Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Biol Chem ; 300(1): 105526, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38043797

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica , Reservorios de Enfermedades , Infecciones por VIH , VIH-1 , Infección Latente , Pericitos , Humanos , Barrera Hematoencefálica/virología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/transmisión , Infecciones por VIH/virología , Infección Latente/virología , Pericitos/virología , Proteínas Proto-Oncogénicas c-akt/genética , Calidad de Vida , Latencia del Virus , Reservorios de Enfermedades/virología
2.
Am J Physiol Cell Physiol ; 326(2): C487-C504, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38145295

RESUMEN

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.


Asunto(s)
Infecciones por VIH , VIH-1 , Imidazoles , Accidente Cerebrovascular Isquémico , Sulfóxidos , Humanos , Astrocitos/metabolismo , Inflamasomas/metabolismo , Inflamasomas/farmacología , VIH-1/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Células Endoteliales/metabolismo , Piroptosis , Proteínas NLR/metabolismo , Oxígeno/metabolismo , Isquemia/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Glucosa/metabolismo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/metabolismo
3.
Retrovirology ; 19(1): 27, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36476484

RESUMEN

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.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Células Endoteliales
4.
J Neuroinflammation ; 18(1): 167, 2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34325716

RESUMEN

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.


Asunto(s)
Vasos Sanguíneos/metabolismo , COVID-19/complicaciones , Infecciones por VIH/metabolismo , VIH-1 , Neuronas/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Astrocitos/metabolismo , Encefalopatías/etiología , Células Cultivadas , Endotelio Vascular/metabolismo , Humanos , Microglía/metabolismo , Enfermedades del Sistema Nervioso/etiología , Cultivo Primario de Células , Receptor de Angiotensina Tipo 2 , Replicación Viral
5.
Int J Obes (Lond) ; 42(12): 2036-2046, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29907847

RESUMEN

BACKGROUND/OBJECTIVES: The frequency of metabolic syndrome (MetS) is significantly higher in schizophrenia (SCH) patients, when compared to the general populatiotin. The goal of this study was to evaluate whether genetic variants T-786C (rs2070744), G894T (rs1799983) and C774T (rs1549758) in the endothelial nitric oxide (NOS3) gene and/or their haplotypes could be associated with the risk of MetS in SCH patients or healthy subjects from Russian population. SUBJECTS/METHODS: We performed two case-control comparisons. NOS3 polymorphisms were genotyped in 70 SCH patients with MetS, 190 normal weight SCH patients, 155 MetS patients, and 100 healthy controls. MetS was defined as per the criteria proposed by the International Diabetes Federation (IDF). Anthropometric, clinical, biochemical parameters, and serum nitrite concentrations were measured in all samples. Haplotype frequency estimations and linkage disequilibrium measures were made using Haploview 4.2. RESULTS: The higher C allele (P = 0.009) and lower TT genotype (P = 0.008) frequencies of T-786C polymorphism were found in SCH patients with MetS compared to those in normal weight SCH patients. SCH patients with MetS who were carriers of the T-786C TT genotype had lower serum total cholesterol levels in comparison to the CC genotype (P = 0.016). Furthermore, the 774T/894T haplotype was more frequent in non-SCH individuals with MetS compared to healthy controls (P = 0.0004, odds ratio = 2.18, 95% confidence interval 1.4-3.37). Conversely, the most common haplotype 774C/894G was less frequent in MetS patients than in healthy controls (P = 0.013, odds ratio = 0.61, 95% confidence interval 0.41-0.9). CONCLUSIONS: These results indicate that the NOS3 T-786C promoter polymorphism was closely associated with MetS risk in SCH patients. In addition, the haplotypes composed of G894T and C774T polymorphisms are associated with the MetS susceptibility in Russian population.


Asunto(s)
Síndrome Metabólico , Óxido Nítrico Sintasa de Tipo III/genética , Polimorfismo de Nucleótido Simple/genética , Esquizofrenia , Adulto , Estudios de Casos y Controles , Femenino , Haplotipos/genética , Humanos , Masculino , Síndrome Metabólico/complicaciones , Síndrome Metabólico/epidemiología , Síndrome Metabólico/genética , Persona de Mediana Edad , Esquizofrenia/complicaciones , Esquizofrenia/epidemiología , Esquizofrenia/genética
6.
Res Sq ; 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36824983

RESUMEN

Brain endothelial extracellular vesicles carrying amyloid beta (EV-Aß) can be transferred to neural progenitor cells (NPCs) leading to NPC dysfunction. However, the events involved in this EV-mediated Aß pathology are unclear. EV-proteomics studies identified Serpine-1 (plasminogen activator inhibitor 1, PAI-1) as a major connecting "hub" on several protein-protein interaction maps. Serpine-1 was described as a key player in Aß pathology and was linked to HIV-1 infection as well. Therefore, the aim of this work was to address the hypothesis that Serpine-1 can be transferred via EVs from brain endothelial cells to NPCs and contribute to NPC dysfunction. HBMEC concentrated and released Serpine-1 via EVs, the effect that was potentiated by HIV-1 and Aß. EVs loaded with Serpine-1 were readily taken up by NPCs, and HIV-1 enhanced this event. Interestingly, a highly specific Serpine-1 inhibitor PAI039 increased EV-Aß transfer to NPCs in the presence of HIV-1. PAI039 also partially blocked mitochondrial network morphology and mitochondrial function alterations in the recipient NPCs, which developed mainly after HIV + Aß-EV transfer. PAI039 partly attenuated HIV-EV-mediated decreased synaptic protein levels in NPCs, while increased synaptic protein levels in NPC projections. These findings contribute to a better understanding of the complex mechanisms underlying EV-Serpine-1 related Aß pathology in the context of HIV infection. They are relevant to HIV-1 associated neurocognitive disorders (HAND) in an effort to elucidate the mechanisms of neuropathology in HIV infection.

7.
Mol Neurobiol ; 60(11): 6441-6465, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37458985

RESUMEN

Brain endothelial extracellular vesicles carrying amyloid beta (EV-Aß) can be transferred to neural progenitor cells (NPCs) leading to NPC dysfunction. However, the events involved in this EV-mediated Aß pathology are unclear. EV-proteomics studies identified Serpine-1 (plasminogen activator inhibitor 1, PAI-1) as a major connecting "hub" on several protein-protein interaction maps. Serpine-1 was described as a key player in Aß pathology and was linked to HIV-1 infection as well. Therefore, the aim of this work was to address the hypothesis that Serpine-1 can be transferred via EVs from brain endothelial cells (HBMEC) to NPCs and contribute to NPC dysfunction. HBMEC concentrated and released Serpine-1 via EVs, the effect that was potentiated by HIV-1 and Aß. EVs loaded with Serpine-1 were readily taken up by NPCs, and HIV-1 enhanced this event. Interestingly, a highly specific Serpine-1 inhibitor PAI039 increased EV-Aß transfer to NPCs in the presence of HIV-1. PAI039 also partially blocked mitochondrial network morphology alterations in the recipient NPCs, which developed mainly after HIV + Aß-EV transfer. PAI039 partly attenuated HIV-EV-mediated decreased synaptic protein levels in NPCs, while increased synaptic protein levels in NPC projections. These findings contribute to a better understanding of the complex mechanisms underlying EV-Serpine-1 related Aß pathology in the context of HIV infection. They are relevant to HIV-1 associated neurocognitive disorders (HAND) in an effort to elucidate the mechanisms of neuropathology in HIV infection.


Asunto(s)
Vesículas Extracelulares , Infecciones por VIH , VIH-1 , Células-Madre Neurales , Humanos , Péptidos beta-Amiloides/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Células Endoteliales/metabolismo , Células-Madre Neurales/metabolismo , Vesículas Extracelulares/metabolismo
8.
Res Sq ; 2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36778388

RESUMEN

HIV-1-associated blood brain barrier (BBB) alterations and neurocognitive disorders are frequent clinical manifestations in HIV-1 infected patients. The BBB is formed by cells of the neurovascular unit (NVU) and sealed together by tight junction (TJ) proteins, such as occludin (ocln). Pericytes are a key cell type of NVU that can harbor HIV-1 infection via a mechanism that is regulated, at least in part, by ocln. After viral infection, the immune system starts the production of interferons, which induce the expression of the 2'-5'-oligoadenylate synthetase (OAS) family of interferon stimulated genes and activate the endoribonuclease RNaseL that provides antiviral protection by viral RNA degradation. The current study evaluated the involvement of the OAS genes in HIV-1 infection of cells of NVU and the role of ocln in controlling OAS antiviral signaling pathway. We identified that ocln modulates the expression levels of the OAS1, OAS2, OAS3, and OASL genes and proteins and, in turn, that the members of the OAS family can influence HIV replication in human brain pericytes. Mechanistically, this effect was regulated via the STAT signaling. HIV-1 infection of pericytes significantly upregulated expression of all OAS genes at the mRNA level but selectively OAS1, OAS2 and OAS3 at the protein level. Interestingly no changes were found in RNaseL after HIV-1 infection. Overall, these results contribute to a better understanding of the molecular mechanisms implicated in the regulation of HIV-1 infection in human brain pericytes and suggest a novel role for ocln in controlling of this process.

9.
Eur Neuropsychopharmacol ; 67: 66-79, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36513018

RESUMEN

The neuronal isoform of nitric oxide synthase (nNOS) and its interacting protein NOS1AP have been linked to several mental disorders including schizophrenia and depression. An increase in the interaction between nNOS and NOS1AP in the frontal cortex has been suggested to contribute to the emergence of these disorders. Here we aimed to uncover whether disruption of their interactions in the frontal cortex leads to mental disorder endophenotypes. Targeting the medial prefrontal cortex (mPFC), we stereotaxically injected wild-type C57BL/6J mice with recombinant adeno-associated virus (rAAV) expressing either full-length NOS1AP, the nNOS binding region of NOS1AP (i.e. NOS1AP396-503), or the nNOS amino-terminus (i.e. nNOS1-133), which was shown to disrupt the interaction of endogenous nNOS with PSD-95. We tested these mice in a comprehensive behavioural battery, assessing different endophenotypes related to mental disorders. We found no differences in anxiety-related and exploratory behaviours. Likewise, social interaction was comparable in all groups. However, social recognition was impaired in NOS1AP and NOS1AP396-503 mice. These mice, as well as mice overexpressing nNOS1-133 also displayed impaired spatial working memory (SWM) capacity, while spatial reference memory (SRM) remained intact. Finally, mice overexpressing NOS1AP and nNOS1-133, but not NOS1AP396-503, failed to habituate to the startling pulses in an acoustic startle response (ASR) paradigm, though we found no difference in overall startle intensity or prepulse inhibition (PPI) of the ASR. Our findings indicate a distinct role of NOS1AP/nNOS/PSD-95 interactions in the mPFC to contribute to specific endophenotypic changes observed in different mental disorders.


Asunto(s)
Memoria a Corto Plazo , Reflejo de Sobresalto , Ratones , Animales , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo I/genética , Reconocimiento en Psicología , Trastornos de la Memoria , Homólogo 4 de la Proteína Discs Large/metabolismo , Corteza Prefrontal/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética
10.
Trends Neurosci ; 46(8): 682-693, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37330380

RESUMEN

HIV-associated comorbidities, such as ischemic stroke, are prevalent in people with HIV (PWH). Several studies both in animal models and humans have revealed an association between activation of the inflammasome in HIV-1 infection and stroke. The gut microbiota is an important component in controlling neuroinflammation in the CNS. It has also been proposed to be involved in the pathobiology of HIV-1 infection, and has been associated with an increase in activation of the inflammasome. In this review, we provide an overview of the microbiota-gut-inflammasome-brain axis, focusing on the NLRP3 inflammasome and dysregulation of the microbiome as risk factors that may contribute to the outcome of ischemic stroke and recovery in PWH. We also focus on the potential of targeting the NLRP3 inflammasome as a novel therapeutic approach for PWH who are at risk of developing cerebrovascular diseases.


Asunto(s)
Infecciones por VIH , VIH-1 , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Humanos , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Accidente Cerebrovascular Isquémico/complicaciones , Disbiosis/complicaciones , Accidente Cerebrovascular/complicaciones , Infecciones por VIH/complicaciones
11.
Mol Neurobiol ; 60(9): 4966-4982, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37209263

RESUMEN

HIV-1-associated blood brain barrier (BBB) alterations and neurocognitive disorders are frequent clinical manifestations in HIV-1 infected patients. The BBB is formed by cells of the neurovascular unit (NVU) and sealed together by tight junction proteins, such as occludin (ocln). Pericytes are a key cell type of NVU that can harbor HIV-1 infection via a mechanism that is regulated, at least in part, by ocln. After viral infection, the immune system starts the production of interferons, which induce the expression of the 2'-5'-oligoadenylate synthetase (OAS) family of interferon stimulated genes and activate the endoribonuclease RNaseL that provides antiviral protection by viral RNA degradation. The current study evaluated the involvement of the OAS genes in HIV-1 infection of cells of NVU and the role of ocln in controlling OAS antiviral signaling pathway. We identified that ocln modulates the expression levels of the OAS1, OAS2, OAS3, and OASL genes and proteins and, in turn, that the members of the OAS family can influence HIV replication in human brain pericytes. Mechanistically, this effect was regulated via the STAT signaling. HIV-1 infection of pericytes significantly upregulated expression of all OAS genes at the mRNA level but selectively OAS1, OAS2, and OAS3 at the protein level. Interestingly no changes were found in RNaseL after HIV-1 infection. Overall, these results contribute to a better understanding of the molecular mechanisms implicated in the regulation of HIV-1 infection in human brain pericytes and suggest a novel role for ocln in controlling of this process.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Interferones , Ocludina/genética , VIH-1/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , Infecciones por VIH/genética , Antivirales
12.
Elife ; 122023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37725078

RESUMEN

Development of the nervous system depends on signaling centers - specialized cellular populations that produce secreted molecules to regulate neurogenesis in the neighboring neuroepithelium. In some cases, signaling center cells also differentiate to produce key types of neurons. The formation of a signaling center involves its induction, the maintenance of expression of its secreted molecules, and cell differentiation and migration events. How these distinct processes are coordinated during signaling center development remains unknown. By performing studies in mice, we show that Lmx1a acts as a master regulator to orchestrate the formation and function of the cortical hem (CH), a critical signaling center that controls hippocampus development. Lmx1a co-regulates CH induction, its Wnt signaling, and the differentiation and migration of CH-derived Cajal-Retzius neurons. Combining RNAseq, genetic, and rescue experiments, we identified major downstream genes that mediate distinct Lmx1a-dependent processes. Our work revealed that signaling centers in the mammalian brain employ master regulatory genes and established a framework for analyzing signaling center development.


Asunto(s)
Neurogénesis , Neuronas , Animales , Ratones , Transporte Biológico , Diferenciación Celular , Mamíferos , Neurogénesis/genética , Vía de Señalización Wnt
13.
Fluids Barriers CNS ; 20(1): 73, 2023 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-37840143

RESUMEN

Compromised structure and function of the blood-brain barrier (BBB) is one of the pathological hallmarks of brain infection by HIV-1. BBB damage during HIV-1 infection has been associated with modified expression of tight junction (TJ) proteins, including occludin. Recent evidence indicated occludin as a redox-sensitive, multifunctional protein that can act as both an NADH oxidase and influence cellular metabolism through AMPK kinase. One of the newly identified functions of occludin is its involvement in regulating HIV-1 infection. Studies suggest that occludin expression levels and the rate of HIV-1 infection share a reverse, bidirectional relationship; however, the mechanisms of this relationship are unclear. In this review, we describe the pathways involved in the regulation of HIV-1 infection by occludin. We propose that occludin may serve as a potential therapeutic target to control HIV-1 infection and to improve the lives of people living with HIV-1.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , Ocludina/metabolismo , VIH-1/metabolismo , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo
14.
J Vis Exp ; (188)2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36282703

RESUMEN

Ischemic stroke is a major cause of death and disability worldwide with limited therapeutic options. The neuropathology of ischemic stroke is characterized by an interruption in blood supply to the brain leading to cell death and cognitive dysfunction. During and after ischemic stroke, blood-brain barrier (BBB) dysfunction facilitates injury progression and contributes to poor patient recovery. Current BBB models primarily include endothelial monocultures and double co-cultures with either astrocytes or pericytes. Such models lack the ability to fully imitate a dynamic brain microenvironment, which is essential for cell-to-cell communication. Additionally, commonly used BBB models often contain immortalized human endothelial cells or animal-derived (rodent, porcine, or bovine) cell cultures that pose translational limitations. This paper describes a novel well-insert-based BBB model containing only primary human cells (brain microvascular endothelial cells, astrocytes, and brain vascular pericytes) enabling the investigation of ischemic brain injury in vitro. The effects of oxygen-glucose deprivation (OGD) on barrier integrity were assessed by passive permeability, transendothelial electrical resistance (TEER) measurements,and direct visualization of hypoxic cells. The presented protocol offers a distinct advantage inmimicking the intercellular environment of the BBB in vivo, serving as a more realistic in vitro BBB model for developing new therapeutic strategies in the setting of ischemic brain injury.


Asunto(s)
Lesiones Encefálicas , Accidente Cerebrovascular Isquémico , Humanos , Animales , Bovinos , Porcinos , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Cultivo Primario de Células , Técnicas de Cocultivo , Astrocitos/metabolismo , Oxígeno/metabolismo , Glucosa/metabolismo , Lesiones Encefálicas/patología
15.
Viruses ; 13(9)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34578464

RESUMEN

The neurovascular units (NVU) are the minimal functional units of the blood-brain barrier (BBB), composed of endothelial cells, pericytes, astrocytes, microglia, neurons, and the basement membrane. The BBB serves as an important interface for immune communication between the brain and peripheral circulation. Disruption of the NVU by the human immunodeficiency virus-1 (HIV-1) induces dysfunction of the BBB and triggers inflammatory responses, which can lead to the development of neurocognitive impairments collectively known as HIV-1-associated neurocognitive disorders (HAND). Methamphetamine (METH) use disorder is a frequent comorbidity among individuals infected with HIV-1. METH use may be associated not only with rapid HIV-1 disease progression but also with accelerated onset and increased severity of HAND. However, the molecular mechanisms of METH-induced neuronal injury and cognitive impairment in the context of HIV-1 infection are poorly understood. In this review, we summarize recent progress in the signaling pathways mediating synergistic impairment of the BBB and neuronal injury induced by METH and HIV-1, potentially accelerating the onset or severity of HAND in HIV-1-positive METH abusers. We also discuss potential therapies to limit neuroinflammation and NVU damage in HIV-1-infected METH abusers.


Asunto(s)
Complejo SIDA Demencia/fisiopatología , Trastornos Relacionados con Anfetaminas/complicaciones , Barrera Hematoencefálica/fisiopatología , Infecciones por VIH/complicaciones , Metanfetamina , Trastornos Neurocognitivos/fisiopatología , Trastornos Relacionados con Anfetaminas/fisiopatología , Animales , Comorbilidad , Infecciones por VIH/fisiopatología , Humanos , Procesos Mentales , Enfermedades Neuroinflamatorias/fisiopatología
16.
Exp Neurol ; 336: 113537, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33259808

RESUMEN

Intrauterine growth restriction (IUGR) affects ~10% of human pregnancies, results in infants born small for gestational age (SGA), and is associated with motor and cognitive deficits. Human studies suggest that some deficits in SGA patients originate in the cerebellum, a major motor-coordination and cognitive center, but the underlying mechanisms remain unknown. To identify the cerebellar developmental program affected by IUGR, we analyzed the pig as a translational animal model in which some fetuses spontaneously develop IUGR due to early-onset chronic placental insufficiency. Similar to humans, SGA pigs revealed small cerebella, which contained fewer mature granule cells (GCs) in the internal granule cell layer (IGL). Surprisingly, newborn SGA pigs had increased proliferation of GC precursors in the external granule cell layer (EGL), which was associated with an increased density of Purkinje cells, known to non-autonomously promote the proliferation of GCs. However, the GCs of SGA pigs did not properly initiate exit from the EGL to IGL, which was associated with a decreased density of guiding Bergmann glial fibers, reduced expression of pro-migratory genes Pard3a, JamC and Sema6a, and increased apoptosis. While proliferation spontaneously normalized during postnatal development, accumulation of pre-migratory GCs and apoptosis in the EGL were long-lasting consequences of IUGR. Using organotypic cerebellar slice cultures, we showed that normalizing expression of Pard3a and JamC, which operate in the same molecular pathway in GCs, was sufficient to rescue both migratory and, at a later time point, apoptotic defects of IUGR. Thus, a decreased exit of GCs from the EGL, due to disrupted Pard3a/JamC radial migration initiation pathway, is a major mechanism of IUGR-related cerebellar pathology.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Retardo del Crecimiento Fetal/patología , Transducción de Señal/genética , Animales , Animales Recién Nacidos , Apoptosis , Recuento de Células , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Cerebelo/patología , Gránulos Citoplasmáticos , Femenino , Embarazo , Células de Purkinje/patología , Porcinos
17.
Neuroscience ; 452: 247-264, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33246067

RESUMEN

The inner ear, projections, and brainstem nuclei are essential components of the auditory and vestibular systems. It is believed that the evolution of complex systems depends on duplicated sets of genes. The contribution of duplicated genes to auditory or vestibular system development, however, is poorly understood. We describe that Lmx1a and Lmx1b, which originate from the invertebrate Lmx1b-like gene, redundantly regulate development of multiple essential components of the mammalian auditory/vestibular systems. Combined, but not individual, loss of Lmx1a/b eliminated the auditory inner ear organ of Corti (OC) and disrupted the spiral ganglion, which was preceded by a diminished expression of their critical regulator Pax2. Innervation of the remaining inner ear vestibular organs revealed unusual sizes or shapes and was more affected compared to Lmx1a/b single-gene mutants. Individual loss of Lmx1a/b genes did not disrupt brainstem auditory nuclei or inner ear central projections. Combined loss of Lmx1a/b, however, eliminated excitatory neurons in cochlear/vestibular nuclei, and also the expression of a master regulator Atoh1 in their progenitors in the lower rhombic lip (RL). Finally, in Lmx1a/b double mutants, vestibular afferents aberrantly projected to the roof plate. This phenotype was associated with altered expression of Wnt3a, a secreted ligand of the Wnt pathway that regulates pathfinding of inner ear projections. Thus, Lmx1a/b are redundantly required for the development of the mammalian inner ear, inner ear central projections, and cochlear/vestibular nuclei.


Asunto(s)
Órgano Espiral , Factores de Transcripción , Animales , Cóclea , Proteínas con Homeodominio LIM/genética , Ganglio Espiral de la Cóclea , Factores de Transcripción/genética
18.
Antioxidants (Basel) ; 10(1)2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467773

RESUMEN

Inflammation and an increase in antioxidant responses mediated by oxidative stress play an important role in the pathogenesis of acute liver injury (ALI). We utilized in silico prediction of biological activity spectra for substances (PASS) analysis to estimate the potential biological activity profile of deethylated ethoxyquin (DEQ) and hypothesized that DEQ exhibits antioxidant and anti-inflammatory effects in a rat model of carbon tetrachloride (CCl4)-induced ALI. Our results demonstrate that DEQ improved liver function which was indicated by the reduction of histopathological liver changes. Treatment with DEQ reduced CCl4-induced elevation of gene expression, and the activity of antioxidant enzymes (AEs), as well as the expression of transcription factors Nfe2l2 and Nfkb2. Furthermore, DEQ treatment inhibited apoptosis, downregulated gene expression of pro-inflammatory cytokines (Tnf and Il6), cyclooxygenase 2 (Ptgs2), decreased glutathione (GSH) level and myeloperoxidase (MPO) activity in rats with ALI. Notably, DEQ treatment led to an inhibition of CCl4-induced NLRP3-inflammasome activation which was indicated by the reduced protein expression of IL-1ß, caspase-1, and NLRP3 in the liver. Our data suggest that DEQ has a hepatoprotective effect mediated by redox-homeostasis regulation, NLRP3 inflammasome, and apoptosis inhibition, which makes that compound a promising candidate for future clinical studies.

19.
EBioMedicine ; 71: 103565, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34455393

RESUMEN

BACKGROUND: Nitric oxide synthase 1 adaptor protein (NOS1AP; previously named CAPON) is linked to the glutamatergic postsynaptic density through interaction with neuronal nitric oxide synthase (nNOS). NOS1AP and its interaction with nNOS have been associated with several mental disorders. Despite the high levels of NOS1AP expression in the hippocampus and the relevance of this brain region in glutamatergic signalling as well as mental disorders, a potential role of hippocampal NOS1AP in the pathophysiology of these disorders has not been investigated yet. METHODS: To uncover the function of NOS1AP in hippocampus, we made use of recombinant adeno-associated viruses to overexpress murine full-length NOS1AP or the NOS1AP carboxyterminus in the hippocampus of mice. We investigated these mice for changes in gene expression, neuronal morphology, and relevant behavioural phenotypes. FINDINGS: We found that hippocampal overexpression of NOS1AP markedly increased the interaction of nNOS with PSD-95, reduced dendritic spine density, and changed dendritic spine morphology at CA1 synapses. At the behavioural level, we observed an impairment in social memory and decreased spatial working memory capacity. INTERPRETATION: Our data provide a mechanistic explanation for a highly selective and specific contribution of hippocampal NOS1AP and its interaction with the glutamatergic postsynaptic density to cross-disorder pathophysiology. Our findings allude to therapeutic relevance due to the druggability of this molecule. FUNDING: This study was funded in part by the DFG, the BMBF, the Academy of Finland, the NIH, the Japanese Society of Clinical Neuropsychopharmacology, the Ministry of Education of the Russian Federation, and the European Community.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Endofenotipos , Expresión Génica , Hipocampo/metabolismo , Trastornos Mentales/etiología , Trastornos Mentales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Homólogo 4 de la Proteína Discs Large/metabolismo , Regulación de la Expresión Génica , Hipocampo/fisiopatología , Masculino , Trastornos Mentales/diagnóstico , Ratones , Óxido Nítrico Sintasa de Tipo I/metabolismo , Unión Proteica , Transducción de Señal
20.
Nat Commun ; 10(1): 5192, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31729356

RESUMEN

The extent of neocortical gyrification is an important determinant of a species' cognitive abilities, yet the mechanisms regulating cortical gyrification are poorly understood. We uncover long-range regulation of this process originating at the telencephalic dorsal midline, where levels of secreted Bmps are maintained by factors in both the neuroepithelium and the overlying mesenchyme. In the mouse, the combined loss of transcription factors Lmx1a and Lmx1b, selectively expressed in the midline neuroepithelium and the mesenchyme respectively, causes dorsal midline Bmp signaling to drop at early neural tube stages. This alters the spatial and temporal Wnt signaling profile of the dorsal midline cortical hem, which in turn causes gyrification of the distal neocortex. Our study uncovers early mesenchymal-neuroepithelial interactions that have long-range effects on neocortical gyrification and shows that lissencephaly in mice is actively maintained via redundant genetic regulation of dorsal midline development and signaling.


Asunto(s)
Mesodermo/embriología , Neocórtex/embriología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neocórtex/metabolismo , Células Neuroepiteliales/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA