Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 740
Filter
1.
Sci Rep ; 14(1): 18002, 2024 08 03.
Article in English | MEDLINE | ID: mdl-39097642

ABSTRACT

Zika virus (ZIKV) infection was first reported in 2015 in Brazil as causing microcephaly and other developmental abnormalities in newborns, leading to the identification of Congenital Zika Syndrome (CZS). Viral infections have been considered an environmental risk factor for neurodevelopmental disorders outcome, such as Autism Spectrum Disorder (ASD). Moreover, not only the infection per se, but maternal immune system activation during pregnancy, has been linked to fetal neurodevelopmental disorders. To understand the impact of ZIKV vertical infection on brain development, we derived induced pluripotent stem cells (iPSC) from Brazilian children born with CZS, some of the patients also being diagnosed with ASD. Comparing iPSC-derived neurons from CZS with a control group, we found lower levels of pre- and postsynaptic proteins and reduced functional synapses by puncta co-localization. Furthermore, neurons and astrocytes derived from the CZS group showed decreased glutamate levels. Additionally, the CZS group exhibited elevated levels of cytokine production, one of which being IL-6, already associated with the ASD phenotype. These preliminary findings suggest that ZIKV vertical infection may cause long-lasting disruptions in brain development during fetal stages, even in the absence of the virus after birth. These disruptions could contribute to neurodevelopmental disorders manifestations such as ASD. Our study contributes with novel knowledge of the CZS outcomes and paves the way for clinical validation and the development of potential interventions to mitigate the impact of ZIKV vertical infection on neurodevelopment.


Subject(s)
Brain , Induced Pluripotent Stem Cells , Infectious Disease Transmission, Vertical , Synapses , Zika Virus Infection , Zika Virus , Humans , Zika Virus Infection/virology , Zika Virus Infection/pathology , Female , Zika Virus/pathogenicity , Synapses/pathology , Synapses/metabolism , Brain/virology , Brain/pathology , Pregnancy , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/virology , Neurons/virology , Neurons/metabolism , Neurons/pathology , Male , Astrocytes/virology , Astrocytes/metabolism , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/pathology , Brazil , Infant, Newborn , Autism Spectrum Disorder/virology , Child
2.
Neurochem Res ; 49(11): 3069-3077, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39120794

ABSTRACT

Autism spectrum disorder (ASD) is known as a group of neurodevelopmental conditions including stereotyped and repetitive behaviors, besides social and sensorimotor deficits. Anatomical and functional evidence indicates atypical maturation of the striatum. Astrocytes regulate the maturation and plasticity of synaptic circuits, and impaired calcium signaling is associated with repetitive behaviors and atypical social interaction. Spontaneous calcium transients (SCT) recorded in the striatal astrocytes of the rat were investigated in the preclinical model of ASD by prenatal exposure to valproic acid (VPA). Our results showed sensorimotor delay, augmented glial fibrillary acidic protein -a typical intermediate filament protein expressed by astrocytes- and diminished expression of GABAA-ρ3 through development, and increased frequency of SCT with a reduced latency that resulted in a diminished amplitude in the VPA model. The convulsant picrotoxin, a GABAA (γ-aminobutyric acid type A) receptor antagonist, reduced the frequency of SCT in both experimental groups but rescued this parameter to control levels in the preclinical ASD model. The amplitude and latency of SCT were decreased by picrotoxin in both experimental groups. Nipecotic acid, a GABA uptake inhibitor, reduced the mean amplitude only for the control group. Nevertheless, nipecotic acid increased the frequency but diminished the latency in both experimental groups. Thus, we conclude that striatal astrocytes exhibit SCT modulated by GABAA-mediated signaling, and prenatal exposure to VPA disturbs this tuning.


Subject(s)
Astrocytes , Corpus Striatum , Animals , Astrocytes/metabolism , Astrocytes/drug effects , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Female , Pregnancy , Rats , Valproic Acid/pharmacology , Rats, Wistar , Picrotoxin/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Disease Models, Animal , Male , Calcium/metabolism , Autism Spectrum Disorder/metabolism , Autistic Disorder/metabolism , Prenatal Exposure Delayed Effects/metabolism
3.
Am J Physiol Regul Integr Comp Physiol ; 327(4): R423-R441, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39102465

ABSTRACT

There is evidence that astrocytes modulate synaptic transmission in the nucleus tractus solitarius (NTS) interacting with glutamatergic and purinergic mechanisms. Here, using in situ working heart-brainstem preparations, we evaluated the involvement of astrocyte and glutamatergic/purinergic neurotransmission in the processing of autonomic and respiratory pathways in the NTS of control and rats exposed to sustained hypoxia (SH). Baseline autonomic and respiratory activities and the responses to chemoreflex activation (KCN) were evaluated before and after microinjections of fluorocitrate (FCt, an astrocyte metabolic inhibitor), kynurenic acid, and pyridoxalphosphate-6-azophenyl-2',4'-disulfonate (PPADS) (nonselective antagonists of glutamatergic and purinergic receptors) into the rostral aspect of the caudal commissural NTS. FCt had no effects on the baseline parameters evaluated but reduced the bradycardic response to chemoreflex activation in SH rats. FCt combined with kynurenic acid and PPADS in control rats reduced the baseline duration of expiration, which was attenuated after SH. FCt produced a large increase in PN frequency discharge in control rats, which was reduced after SH, indicating a reduction in the astrocyte modulation after SH. The data show that 1) the bradycardic component of the peripheral chemoreflex is reduced in SH rats after astrocytes inhibition, 2) the inhibition of astrocytes in the presence of double antagonists in the NTS affects the modulation of baseline duration of expiration in control but not in SH rats, and 3) the autonomic and respiratory responses to chemoreflex activation are mediated by glutamatergic and purinergic receptors in the rostral aspect of the caudal commissural NTS.NEW & NOTEWORTHY Our findings indicate that the neurotransmission of autonomic and respiratory components of the peripheral chemoreflex in the nucleus tractus solitarius (NTS) is mediated by glutamatergic and purinergic mechanisms and reveal a selective involvement of NTS astrocytes in controlling the chemoreflex parasympathetic response in rats exposed to sustained hypoxia (SH) and the baseline duration of expiration mainly in control rats, indicating a selective role for astrocytes modulation in the NTS of control and SH rats.


Subject(s)
Astrocytes , Glutamic Acid , Hypoxia , Receptors, Purinergic , Solitary Nucleus , Synaptic Transmission , Animals , Solitary Nucleus/metabolism , Solitary Nucleus/drug effects , Astrocytes/metabolism , Astrocytes/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Hypoxia/physiopathology , Hypoxia/metabolism , Male , Glutamic Acid/metabolism , Receptors, Purinergic/metabolism , Rats , Rats, Wistar , Kynurenic Acid/pharmacology , Chemoreceptor Cells/metabolism , Chemoreceptor Cells/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Citrates/pharmacology , Time Factors
4.
Neurobiol Dis ; 200: 106612, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39032798

ABSTRACT

Astrocytes play key roles in the brain. When astrocyte support fails, neurological disorders follow, resulting in disrupted synaptic communication, neuronal degeneration, and cell death. We posit that astrocytes overexpressing neurotrophic factors, such as Insulin Like Growth Factor 1 (IGF1), prevent the onset of neurodegeneration. We overexpressed IGF1 and the reporter TdTomato (TOM) in hippocampal astrocytes with bicistronic Adeno-Associated Virus (AAV) harboring the Glial Fibrillary Acidic Protein (GFAP) promoter and afterwards induced neurodegeneration by the intracerebroventricular (ICV) injection of streptozotocin (STZ), a rat model of behavioral impairment, neuroinflammation and shortening of hippocampal astrocytes. We achieved a thorough transgene expression along the hippocampus with a single viral injection. Although species typical behavior was impaired, memory deficit was prevented by IGF1. STZ prompted astrocyte shortening, albeit the length of these cells in animals injected with GFP and IGF1 vectors did not statistically differ from the other groups. In STZ control animals, hippocampal microglial reactive cells increased dramatically, but this was alleviated in IGF1 rats. We conclude that overexpression of IGF1 in astrocytes prevents neurodegeneration onset. Hence, individuals with early neurotrophic exhaustion would be vulnerable to age-related neurodegeneration.


Subject(s)
Astrocytes , Dependovirus , Hippocampus , Insulin-Like Growth Factor I , Animals , Astrocytes/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics , Hippocampus/metabolism , Dependovirus/genetics , Rats , Male , Rats, Wistar , Glial Fibrillary Acidic Protein/metabolism
5.
ACS Biomater Sci Eng ; 10(7): 4279-4296, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38870483

ABSTRACT

After traumatic brain injury, the brain extracellular matrix undergoes structural rearrangement due to changes in matrix composition, activation of proteases, and deposition of chondroitin sulfate proteoglycans by reactive astrocytes to produce the glial scar. These changes lead to a softening of the tissue, where the stiffness of the contusion "core" and peripheral "pericontusional" regions becomes softer than that of healthy tissue. Pioneering mechanotransduction studies have shown that soft substrates upregulate intermediate filament proteins in reactive astrocytes; however, many other aspects of astrocyte biology remain unclear. Here, we developed a platform for the culture of cortical astrocytes using polyacrylamide (PA) gels of varying stiffness (measured in Pascal; Pa) to mimic injury-related regions in order to investigate the effects of tissue stiffness on astrocyte reactivity and morphology. Our results show that substrate stiffness influences astrocyte phenotype; soft 300 Pa substrates led to increased GFAP immunoreactivity, proliferation, and complexity of processes. Intermediate 800 Pa substrates increased Aggrecan+, Brevican+, and Neurocan+ astrocytes. The stiffest 1 kPa substrates led to astrocytes with basal morphologies, similar to a physiological state. These results advance our understanding of astrocyte mechanotransduction processes and provide evidence of how substrates with engineered stiffness can mimic the injury microenvironment.


Subject(s)
Acrylic Resins , Astrocytes , Mechanotransduction, Cellular , Astrocytes/metabolism , Animals , Acrylic Resins/chemistry , Cells, Cultured , Glial Fibrillary Acidic Protein/metabolism , Rats , Gels/chemistry , Cell Proliferation , Rats, Sprague-Dawley
6.
Cells ; 13(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38891053

ABSTRACT

The astrocyte population, around 50% of human brain cells, plays a crucial role in maintaining the overall health and functionality of the central nervous system (CNS). Astrocytes are vital in orchestrating neuronal development by releasing synaptogenic molecules and eliminating excessive synapses. They also modulate neuronal excitability and contribute to CNS homeostasis, promoting neuronal survival by clearance of neurotransmitters, transporting metabolites, and secreting trophic factors. Astrocytes are highly heterogeneous and respond to CNS injuries and diseases through a process known as reactive astrogliosis, which can contribute to both inflammation and its resolution. Recent evidence has revealed remarkable alterations in astrocyte transcriptomes in response to several diseases, identifying at least two distinct phenotypes called A1 or neurotoxic and A2 or neuroprotective astrocytes. However, due to the vast heterogeneity of these cells, it is limited to classify them into only two phenotypes. This review explores the various physiological and pathophysiological roles, potential markers, and pathways that might be activated in different astrocytic phenotypes. Furthermore, we discuss the astrocyte heterogeneity in the main neurodegenerative diseases and identify potential therapeutic strategies. Understanding the underlying mechanisms in the differentiation and imbalance of the astrocytic population will allow the identification of specific biomarkers and timely therapeutic approaches in various neurodegenerative diseases.


Subject(s)
Astrocytes , Neurodegenerative Diseases , Astrocytes/metabolism , Astrocytes/pathology , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Animals , Phenotype
7.
Neurochem Res ; 49(9): 2505-2518, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38886329

ABSTRACT

Sulforaphane is a natural compound with neuroprotective activity, but its effects on hypothalamus remain unknown. In line with this, astrocytes are critical cells to maintain brain homeostasis, and hypothalamic astrocytes are fundamental for sensing and responding to environmental changes involved in a variety of homeostatic functions. Changes in brain functionality, particularly associated with hypothalamic astrocytes, can contribute to age-related neurochemical alterations and, consequently, neurodegenerative diseases. Thus, here, we investigated the glioprotective effects of sulforaphane on hypothalamic astrocyte cultures and hypothalamic cell suspension obtained from aged Wistar rats (24 months old). Sulforaphane showed anti-inflammatory and antioxidant properties, as well as modulated the mRNA expression of astroglial markers, such as aldehyde dehydrogenase 1 family member L1, aquaporin 4, and vascular endothelial growth factor. In addition, it increased the expression and extracellular levels of trophic factors, such as glia-derived neurotrophic factor and nerve growth factor, as well as the release of brain-derived neurotrophic factor and the mRNA of TrkA, which is a receptor associated with trophic factors. Sulforaphane also modulated the expression of classical pathways associated with glioprotection, including nuclear factor erythroid-derived 2-like 2, heme oxygenase-1, nuclear factor kappa B p65 subunit, and AMP-activated protein kinase. Finally, a cell suspension with neurons and glial cells was used to confirm the predominant effect of sulforaphane in glial cells. In summary, this study indicated the anti-aging and glioprotective activities of sulforaphane in aged astrocytes.


Subject(s)
Aging , Astrocytes , Hypothalamus , Isothiocyanates , Neuroprotective Agents , Rats, Wistar , Sulfoxides , Animals , Isothiocyanates/pharmacology , Aging/drug effects , Aging/metabolism , Neuroprotective Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Rats , Male , Cells, Cultured , Antioxidants/pharmacology
8.
Glia ; 72(10): 1746-1765, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38856177

ABSTRACT

Cholesterol is crucial for the proper functioning of eukaryotic cells, especially neurons, which rely on cholesterol to maintain their complex structure and facilitate synaptic transmission. However, brain cells are isolated from peripheral cholesterol by the blood-brain barrier and mature neurons primarily uptake the cholesterol synthesized by astrocytes for proper function. This study aimed to investigate the effect of aging on cholesterol trafficking in astrocytes and its delivery to neurons. We found that aged astrocytes accumulated high levels of cholesterol in the lysosomal compartment, and this cholesterol buildup can be attributed to the simultaneous occurrence of two events: decreased levels of the ABCA1 transporter, which impairs ApoE-cholesterol export from astrocytes, and reduced expression of NPC1, which hinders cholesterol release from lysosomes. We show that these two events are accompanied by increased microR-33 in aged astrocytes, which targets ABCA1 and NPC1. In addition, we demonstrate that the microR-33 increase is triggered by oxidative stress, one of the hallmarks of aging. By coculture experiments, we show that cholesterol accumulation in astrocytes impairs the cholesterol delivery from astrocytes to neurons. Remarkably, we found that this altered transport of cholesterol could be alleviated through treatment with endocannabinoids as well as cannabidiol or CBD. Finally, according to data demonstrating that aged astrocytes develop an A1 phenotype, we found that cholesterol buildup is also observed in reactive C3+ astrocytes. Given that reduced neuronal cholesterol affects synaptic plasticity, the ability of cannabinoids to restore cholesterol transport from aged astrocytes to neurons holds significant implications in aging and inflammation.


Subject(s)
ATP Binding Cassette Transporter 1 , Astrocytes , Cannabinoids , Cholesterol , Lysosomes , Neurons , Astrocytes/metabolism , Astrocytes/drug effects , Animals , Cholesterol/metabolism , Neurons/metabolism , Neurons/drug effects , Lysosomes/metabolism , Lysosomes/drug effects , ATP Binding Cassette Transporter 1/metabolism , Cannabinoids/pharmacology , Cannabinoids/metabolism , Cells, Cultured , Niemann-Pick C1 Protein , Mice , Aging/metabolism , Coculture Techniques , Mice, Inbred C57BL
9.
J Neurochem ; 168(9): 2935-2955, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38943350

ABSTRACT

Astrocytes provide metabolic support to neurons, maintain ionic and water homeostasis, and uptake and recycle neurotransmitters. After exposure to the prototypical PAMP lipopolysaccharide (LPS), reactive astrocytes increase the expression of pro-inflammatory genes, facilitating neurodegeneration. In this study, we analyzed the expression of homeostatic genes in astrocytes exposed to LPS and identified the epigenetic factors contributing to the suppression of homeostatic genes in reactive astrocytes. Primary astrocytic cultures were acutely exposed to LPS and allowed to recover for 24, 72 h, and 7 days. As expected, LPS exposure induced reactive astrogliosis and increased the expression of pro-inflammatory IL-1B and IL-6. Interestingly, the acute exposure resulted in persistent hypermethylation of astroglial DNA. Similar hypermethylation was observed in highly reactive astrocytes from the traumatic brain injury (TBI) penumbra in vivo. Hypermethylation was accompanied by decreased expression of homeostatic genes including LDHA and Scl16a1 (MCT1) both involved in the lactate shuttle to neurons; glutamine synthase (GS) responsible for glutamate processing; Kcnj10 (Kir4.1) important for K+ homeostasis, and the water channel aquaporin-4 (Aqp4). Furthermore, the master regulator of DNA methylation, MAFG-1, as well as DNA methyl transferases DNMT1 and DNMT3a were overexpressed. The downregulation of homeostatic genes correlated with increased methylation of CpG islands in their promoters, as assessed by methylation-sensitive PCR and increased DNMT3a binding to the GS promoter. Treatment with decitabine, a DNMT inhibitor, prevented the LPS- and the HMGB-1-induced downregulation of homeostatic genes. Decitabine treatment also prevented the neurotoxic effects of these astrocytes in primary cortical cultures. In summary, our findings reveal that the pathological remodeling of reactive astrocytes encompasses not only the pro-inflammatory response but, significantly, also entails a long-term suppression of homeostatic gene expression with methylation of crucial CpG islands within their promoters.


Subject(s)
Astrocytes , DNA Methylation , Down-Regulation , Homeostasis , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , DNA Methylation/drug effects , Animals , Homeostasis/drug effects , Down-Regulation/drug effects , Cells, Cultured , Lipopolysaccharides/pharmacology , Male , Mice , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/genetics , Rats , Mice, Inbred C57BL
10.
Br J Pharmacol ; 181(20): 4028-4049, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38936407

ABSTRACT

BACKGROUND AND PURPOSE: Inhibitors of histone deacetylases (iHDACs) are promising drugs for neurodegenerative diseases. We have evaluated the therapeutic potential of the new iHDAC LASSBio-1911 in Aß oligomer (AßO) toxicity models and astrocytes, key players in neuroinflammation and Alzheimer's disease (AD). EXPERIMENTAL APPROACH: Astrocyte phenotype and synapse density were evaluated by flow cytometry, Western blotting, immunofluorescence and qPCR, in vitro and in mice. Cognitive function was evaluated by behavioural assays using a mouse model of intracerebroventricular infusion of AßO. KEY RESULTS: LASSBio-1911 modulates reactivity and synaptogenic potential of cultured astrocytes and improves synaptic markers in cultured neurons and in mice. It prevents AßO-triggered astrocytic reactivity in mice and enhances the neuroprotective potential of astrocytes. LASSBio-1911 improves behavioural performance and rescues synaptic and memory function in AßO-infused mice. CONCLUSION AND IMPLICATIONS: These results contribute to unveiling the mechanisms underlying astrocyte role in AD and provide the rationale for using astrocytes as targets to new drugs for AD.


Subject(s)
Amyloid beta-Peptides , Astrocytes , Cognitive Dysfunction , Histone Deacetylase Inhibitors , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Histone Deacetylase Inhibitors/pharmacology , Mice , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/chemically induced , Male , Mice, Inbred C57BL , Cells, Cultured , Synapses/drug effects , Synapses/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/administration & dosage
11.
Sci Rep ; 14(1): 11435, 2024 05 19.
Article in English | MEDLINE | ID: mdl-38763939

ABSTRACT

Autism spectrum disorder (ASD) exhibits a gender bias, with boys more frequently affected than girls. Similarly, in mouse models induced by prenatal exposure to valproic acid (VPA), males typically display reduced sociability, while females are less affected. Although both males and females exhibit VPA effects on neuroinflammatory parameters, these effects are sex-specific. Notably, females exposed to VPA show increased microglia and astrocyte density during the juvenile period. We hypothesized that these distinct neuroinflammatory patterns contribute to the resilience of females to VPA. To investigate this hypothesis, we treated juvenile animals with intraperitoneal bacterial lipopolysaccharides (LPS), a treatment known to elicit brain neuroinflammation. We thus evaluated the impact of juvenile LPS-induced inflammation on adult sociability and neuroinflammation in female mice prenatally exposed to VPA. Our results demonstrate that VPA-LPS females exhibit social deficits in adulthood, overriding the resilience observed in VPA-saline littermates. Repetitive behavior and anxiety levels were not affected by either treatment. We also evaluated whether the effect on sociability was accompanied by heightened neuroinflammation in the cerebellum and hippocampus. Surprisingly, we observed reduced astrocyte and microglia density in the cerebellum of VPA-LPS animals. These findings shed light on the complex interactions between prenatal insults, juvenile inflammatory stimuli, and sex-specific vulnerability in ASD-related social deficits, providing insights into potential therapeutic interventions for ASD.


Subject(s)
Autism Spectrum Disorder , Lipopolysaccharides , Prenatal Exposure Delayed Effects , Social Behavior , Valproic Acid , Animals , Female , Prenatal Exposure Delayed Effects/chemically induced , Pregnancy , Mice , Valproic Acid/adverse effects , Male , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/etiology , Microglia/drug effects , Microglia/metabolism , Disease Models, Animal , Behavior, Animal/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Mice, Inbred C57BL
12.
Neurochem Res ; 49(7): 1823-1837, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38727985

ABSTRACT

Methylglyoxal (MG) is considered a classical biomarker of diabetes mellitus and its comorbidities. However, a role for this compound in exacerbated immune responses, such as septicemia, is being increasingly observed and requires clarification, particularly in the context of neuroinflammatory responses. Herein, we used two different approaches (in vivo and acute hippocampal slice models) to investigate MG as a biomarker of neuroinflammation and the neuroimmunometabolic shift to glycolysis in lipopolysaccharide (LPS) inflammation models. Our data reinforce the hypothesis that LPS-induced neuroinflammation stimulates the cerebral innate immune response by increasing IL-1ß, a classical pro-inflammatory cytokine, and the astrocyte reactive response, via elevating S100B secretion and GFAP levels. Acute neuroinflammation promotes an early neuroimmunometabolic shift to glycolysis by elevating glucose uptake, lactate release, PFK1, and PK activities. We observed high serum and cerebral MG levels, in association with a reduction in glyoxalase 1 detoxification activity, and a close correlation between serum and hippocampus MG levels with the systemic and neuroinflammatory responses to LPS. Findings strongly suggest a role for MG in immune responses.


Subject(s)
Biomarkers , Hippocampus , Lipopolysaccharides , Neuroinflammatory Diseases , Pyruvaldehyde , Pyruvaldehyde/metabolism , Lipopolysaccharides/pharmacology , Animals , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Biomarkers/metabolism , Male , Hippocampus/metabolism , Hippocampus/drug effects , Rats, Wistar , S100 Calcium Binding Protein beta Subunit/metabolism , Glycolysis/drug effects , Interleukin-1beta/metabolism , Inflammation/metabolism , Inflammation/chemically induced , Glial Fibrillary Acidic Protein/metabolism , Lactoylglutathione Lyase/metabolism , Rats , Astrocytes/metabolism , Astrocytes/drug effects
13.
Neurochem Res ; 49(7): 1851-1862, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38733521

ABSTRACT

Alzheimer's disease (AD) is an age-dependent neurodegenerative disease that is typically sporadic and has a high social and economic cost. We utilized the intracerebroventricular administration of streptozotocin (STZ), an established preclinical model for sporadic AD, to investigate hippocampal astroglial changes during the first 4 weeks post-STZ, a period during which amyloid deposition has yet to occur. Astroglial proteins aquaporin 4 (AQP-4) and connexin-43 (Cx-43) were evaluated, as well as claudins, which are tight junction (TJ) proteins in brain barriers, to try to identify changes in the glymphatic system and brain barrier during the pre-amyloid phase. Glial commitment, glucose hypometabolism and cognitive impairment were characterized during this phase. Astroglial involvement was confirmed by an increase in glial fibrillary acidic protein (GFAP); concurrent proteolysis was also observed, possibly mediated by calpain. Levels of AQP-4 and Cx-43 were elevated in the fourth week post-STZ, possibly accelerating the clearance of extracellular proteins, since these proteins actively participate in the glymphatic system. Moreover, although we did not see a functional disruption of the blood-brain barrier (BBB) at this time, claudin 5 (present in the TJ of the BBB) and claudin 2 (present in the TJ of the blood-cerebrospinal fluid barrier) were reduced. Taken together, data support a role for astrocytes in STZ brain damage, and suggest that astroglial dysfunction accompanies or precedes neuronal damage in AD.


Subject(s)
Alzheimer Disease , Aquaporin 4 , Astrocytes , Streptozocin , Astrocytes/metabolism , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Male , Aquaporin 4/metabolism , Connexin 43/metabolism , Blood-Brain Barrier/metabolism , Water/metabolism , Hippocampus/metabolism , Rats, Wistar , Rats , Disease Models, Animal
14.
J Mater Chem B ; 12(21): 5085-5097, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38713059

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive impairment associated with the accumulation of beta-amyloid protein (Aß). Aß activates glial cells in the brain, increasing the secretion of proinflammatory cytokines, which leads to neuroinflammation and neuronal death. Currently, there are no effective treatments that cure or stop its progression; therefore, AD is considered a global health priority. The main limitations are the low drug bioavailability and impermeability of the blood-brain barrier (BBB). Fortunately, nanomedicine has emerged as a promising field for the development of new nanosystems for the controlled and targeted delivery of drugs to the brain. Therefore, in this work, lipid-polymer hybrid nanoparticles (LPHNPs) conjugated with transferrin (Tf) to facilitate crossing the BBB and loaded with N-acetylcysteine (NAC) for its anti-inflammatory effect were synthesized, and their physicochemical characterization was carried out. Subsequently, an in vitro model involving human astrocytes derived from induced pluripotent stem cells (iPSC) from an AD-diagnosed patient was developed, which was brought to a reactive state by stimulation with lipopolysaccharides (LPSs). The cell culture was treated with either Tf-conjugated LPHNPs loaded with NAC (NAC-Tf-LPHNPs) at 0.25 mg mL-1, or free NAC at 5 mM. The results showed that NAC-Tf-LPHNPs favorably modulated the expression of proinflammatory genes such as interleukin-1ß (IL-1ß), amyloid precursor protein (APP) and glial fibrillary acidic protein (GFAP). In addition, they reduced the secretion of the proinflammatory cytokines interleukin 6 (IL-6), IL-1ß and interferon-gamma (INF-γ). Results for both cases were compared to the group of cells that did not receive any treatment. In contrast, free NAC only had this effect on the expression of IL-1ß and the secretion of the cytokines IL-6 and INF-γ. These results indicate the potential of NAC-Tf-LPHNPs for AD treatment.


Subject(s)
Acetylcysteine , Alzheimer Disease , Astrocytes , Induced Pluripotent Stem Cells , Nanoparticles , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Nanoparticles/chemistry , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Acetylcysteine/chemistry , Acetylcysteine/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Polymers/chemistry , Polymers/pharmacology , Lipids/chemistry , Biomarkers/metabolism , Particle Size , Neuroinflammatory Diseases/drug therapy
15.
J Neuroendocrinol ; 36(10): e13415, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38808481

ABSTRACT

Ischemic stroke is a significant global health issue, ranking fifth among all causes of death and a leading cause of serious long-term disability. Ischemic stroke leads to severe outcomes, including permanent brain damage and neuronal dysfunction. Therefore, decreasing and preventing neuronal injuries caused by stroke has been the focus of therapeutic research. In recent years, many studies have shown that fluctuations in hormonal levels influence the prognosis of ischemic stroke. Thus, it is relevant to understand the role of hormones in the pathophysiological mechanisms of ischemic stroke for preventing and treating this health issue. Here, we investigate the contribution of the prolactin/vasoinhibin axis, an endocrine system regulating blood vessel growth, immune processes, and neuronal survival, to the pathophysiology of ischemic stroke. Male mice with brain overexpression of prolactin or vasoinhibin by adeno-associated virus (AAV) intracerebroventricular injection or lacking the prolactin receptor (Prlr-/-) were exposed to transient middle cerebral artery occlusion (tMCAO) for 45 min followed by 48 h of reperfusion. Overexpression of vasoinhibin or the absence of the prolactin receptor led to an increased lesion volume and decreased survival rates in mice following tMCAO, whereas overexpression of prolactin had no effect. In addition, astrocytic distribution in the penumbra was altered, glial fibrillary acidic protein and S100b mRNA expressions were reduced, and interleukin-6 mRNA expression increased in the ischemic hemisphere of mice overexpressing vasoinhibin. Of note, prolactin receptor-null mice (Prlr-/-) showed a marked increase in serum vasoinhibin levels. Furthermore, vasoinhibin decreased astrocyte numbers in mixed hippocampal neuron-glia cultures. These observations suggest that increased vasoinhibin levels may hinder astrocytes' protective reactivity. Overall, this study suggests the involvement of the prolactin/vasoinhibin axis in the pathophysiology of ischemic stroke-induced brain injury and provides insights into the impact of its dysregulation on astrocyte reactivity and lesion size. Understanding these mechanisms could help develop therapeutic interventions in ischemic stroke and other related neurological disorders.


Subject(s)
Cell Cycle Proteins , Gliosis , Prolactin , Receptors, Prolactin , Animals , Prolactin/metabolism , Male , Mice , Gliosis/pathology , Gliosis/metabolism , Receptors, Prolactin/metabolism , Receptors, Prolactin/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Mice, Inbred C57BL , Astrocytes/metabolism , Astrocytes/pathology , Mice, Knockout , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/metabolism
16.
Neuroreport ; 35(8): 542-550, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38597273

ABSTRACT

Wnt signaling plays an important role in adult brain function, and its dysregulation has been implicated in the loss of neuronal homeostasis. Despite the existence of many studies on the participation of the Wnt pathway in adult neurons, its regulation in astrocytes has been scarcely explored. Several reports point to the presence of Wnt ligands in astrocytes and their possible impact on neuronal plasticity or neuronal death. We aimed to analyze the effect of the neurotransmitter glutamate and the inflammatory cytokine TNFα on the mRNA and protein levels of the canonical Wnt agonist Wnt7a and the antagonist Dkk1 in cultured astrocytes. Primary astrocyte cultures from rat cerebral cortices were exposed to glutamate or TNFα. Wnt7a and Dkk1 expression was analyzed by RT-qPCR and its protein abundance and distribution was assessed by immunofluorescence. We found high basal expression and protein levels of Wnt7a and Dkk1 in unstimulated astrocytes and overproduction of Dkk1 mRNA induced by the two stimuli. These results reveal the astrocytic source of the canonical Wnt ligands Wnt7a and Dkk1, whose levels are differentially regulated by glutamate and TNFα. Astrocytes are a significant source of Wnt ligands, the production of which can be differentially regulated under excitatory or proinflammatory conditions, thereby impacting neuronal function.


Subject(s)
Astrocytes , Glutamic Acid , Intercellular Signaling Peptides and Proteins , Proto-Oncogene Proteins , Tumor Necrosis Factor-alpha , Wnt Proteins , Astrocytes/metabolism , Astrocytes/drug effects , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Glutamic Acid/metabolism , Wnt Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , Rats , RNA, Messenger/metabolism , Rats, Wistar , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/cytology
17.
Toxins (Basel) ; 16(4)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38668612

ABSTRACT

Accidents caused by Bothrops jararaca (Bj) snakes result in several local and systemic manifestations, with pain being a fundamental characteristic. The inflammatory process responsible for hyperalgesia induced by Bj venom (Bjv) has been studied; however, the specific roles played by the peripheral and central nervous systems in this phenomenon remain unclear. To clarify this, we induced hyperalgesia in rats using Bjv and collected tissues from dorsal root ganglia (DRGs) and spinal cord (SC) at 2 and 4 h post-induction. Samples were labeled for Iba-1 (macrophage and microglia), GFAP (satellite cells and astrocytes), EGR1 (neurons), and NK1 receptors. Additionally, we investigated the impact of minocycline, an inhibitor of microglia, and GR82334 antagonist on Bjv-induced hyperalgesia. Our findings reveal an increase in Iba1 in DRG at 2 h and EGR1 at 4 h. In the SC, markers for microglia, astrocytes, neurons, and NK1 receptors exhibited increased expression after 2 h, with EGR1 continuing to rise at 4 h. Minocycline and GR82334 inhibited venom-induced hyperalgesia, highlighting the crucial roles of microglia and NK1 receptors in this phenomenon. Our results suggest that the hyperalgesic effects of Bjv involve the participation of microglial and astrocytic cells, in addition to the activation of NK1 receptors.


Subject(s)
Bothrops , Crotalid Venoms , Ganglia, Spinal , Hyperalgesia , Receptors, Neurokinin-1 , Animals , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Crotalid Venoms/toxicity , Male , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Receptors, Neurokinin-1/metabolism , Minocycline/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Microglia/drug effects , Microglia/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Rats , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Microfilament Proteins/metabolism , Neurokinin-1 Receptor Antagonists/pharmacology , Rats, Sprague-Dawley
18.
J Neurosci Res ; 102(4): e25334, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38656648

ABSTRACT

Iron deficiency (ID) has been shown to affect central nervous system (CNS) development and induce hypomyelination. Previous work from our laboratory in a gestational ID model showed that both oligodendrocyte (OLG) and astrocyte (AST) maturation was impaired. To explore the contribution of AST iron to the myelination process, we generated an in vitro ID model by silencing divalent metal transporter 1 (DMT1) in AST (siDMT1 AST) or treating AST with Fe3+ chelator deferoxamine (DFX; DFX AST). siDMT1 AST showed no changes in proliferation but remained immature. Co-cultures of oligodendrocyte precursors cells (OPC) with siDMT1 AST and OPC cultures incubated with siDMT1 AST-conditioned media (ACM) rendered a reduction in OPC maturation. These findings correlated with a decrease in the expression of AST-secreted factors IGF-1, NRG-1, and LIF, known to promote OPC differentiation. siDMT1 AST also displayed increased mitochondrial number and reduced mitochondrial size as compared to control cells. DFX AST also remained immature and DFX AST-conditioned media also hampered OPC maturation in culture, in keeping with a decrease in the expression of AST-secreted growth factors IGF-1, NRG-1, LIF, and CNTF. DFX AST mitochondrial morphology and number showed results similar to those observed in siDMT1 AST. In sum, our results show that ID, induced through two different methods, impacts AST maturation and mitochondrial functioning, which in turn hampers OPC differentiation.


Subject(s)
Astrocytes , Cell Differentiation , Iron Deficiencies , Oligodendroglia , Astrocytes/metabolism , Astrocytes/drug effects , Oligodendroglia/metabolism , Oligodendroglia/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cation Transport Proteins/metabolism , Coculture Techniques , Culture Media, Conditioned/pharmacology , Rats , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Deferoxamine/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Iron/metabolism
19.
J Neurosci Res ; 102(4): e25336, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38656664

ABSTRACT

Chronic neuroinflammation has been implicated in neurodegenerative disease pathogenesis. A key feature of neuroinflammation is neuronal loss and glial activation, including microglia and astrocytes. 4R-cembranoid (4R) is a natural compound that inhibits hippocampal pro-inflammatory cytokines and increases memory function in mice. We used the lipopolysaccharide (LPS) injection model to study the effect of 4R on neuronal density and microglia and astrocyte activation. C57BL/6J wild-type mice were injected with LPS (5 mg/kg) and 2 h later received either 4R (6 mg/kg) or vehicle. Mice were sacrificed after 72 h for analysis of brain pathology. Confocal images of brain sections immunostained for microglial, astrocyte, and neuronal markers were used to quantify cellular hippocampal phenotypes and neurons. Hippocampal lysates were used to measure the expression levels of neuronal nuclear protein (NeuN), inducible nitrous oxide synthase (iNOS), arginase-1, thrombospondin-1 (THBS1), glial cell-derived neurotrophic factor (GDNF), and orosomucoid-2 (ORM2) by western blot. iNOS and arginase-1 are widely used protein markers of pro- and anti-inflammatory microglia, respectively. GDNF promotes neuronal survival, and ORM2 and THBS1 are astrocytic proteins that regulate synaptic plasticity and inhibit microglial activation. 4R administration significantly reduced neuronal loss and the number of pro-inflammatory microglia 72 h after LPS injection. It also decreased the expression of the pro-inflammatory protein iNOS while increasing arginase-1 expression, supporting its anti-inflammatory role. The protein expression of THBS1, GDNF, and ORM2 was increased by 4R. Our data show that 4R preserves the integrity of hippocampal neurons against LPS-induced neuroinflammation in mice.


Subject(s)
Hippocampus , Lipopolysaccharides , Mice, Inbred C57BL , Neuroglia , Neurons , Animals , Lipopolysaccharides/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Male , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/drug therapy , Phenotype , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology
20.
Biol Res ; 57(1): 19, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689353

ABSTRACT

BACKGROUND: Astrocytes Ca2+ signaling play a central role in the modulation of neuronal function. Activation of metabotropic glutamate receptors (mGluR) by glutamate released during an increase in synaptic activity triggers coordinated Ca2+ signals in astrocytes. Importantly, astrocytes express the Ca2+-dependent nitric oxide (NO)-synthetizing enzymes eNOS and nNOS, which might contribute to the Ca2+ signals by triggering Ca2+ influx or ATP release through the activation of connexin 43 (Cx43) hemichannels, pannexin-1 (Panx-1) channels or Ca2+ homeostasis modulator 1 (CALHM1) channels. Hence, we aim to evaluate the participation of NO in the astrocytic Ca2+ signaling initiated by stimulation of mGluR in primary cultures of astrocytes from rat brain cortex. RESULTS: Astrocytes were stimulated with glutamate or t-ACPD and NO-dependent changes in [Ca2+]i and ATP release were evaluated. In addition, the activity of Cx43 hemichannels, Panx-1 channels and CALHM1 channels was also analyzed. The expression of Cx43, Panx-1 and CALHM1 in astrocytes was confirmed by immunofluorescence analysis and both glutamate and t-ACPD induced NO-mediated activation of CALHM1 channels via direct S-nitrosylation, which was further confirmed by assessing CALHM1-mediated current using the two-electrode voltage clamp technique in Xenopus oocytes. Pharmacological blockade or siRNA-mediated inhibition of CALHM1 expression revealed that the opening of these channels provides a pathway for ATP release and the subsequent purinergic receptor-dependent activation of Cx43 hemichannels and Panx-1 channels, which further contributes to the astrocytic Ca2+ signaling. CONCLUSIONS: Our findings demonstrate that activation of CALHM1 channels through NO-mediated S-nitrosylation in astrocytes in vitro is critical for the generation of glutamate-initiated astrocytic Ca2+ signaling.


Subject(s)
Astrocytes , Calcium Signaling , Nitric Oxide , Animals , Rats , Astrocytes/metabolism , Astrocytes/drug effects , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling/physiology , Calcium Signaling/drug effects , Cells, Cultured , Connexin 43/metabolism , Glutamic Acid/metabolism , Nitric Oxide/metabolism , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL