-1 infection. Occludin (ocln) and caveolin-1 (cav-1) are critical determinants of BBB integrity that can regulate barrier properties of the BBB in response to HIV-1 infection. Additionally, Alix is an early acting endosomal factor involved in HIV-1 budding from the cells. The aim of the present study was to evaluate the role of cav-1, ocln, and Alix in HIV-1 infection of brain pericytes. Our results indicated that cav-1, ocln, and Alix form a multi-protein complex in which they cross-regulate each other's expression. Importantly, the stability of this complex was affected by HIV-1 infection. Modifications of the complex resulted in diminished HIV-1 infection and alterations of the cytokine profile produced by brain pericytes. These results identify a novel mechanism involved in HIV-1 infection contributing to a better understanding of the HIV-1 pathology and the associated neuroinflammatory responses.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Caveolina 1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Infecções por HIV/metabolismo , Ocludina/metabolismo , Pericitos/metabolismo , Transporte Biológico/fisiologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/virologia , Encéfalo/virologia , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Células HEK293 , Infecções por HIV/virologia , HIV-1/patogenicidade , Humanos
13.
Biol Sport ; 38(4): 545-553, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34937963

RESUMO

The aim of this study was the assessment of sodium bicarbonate supplementation (NaHCO3 -) on anaerobic and cognitive performance, assuming ergogenic effect of HCO3 by improving buffering capacity and greater lactate efflux, which may have indirect effect on circulating neurotrophin level (e.g BDNF, IGF-1) and memory. Sixteen well-trained judo athletes completed a randomized trial of either a NaHCO3 - (EG) (5000 mg x 2/day/90 min before training)or placebo for 21 days (CG). Before and after treatment, athletes completed double Wingate test (Wt) protocol following which they performed perceived Working Memory test (pWM). Results suggested significant increase in Upper Limb Total Work (with p = 0.011), Mean Power (with p = 0.001), post exercise LA concentration (from 15.51 mmol/L to 18.10 mmol/L with p = 0.01) and HCO3rest concentrations (from 27.37 mmol/l to 28.91 mmol/l with p = 0.001), when compared to baseline values in EG. The analysis showed statistically significant increase in values for IGF-1 (with p = 0.001) and decrease for cortisol and BDNF (with p = 0.001) in EG and CG, when pre and post exercise values were compared. We also revealed statistically significant decrease in values for display time after ingestion of HCO3 between pre and post exercise (with p = 0.002) In conclusion, the lack of a substantial relationship between exerkines (IGF-1, BDNF) and memory in the present study might suggest that exercise induced lactate levels is dominant mechanism improving working memory in well-train athletes.

14.
Brain ; 142(3): 502-511, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668645

RESUMO

Pericytes are multifunctional cells wrapped around endothelial cells via cytoplasmic processes that extend along the abluminal surface of the endothelium. The interactions between endothelial cells and pericytes of the blood-brain barrier are necessary for proper formation, development, stabilization, and maintenance of the blood-brain barrier. Blood-brain barrier pericytes regulate paracellular flow between cells, transendothelial fluid transport, maintain optimal chemical composition of the surrounding microenvironment, and protect endothelial cells from potential harmful substances. Thus, dysfunction or loss of blood-brain barrier pericytes is an important factor in the pathogenesis of several diseases that are associated with microvascular instability. Importantly, recent research indicates that blood-brain barrier pericytes can be a target of HIV-1 infection able to support productive HIV-1 replication. In addition, blood-brain barrier pericytes are prone to establish a latent infection, which can be reactivated by a mixture of histone deacetylase inhibitors in combination with TNF. HIV-1 infection of blood-brain barrier pericytes has been confirmed in a mouse model of HIV-1 infection and in human post-mortem samples of HIV-1-infected brains. Overall, recent evidence indicates that blood-brain barrier pericytes can be a previously unrecognized HIV-1 target and reservoir in the brain.


Assuntos
Barreira Hematoencefálica/patologia , Pericitos/metabolismo , Pericitos/virologia , Animais , Transporte Biológico , Encéfalo/patologia , Células Endoteliais/patologia , Infecções por HIV/metabolismo , HIV-1/patogenicidade , Humanos , Camundongos , Pericitos/fisiologia
15.
Environ Toxicol ; 35(1): 87-96, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31515868

RESUMO

Increasing consumption of metal-oxide nanoparticles (NPs) and carbon-based nanomaterials has caused significant concern about their potential hazards in aquatic environments. The release of NPs into aquatic environments could result in adsorption of NPs on microorganisms. While metal-oxide NP-conjugated carbon-based nanohybrids (NHs) may exhibit enhanced toxicity toward microorganisms due to their large surface area and the generation of reactive oxygen species (ROS), there is a lack of information regarding the ecotoxicological effects of NHs on marine diatom algae, which are an indicator of marine pollution. Moreover, there is scant information on toxicity mechanisms of NHs on aquatic organisms. In this study, four NHs (ie, ZnO-conjugated graphene oxide [GO], ZnO-conjugated carbon nanotubes [CNTs], TiO2 -conjugated GO, and TiO2 -conjugated CNT) that were synthesized by a hydrothermal method were investigated for their toxicity effects on a Thalassiosira pseudonana marine diatom. The in vitro cellular viability and ROS formation employed at the concentration ranges of 50 and 100 mg/L of NHs over 72 hours revealed that ZnO-GO had the most negative effect on T. pseudonana. The primary mechanism identified was the generation of ROS and GO-induced dispersion that caused electrostatic repulsion, preventing aggregation, and an increase in surface areas of NHs. In contrast to GO-induced dispersion, large aggregates were observed in ZnO/TiO2 -conjugated CNT-based NHs. The scanning electron microscopy images suggest that NHs covered algae cells and interacted with them (shading effects); this reduced light availability for photosynthesis. Detailed in vitro toxicity effects and mechanisms that cause high adverse acute toxicity on T. pseudonana were unveiled; this implied concerns about potential hazards of these mechanisms in aquatic ecosystems.


Assuntos
Diatomáceas/efeitos dos fármacos , Grafite/toxicidade , Nanotubos de Carbono/toxicidade , Titânio/toxicidade , Poluentes Químicos da Água/toxicidade , Óxido de Zinco/toxicidade , Organismos Aquáticos/efeitos dos fármacos , Organismos Aquáticos/crescimento & desenvolvimento , Sobrevivência Celular/efeitos dos fármacos , Diatomáceas/crescimento & desenvolvimento , Ecossistema , Grafite/química , Nanotubos de Carbono/química , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Propriedades de Superfície , Titânio/química , Poluentes Químicos da Água/química , Óxido de Zinco/química
16.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-32326569

RESUMO

Amyloid beta (Aß) depositions are more abundant in HIV-infected brains. The blood-brain barrier, with its backbone created by endothelial cells, is assumed to be a core player in Aß homeostasis and may contribute to Aß accumulation in the brain. Exposure to HIV increases shedding of extracellular vesicles (EVs) from human brain endothelial cells and alters EV-Aß levels. EVs carrying various cargo molecules, including a complex set of proteins, can profoundly affect the biology of surrounding neurovascular unit cells. In the current study, we sought to examine how exposure to HIV, alone or together with Aß, affects the surface and total proteomic landscape of brain endothelial EVs. By using this unbiased approach, we gained an unprecedented, high-resolution insight into these changes. Our data suggest that HIV and Aß profoundly remodel the proteome of brain endothelial EVs, altering the pathway networks and functional interactions among proteins. These events may contribute to the EV-mediated amyloid pathology in the HIV-infected brain and may be relevant to HIV-1-associated neurocognitive disorders.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Vesículas Extracelulares/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Proteoma/metabolismo , Peptídeos beta-Amiloides/farmacologia , Barreira Hematoencefálica/metabolismo , Encéfalo/virologia , Células Cultivadas , Cromatografia Líquida , Vesículas Extracelulares/virologia , Ontologia Genética , Células HEK293 , Humanos , Mapas de Interação de Proteínas , Proteômica , Software , Espectrometria de Massas em Tandem
17.
J Biol Chem ; 293(1): 296-311, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29158267

RESUMO

HIV-1 infection and methamphetamine (METH) abuse frequently occur simultaneously and may have synergistic pathological effects. Although HIV-positive/active METH users have been shown to have higher HIV viral loads and experience more severe neurological complications than non-users, the direct impact of METH on HIV infection and its link to the development of neurocognitive alternations are still poorly understood. In the present study, we hypothesized that METH impacts HIV infection of neural progenitor cells (NPCs) by a mechanism encompassing NFκB/SP1-mediated HIV LTR activation. Mouse and human NPCs were infected with EcoHIV (modified HIV virus infectious to mice) and HIV, respectively, in the presence or absence of METH (50 or 100 µm). Pretreatment with METH, but not simultaneous exposure, significantly increased HIV production in both mouse and human NPCs. To determine the mechanisms underlying these effects, cells were transfected with different variants of HIV LTR promoters and then exposed to METH. METH treatment induced transcriptional activity of the HIV LTR promotor, an effect that required both NFκB and SP1 signaling. Pretreatment with METH also decreased neuronal differentiation of HIV-infected NPCs in both in vitro and in vivo settings. Importantly, NPC-derived daughter cells appeared to be latently infected with HIV. This study indicates that METH increases HIV infectivity of NPCs, through the NFκB/SP1-dependent activation of the HIV LTR and with the subsequent alterations of NPC neurogenesis. Such events may underlie METH- exacerbated neurocognitive dysfunction in HIV-infected patients.


Assuntos
Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Metanfetamina/farmacologia , Animais , Linhagem Celular , Repetição Terminal Longa de HIV/efeitos dos fármacos , Humanos , Masculino , Metanfetamina/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição Sp1/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Carga Viral
18.
Brain Behav Immun ; 80: 247-254, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30885840

RESUMO

Methamphetamine (METH) abusers are prone to develop a variety of comorbidities, including cognitive disabilities, and the immunological responses have been recognized as an important component involved in the toxicity of this drug. Cytokines are among the key mediators between systemic inflammatory status and tissue responses. One of these, interleukin 1 (IL-1), has been hypothesized to be involved in cognitive functions and also appears to play a pivotal role among inflammatory molecules. In the present study, we demonstrate that exposure of mice to METH markedly increased the protein level of IL-1ß in hippocampal tissue. Additionally, METH administration induced a decline in spatial learning as determined by the Morris water maze test. We next evaluated the hypothesis that blocking IL-1ß signaling can protect against METH-induced loss of cognitive functioning. The results indicated that METH-induced impaired spatial learning abilities were attenuated by co-administration of mouse IL-1 Trap, a dimeric fusion protein that incorporates the extracellular domains of both of the IL-1 receptor components required for IL-1 signaling (IL-1 receptor type 1 and IL-1 receptor accessory protein), linked to the Fc portion of murine IgG2a. This effect was associated with a decrease in hippocampal IL-1ß level. The current study indicates for the first time that the loss of METH-related cognitive decline can be attenuated by neutralizing IL-1 signaling. Our findings suggest a potential new therapeutic pathway for treatment of altered cognitive abilities that occur in METH abusing individuals.


Assuntos
Estimulantes do Sistema Nervoso Central/administração & dosagem , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Hipocampo/efeitos dos fármacos , Interleucina-1beta/metabolismo , Metanfetamina/administração & dosagem , Animais , Hipocampo/metabolismo , Interleucina-1beta/antagonistas & inibidores , Locomoção/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transdução de Sinais , Aprendizagem Espacial/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos
19.
Mol Pharm ; 16(2): 724-736, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30592424

RESUMO

HIV infection is associated with symptoms of accelerated or accentuated aging that are likely to be driven not only by HIV itself but also by the toxicity of long-term use of antiretroviral drugs. Therefore, it is crucially important to understand the mechanisms by which antiretroviral drugs may contribute to aging. The aim of this study was to investigate the hypothesis that antiretroviral drugs cause increased reactive oxygen species (ROS) generation that results in mitochondrial dysfunction and culminates in promoting cellular senescence. In addition, we applied targeted nanoparticle (NP)-based delivery to specifically enrich mitochondria with coenzyme Q10 (CoQ10) in order to enhance antioxidant protection. The studies employed neural progenitor cells (NPCs), as differentiation of these cells into mature neurons is affected both during HIV infection and in the aging process. Exposure of cultured NPCs to various combinations of HIV antiretroviral therapy (ART) induced a more than 2-fold increase in mitochondrial ROS generation and mitochondrial membrane potential, a more than 50% decrease in oxygen consumption and ATP levels, a 60% decrease in SIRT3 expression, and a 42% decrease in cell proliferation relative to control levels. These alterations were accompanied by a 37% increase in beta-galactosidase staining and a shortening of the telomere length to more than half of the length of controls as assessed by quantitative telomere-FISH labeling, indicating accelerated NPC senescence in response to ART exposure. Importantly, CoQ10 delivered by targeted nanoparticles effectively attenuated these effects. Overall, these results indicate that ART promotes cellular senescence by causing mitochondrial dysfunction, which can be successfully reversed by supplementation with mitochondria-targeted CoQ10.


Assuntos
Antirretrovirais/farmacologia , Senescência Celular/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Ubiquinona/análogos & derivados , Animais , Antioxidantes/metabolismo , Linhagem Celular , Infecções por HIV/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Nanopartículas/química , Células-Tronco Neurais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 3/metabolismo , Ubiquinona/metabolismo
20.
Arterioscler Thromb Vasc Biol ; 38(3): 610-621, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29348119

RESUMO

OBJECTIVE: Cigarette smoke exposure (CSE) is a risk factor for cerebral aneurysm (CA) formation, but the molecular mechanisms are unclear. Although CSE is known to contribute to excess reactive oxygen species generation, the role of oxidative stress on vascular smooth muscle cell (VSMC) phenotypic modulation and pathogenesis of CAs is unknown. The goal of this study was to investigate whether CSE activates a NOX (NADPH oxidase)-dependent pathway leading to VSMC phenotypic modulation and CA formation and rupture. APPROACH AND RESULTS: In cultured cerebral VSMCs, CSE increased expression of NOX1 and reactive oxygen species which preceded upregulation of proinflammatory/matrix remodeling genes (MCP-1, MMPs [matrix metalloproteinase], TNF-α, IL-1ß, NF-κB, KLF4 [Kruppel-like factor 4]) and downregulation of contractile genes (SM-α-actin [smooth muscle α actin], SM-22α [smooth muscle 22α], SM-MHC [smooth muscle myosin heavy chain]) and myocardin. Inhibition of reactive oxygen species production and knockdown of NOX1 with siRNA or antisense decreased CSE-induced upregulation of NOX1 and inflammatory genes and downregulation of VSMC contractile genes and myocardin. p47phox-/- NOX knockout mice, or pretreatment with the NOX inhibitor, apocynin, significantly decreased CA formation and rupture compared with controls. NOX1 protein and mRNA expression were similar in p47phox-/- mice and those pretreated with apocynin but were elevated in unruptured and ruptured CAs. CSE increased CA formation and rupture, which was diminished with apocynin pretreatment. Similarly, NOX1 protein and mRNA and reactive oxygen species were elevated by CSE, and in unruptured and ruptured CAs. CONCLUSIONS: CSE initiates oxidative stress-induced phenotypic modulation of VSMCs and CA formation and rupture. These molecular changes implicate oxidative stress in the pathogenesis of CAs and may provide a potential target for future therapeutic strategies.


Assuntos
Aneurisma Roto/enzimologia , Fumar Cigarros/efeitos adversos , Aneurisma Intracraniano/enzimologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , NADPH Oxidases/metabolismo , Estresse Oxidativo , Fumaça , Acetofenonas/farmacologia , Aneurisma Roto/genética , Aneurisma Roto/patologia , Aneurisma Roto/prevenção & controle , Animais , Antioxidantes/farmacologia , Células Cultivadas , Artérias Cerebrais/enzimologia , Artérias Cerebrais/patologia , Dilatação Patológica , Modelos Animais de Doenças , Aneurisma Intracraniano/genética , Aneurisma Intracraniano/patologia , Aneurisma Intracraniano/prevenção & controle , Fator 4 Semelhante a Kruppel , Masculino , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , NADPH Oxidase 1/genética , NADPH Oxidase 1/metabolismo , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Fenótipo , Ratos Sprague-Dawley , Transdução de Sinais , Remodelação Vascular
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa