Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.711
Filtrar
1.
Cells ; 13(9)2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38727321

RESUMO

Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by deficiency of the survival motor neuron (SMN) protein. Although SMA is a genetic disease, environmental factors contribute to disease progression. Common pathogen components such as lipopolysaccharides (LPS) are considered significant contributors to inflammation and have been associated with muscle atrophy, which is considered a hallmark of SMA. In this study, we used the SMNΔ7 experimental mouse model of SMA to scrutinize the effect of systemic LPS administration, a strong pro-inflammatory stimulus, on disease outcome. Systemic LPS administration promoted a reduction in SMN expression levels in CNS, peripheral lymphoid organs, and skeletal muscles. Moreover, peripheral tissues were more vulnerable to LPS-induced damage compared to CNS tissues. Furthermore, systemic LPS administration resulted in a profound increase in microglia and astrocytes with reactive phenotypes in the CNS of SMNΔ7 mice. In conclusion, we hereby show for the first time that systemic LPS administration, although it may not precipitate alterations in terms of deficits of motor functions in a mouse model of SMA, it may, however, lead to a reduction in the SMN protein expression levels in the skeletal muscles and the CNS, thus promoting synapse damage and glial cells' reactive phenotype.


Assuntos
Modelos Animais de Doenças , Lipopolissacarídeos , Atrofia Muscular Espinal , Animais , Lipopolissacarídeos/farmacologia , Atrofia Muscular Espinal/patologia , Atrofia Muscular Espinal/metabolismo , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/metabolismo , Microglia/metabolismo , Microglia/efeitos dos fármacos , Microglia/patologia , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Camundongos Endogâmicos C57BL , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Inflamação/patologia
2.
CNS Neurosci Ther ; 30(5): e14726, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38715251

RESUMO

AIMS: The preoptic area (POA) of the hypothalamus, crucial in thermoregulation, has long been implicated in the pain process. However, whether nociceptive stimulation affects body temperature and its mechanism remains poorly studied. METHODS: We used capsaicin, formalin, and surgery to induce acute nociceptive stimulation and monitored rectal temperature. Optical fiber recording, chemical genetics, confocal imaging, and pharmacology assays were employed to confirm the role and interaction of POA astrocytes and extracellular adenosine. Immunofluorescence was utilized for further validation. RESULTS: Acute nociception could activate POA astrocytes and induce a decrease in body temperature. Manipulation of astrocytes allowed bidirectional control of body temperature. Furthermore, acute nociception and astrocyte activation led to increased extracellular adenosine concentration within the POA. Activation of adenosine A1 or A2A receptors contributed to decreased body temperature, while inhibition of these receptors mitigated the thermo-lowering effect of astrocytes. CONCLUSION: Our results elucidate the interplay between acute nociception and thermoregulation, specifically highlighting POA astrocyte activation. This enriches our understanding of physiological responses to painful stimuli and contributes to the analysis of the anatomical basis involved in the process.


Assuntos
Astrócitos , Hipotermia , Nociceptividade , Área Pré-Óptica , Animais , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Nociceptividade/fisiologia , Hipotermia/induzido quimicamente , Masculino , Camundongos , Receptores Purinérgicos P1/metabolismo , Camundongos Endogâmicos C57BL , Adenosina/metabolismo , Capsaicina/farmacologia , Formaldeído/toxicidade , Formaldeído/farmacologia
3.
CNS Neurosci Ther ; 30(5): e14740, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38715318

RESUMO

AIMS: γ-aminobutyric acid (GABA) from reactive astrocytes is critical for the dysregulation of neuronal activity in various neuroinflammatory conditions. While Scutellaria baicalensis Georgi (S. baicalensis) is known for its efficacy in addressing neurological symptoms, its potential to reduce GABA synthesis in reactive astrocytes and the associated neuronal suppression remains unclear. This study focuses on the inhibitory action of monoamine oxidase B (MAO-B), the key enzyme for astrocytic GABA synthesis. METHODS: Using a lipopolysaccharide (LPS)-induced neuroinflammation mouse model, we conducted immunohistochemistry to assess the effect of S. baicalensis on astrocyte reactivity and its GABA synthesis. High-performance liquid chromatography was performed to reveal the major compounds of S. baicalensis, the effects of which on MAO-B inhibition, astrocyte reactivity, and tonic inhibition in hippocampal neurons were validated by MAO-B activity assay, qRT-PCR, and whole-cell patch-clamp. RESULTS: The ethanolic extract of S. baicalensis ameliorated astrocyte reactivity and reduced excessive astrocytic GABA content in the CA1 hippocampus. Baicalin and baicalein exhibited significant MAO-B inhibition potential. These two compounds downregulate the mRNA levels of genes associated with reactive astrogliosis or astrocytic GABA synthesis. Additionally, LPS-induced aberrant tonic inhibition was reversed by both S. baicalensis extract and its key compounds. CONCLUSIONS: In summary, baicalin and baicalein isolated from S. baicalensis reduce astrocyte reactivity and alleviate aberrant tonic inhibition of hippocampal neurons during neuroinflammation.


Assuntos
Astrócitos , Flavanonas , Flavonoides , Lipopolissacarídeos , Neurônios , Extratos Vegetais , Scutellaria baicalensis , Ácido gama-Aminobutírico , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Flavanonas/farmacologia , Scutellaria baicalensis/química , Camundongos , Ácido gama-Aminobutírico/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Masculino , Flavonoides/farmacologia , Extratos Vegetais/farmacologia , Lipopolissacarídeos/toxicidade , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Monoaminoxidase/metabolismo , Inibição Neural/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo
4.
Sci Rep ; 14(1): 10877, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38740862

RESUMO

In chronic stages of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalitis (EAE), connexin (Cx)43 gap junction channel proteins are overexpressed because of astrogliosis. To elucidate the role of increased Cx43, the central nervous system (CNS)-permeable Cx blocker INI-0602 was therapeutically administered. C57BL6 mice with chronic EAE initiated by MOG35-55 received INI-0602 (40 mg/kg) or saline intraperitoneally every other day from days post-immunization (dpi) 17-50. Primary astroglia were employed to observe calcein efflux responses. In INI-0602-treated mice, EAE clinical signs improved significantly in the chronic phase, with reduced demyelination and decreased CD3+ T cells, Iba-1+ and F4/80+ microglia/macrophages, and C3+GFAP+ reactive astroglia infiltration in spinal cord lesions. Flow cytometry analysis of CD4+ T cells from CNS tissues revealed significantly reduced Th17 and Th17/Th1 cells (dpi 24) and Th1 cells (dpi 50). Multiplex array of cerebrospinal fluid showed significantly suppressed IL-6 and significantly increased IL-10 on dpi 24 in INI-0602-treated mice, and significantly suppressed IFN-γ and MCP-1 on dpi 50 in the same group. In vitro INI-0602 treatment inhibited ATP-induced calcium propagations of Cx43+/+ astroglial cells to similar levels of those of Cx43-/- cells. Astroglial Cx43 hemichannels represent a novel therapeutic target for chronic EAE and MS.


Assuntos
Astrócitos , Conexina 43 , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental , Camundongos Endogâmicos C57BL , Esclerose Múltipla , Animais , Conexina 43/metabolismo , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Camundongos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Feminino
5.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 636-643, 2024 Apr 20.
Artigo em Chinês | MEDLINE | ID: mdl-38708495

RESUMO

OBJECTIVE: To investigate the effect of Jisuikang formula-medicated serum for promoting spinal cord injury (SCI) repair in rats and explore the possible mechanism. METHODS: Thirty adult SD rats were randomized into sham-operated group, SCI (induced using a modified Allen method) model group, and Jisuikang formula-medicated serum treatment group. After the operations, the rats were treated with normal saline or Jisuikang by gavage on a daily basis for 14 days, and the changes in hindlimb motor function of the rats was assessed with Basso-Beattie-Bresnahan (BBB) scores and inclined-plate test. The injured spinal cord tissues were sampled from the SCI rat models for single-cell RNA sequencing, and bioinformatics analysis was performed to identify the target genes of Jisuikang, spinal cord injury and glycolysis. In the cell experiment, cultured astrocytes from neonatal SD rat cortex were treated with SOX2 alone or in combination with Jisuikang-medicated serum for 21 days, and the protein expressions of PKM2, p-PKM2 and YAP and colocalization of PKM2 and YAP in the cells were analyzed with Western blotting and immunofluorescence staining, respectively. RESULTS: The SCI rats with Jisuikang treatment showed significantly improved BBB scores and performance in inclined-plate test. At the injury site, high PKM2 expression was detected in various cell types. Bioinformatic analysis identified the HIPPO-YAP signaling pathway as the target pathway of Jisuikang. In cultured astrocytes, SOX2 combined with the mediated serum, as compared with SOX2 alone, significantly increased PKM2, p-PKM2 and YAP expressions and entry of phosphorylated PKM2 into the nucleus, and promoted PKM2 and YAP co-localization in the cells. CONCLUSION: Jisuikang formula accelerates SCI repair in rats possibly by promoting aerobic glycolysis of the astrocytes via activating the PKM2/YAP axis to induce reprogramming of the astrocytes into neurons.


Assuntos
Astrócitos , Piruvato Quinase , Ratos Sprague-Dawley , Transdução de Sinais , Traumatismos da Medula Espinal , Proteínas de Sinalização YAP , Animais , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Ratos , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas de Ligação a Hormônio da Tireoide , Hormônios Tireóideos/metabolismo , Proteínas de Transporte/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Modelos Animais de Doenças , Proteínas de Membrana/metabolismo
6.
J Neurosci Res ; 102(5): e25339, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38741550

RESUMO

Diets rich in saturated fats are more detrimental to health than those containing mono- or unsaturated fats. Fatty acids are an important source of energy, but they also relay information regarding nutritional status to hypothalamic metabolic circuits and when in excess can be detrimental to these circuits. Astrocytes are the main site of central fatty acid ß-oxidation, and hypothalamic astrocytes participate in energy homeostasis, in part by modulating hormonal and nutritional signals reaching metabolic neurons, as well as in the inflammatory response to high-fat diets. Thus, we hypothesized that how hypothalamic astrocytes process-specific fatty acids participates in determining the differential metabolic response and that this is sex dependent as males and females respond differently to high-fat diets. Male and female primary hypothalamic astrocyte cultures were treated with oleic acid (OA) or palmitic acid (PA) for 24 h, and an untargeted metabolomics study was performed. A clear predictive model for PA exposure was obtained, while the metabolome after OA exposure was not different from controls. The observed modifications in metabolites, as well as the expression levels of key metabolic enzymes, indicate a reduction in the activity of the Krebs and glutamate/glutamine cycles in response to PA. In addition, there were specific differences between the response of astrocytes from male and female mice, as well as between hypothalamic and cerebral cortical astrocytes. Thus, the response of hypothalamic astrocytes to specific fatty acids could result in differential impacts on surrounding metabolic neurons and resulting in varied systemic metabolic outcomes.


Assuntos
Astrócitos , Hipotálamo , Ácido Oleico , Ácido Palmítico , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Ácido Oleico/farmacologia , Feminino , Ácido Palmítico/farmacologia , Hipotálamo/metabolismo , Hipotálamo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Caracteres Sexuais , Células Cultivadas
7.
Neurosci Biobehav Rev ; 161: 105668, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38608826

RESUMO

Neuroinflammation accompanies several brain disorders, either as a secondary consequence or as a primary cause and may contribute importantly to disease pathogenesis. Neurosteroids which act as Positive Steroid Allosteric GABA-A receptor Modulators (Steroid-PAM) appear to modulate neuroinflammation and their levels in the brain may vary because of increased or decreased local production or import from the systemic circulation. The increased synthesis of steroid-PAMs is possibly due to increased expression of the mitochondrial cholesterol transporting protein (TSPO) in neuroinflammatory tissue, and reduced production may be due to changes in the enzymatic activity. Microglia and astrocytes play an important role in neuroinflammation, and their production of inflammatory mediators can be both activated and inhibited by steroid-PAMs and GABA. What is surprising is the finding that both allopregnanolone, a steroid-PAM, and golexanolone, a novel GABA-A receptor modulating steroid antagonist (GAMSA), can inhibit microglia and astrocyte activation and normalize their function. This review focuses on the role of steroid-PAMs in neuroinflammation and their importance in new therapeutic approaches to CNS and liver disease.


Assuntos
Doenças Neuroinflamatórias , Pregnanolona , Pregnanolona/farmacologia , Pregnanolona/metabolismo , Humanos , Animais , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Antagonistas de Receptores de GABA-A/farmacologia
8.
Int J Biol Macromol ; 267(Pt 1): 131433, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38583846

RESUMO

Tannic acid (TA) exhibits low bioavailability in the gastrointestinal tract, limiting its benefits due to small amounts reaching the CNS. Thus, the objective of this study was to develop zein capsules and fibers by electrospraying/electrospinning for encapsulation of TA. Polymeric solutions were evaluated by electrical conductivity, density, and viscosity. In zein capsules, up to 2 % TA was added, and in fibers, up to 1 % TA was added. Zein capsule and fiber with TA were evaluated by morphology, size distribution, encapsulation efficiency, thermal and thermogravimetric properties, and functional groups. Zein capsule with 1.5 % TA was evaluated in astrocyte culture for cytotoxicity and antioxidant activity. TA zein capsules and fibers exhibited high encapsulation efficiency and homogeneous morphology. TA encapsulated in zein presented higher thermal stability than free TA. TA zein capsule did not present toxicity and elicited antioxidant action in lipopolysaccharide-induced astrocyte culture. Capsules and fibers were successfully produced by electrospraying/electrospinning techniques.


Assuntos
Antioxidantes , Astrócitos , Lipopolissacarídeos , Polifenóis , Taninos , Zeína , Taninos/química , Taninos/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Zeína/química , Antioxidantes/farmacologia , Antioxidantes/química , Lipopolissacarídeos/farmacologia , Animais , Escherichia coli/efeitos dos fármacos , Ratos , Células Cultivadas , Cápsulas
9.
Eur J Pharmacol ; 973: 176573, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38642669

RESUMO

Parkinson's disease (PD) is characterised by severe movement defects and the degeneration of dopaminergic neurones in the midbrain. The symptoms of PD can be managed with dopamine replacement therapy using L-3, 4-dihydroxyphenylalanine (L-dopa), which is the gold standard therapy for PD. However, long-term treatment with L-dopa can lead to motor complications. The central renin-angiotensin system (RAS) is associated with the development of neurodegenerative diseases in the brain. However, the role of the RAS in dopamine replacement therapy for PD remains unclear. Here, we tested the co-treatment of the angiotensin-converting enzyme inhibitor (ACEI) with L-dopa altered L-dopa-induced dyskinesia (LID) in a 6-hydroxydopamine (6-OHDA)-lesioned mouse model of PD. Perindopril, captopril, and enalapril were used as ACEIs. The co-treatment of ACEI with L-dopa significantly decreased LID development in 6-OHDA-lesioned mice. In addition, the astrocyte and microglial transcripts involving Ccl2, C3, Cd44, and Iigp1 were reduced by co-treatment with ACEI and L-dopa in the 6-OHDA-lesioned striatum. In conclusion, co-treatment with ACEIs and L-dopa, such as perindopril, captopril, and enalapril, may mitigate the severity of L-DOPA-induced dyskinesia in a mouse model of PD.


Assuntos
Inibidores da Enzima Conversora de Angiotensina , Modelos Animais de Doenças , Discinesia Induzida por Medicamentos , Levodopa , Oxidopamina , Animais , Levodopa/efeitos adversos , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Camundongos , Discinesia Induzida por Medicamentos/tratamento farmacológico , Discinesia Induzida por Medicamentos/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Captopril/farmacologia , Captopril/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Perindopril/farmacologia , Perindopril/uso terapêutico , Enalapril/farmacologia , Enalapril/uso terapêutico , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Antiparkinsonianos/farmacologia
10.
J Neurosci Res ; 102(4): e25334, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38656648

RESUMO

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.


Assuntos
Astrócitos , Diferenciação Celular , Deficiências de Ferro , Oligodendroglia , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Oligodendroglia/metabolismo , Oligodendroglia/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Proteínas de Transporte de Cátions/metabolismo , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Ratos , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/metabolismo , Desferroxamina/farmacologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Ferro/metabolismo
11.
Toxins (Basel) ; 16(4)2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38668612

RESUMO

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.


Assuntos
Bothrops , Venenos de Crotalídeos , Gânglios Espinais , Hiperalgesia , Receptores da Neurocinina-1 , Animais , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Venenos de Crotalídeos/toxicidade , Masculino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Receptores da Neurocinina-1/metabolismo , Minociclina/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Ratos , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Ratos Sprague-Dawley
12.
Cancer Cell ; 42(5): 904-914.e9, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38579724

RESUMO

A subset of patients with IDH-mutant glioma respond to inhibitors of mutant IDH (IDHi), yet the molecular underpinnings of such responses are not understood. Here, we profiled by single-cell or single-nucleus RNA-sequencing three IDH-mutant oligodendrogliomas from patients who derived clinical benefit from IDHi. Importantly, the tissues were sampled on-drug, four weeks from treatment initiation. We further integrate our findings with analysis of single-cell and bulk transcriptomes from independent cohorts and experimental models. We find that IDHi treatment induces a robust differentiation toward the astrocytic lineage, accompanied by a depletion of stem-like cells and a reduction of cell proliferation. Furthermore, mutations in NOTCH1 are associated with decreased astrocytic differentiation and may limit the response to IDHi. Our study highlights the differentiating potential of IDHi on the cellular hierarchies that drive oligodendrogliomas and suggests a genetic modifier that may improve patient stratification.


Assuntos
Neoplasias Encefálicas , Diferenciação Celular , Isocitrato Desidrogenase , Mutação , Oligodendroglioma , Oligodendroglioma/genética , Oligodendroglioma/patologia , Oligodendroglioma/tratamento farmacológico , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/antagonistas & inibidores , Humanos , Diferenciação Celular/efeitos dos fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem da Célula/efeitos dos fármacos , Receptor Notch1/genética , Receptor Notch1/metabolismo , Proliferação de Células/efeitos dos fármacos , Animais , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Camundongos , Análise de Célula Única/métodos
13.
Cancer Cell ; 42(5): 741-743, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38579726

RESUMO

In this issue of Cancer Cell, Spitzer and colleagues demonstrate the role of IDH inhibitors on IDHmutant gliomas in reducing proliferation and enhancing cell differentiation toward an astrocytic-like state, thus altering neurodevelopmental pathways. Despite clinical promise, unresolved questions regarding mechanisms of action and resistance underline the need for further research for treatment optimization.


Assuntos
Neoplasias Encefálicas , Glioma , Isocitrato Desidrogenase , Humanos , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/antagonistas & inibidores , Glioma/tratamento farmacológico , Glioma/patologia , Glioma/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Mutação , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo
14.
Int J Mol Sci ; 25(8)2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38673747

RESUMO

Neuroinflammation and epilepsy are different pathologies, but, in some cases, they are so closely related that the activation of one of the pathologies leads to the development of the other. In this work, we discuss the three main cell types involved in neuroinflammation, namely (i) reactive astrocytes, (ii) activated microglia, and infiltration of (iii) peripheral immune cells in the central nervous system. Then, we discuss how neuroinflammation and epilepsy are interconnected and describe the use of different repurposing drugs with anti-inflammatory properties that have been shown to have a beneficial effect in different epilepsy models. This review reinforces the idea that compounds designed to alleviate seizures need to target not only the neuroinflammation caused by reactive astrocytes and microglia but also the interaction of these cells with infiltrated peripheral immune cells.


Assuntos
Astrócitos , Reposicionamento de Medicamentos , Epilepsia , Microglia , Doenças Neuroinflamatórias , Humanos , Epilepsia/tratamento farmacológico , Reposicionamento de Medicamentos/métodos , Doenças Neuroinflamatórias/tratamento farmacológico , Animais , Microglia/efeitos dos fármacos , Microglia/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/farmacologia , Anticonvulsivantes/uso terapêutico , Anticonvulsivantes/farmacologia
15.
Int J Mol Sci ; 25(8)2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38673879

RESUMO

Reactive astrocytes are key players in HIV-associated neurocognitive disorders (HAND), and different types of reactive astrocytes play opposing roles in the neuropathologic progression of HAND. A recent study by our group found that gp120 mediates A1 astrocytes (neurotoxicity), which secrete proinflammatory factors and promote HAND disease progression. Here, by comparing the expression of A2 astrocyte (neuroprotective) markers in the brains of gp120 tgm mice and gp120+/α7nAChR-/- mice, we found that inhibition of alpha 7 nicotinic acetylcholine receptor (α7nAChR) promotes A2 astrocyte generation. Notably, kynurenine acid (KYNA) is an antagonist of α7nAChR, and is able to promote the formation of A2 astrocytes, the secretion of neurotrophic factors, and the enhancement of glutamate uptake through blocking the activation of α7nAChR/NF-κB signaling. In addition, learning, memory and mood disorders were significantly improved in gp120 tgm mice by intraperitoneal injection of kynurenine (KYN) and probenecid (PROB). Meanwhile, the number of A2 astrocytes in the mouse brain was significantly increased and glutamate toxicity was reduced. Taken together, KYNA was able to promote A2 astrocyte production and neurotrophic factor secretion, reduce glutamate toxicity, and ameliorate gp120-induced neuropathological deficits. These findings contribute to our understanding of the role that reactive astrocytes play in the development of HAND pathology and provide new evidence for the treatment of HAND via the tryptophan pathway.


Assuntos
Astrócitos , Ácido Glutâmico , Cinurenina , Animais , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Ácido Glutâmico/toxicidade , Camundongos , Cinurenina/metabolismo , Ácido Cinurênico/metabolismo , Ácido Cinurênico/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Proteína gp120 do Envelope de HIV/toxicidade , Transdução de Sinais/efeitos dos fármacos , Camundongos Knockout , Probenecid/farmacologia , Camundongos Endogâmicos C57BL , Masculino , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/efeitos dos fármacos , NF-kappa B/metabolismo
16.
Biomed Pharmacother ; 174: 116625, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38643543

RESUMO

AIMS: The purpose of this study was to explore the impacts of salidroside on vascular regeneration, vascular structural changes and long-term neurological recuperation following cerebral ischemia and its possible mechanism. MAIN METHODS: From Day 1 to Day 28, young male mice with middle cerebral artery blockage received daily doses of salidroside and measured neurological deficits. On the 7th day after stroke, the volume of cerebral infarction was determined using TTC and HE staining. Microvascular density, astrocyte coverage, angiogenesis and the expression of the Shh signaling pathway were detected by IF, qRTPCR and WB at 7, 14 and 28 days after stroke. Changes in blood flow, blood vessel density and diameter from stroke to 28 days were measured by the LSCI and TPMI. KEY FINDINGS: Compared with the dMACO group, the salidroside treatment group significantly promoted the recovery of neurological function. Salidroside was found to enhance cerebral blood flow perfusion and reduce the infarct on the 7th day after stroke. From the 7th to the 28th day after stroke, salidroside treatment boosted the expression of CD31, CD31+/BrdU+, and GFAP in the cortex around the infarction site. On the 14th day after stroke, salidroside significantly enhanced the width and density of blood vessels. Salidroside increased the expression of histones and genes in the Shh signaling pathway during treatment, and this effect was weakened by the Shh inhibitor Cyclopamine. SIGNIFICANCE: Salidroside can restore nerve function, improve cerebral blood flow, reduce cerebral infarction volume, increase microvessel density and promote angiogenesis via the Shh signaling pathway.


Assuntos
Isquemia Encefálica , Glucosídeos , Proteínas Hedgehog , Neovascularização Fisiológica , Fenóis , Transdução de Sinais , Animais , Glucosídeos/farmacologia , Fenóis/farmacologia , Masculino , Proteínas Hedgehog/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Camundongos Endogâmicos C57BL , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Modelos Animais de Doenças , Circulação Cerebrovascular/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Angiogênese
17.
Acta Neuropathol Commun ; 12(1): 66, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654316

RESUMO

The elderly frequently present impaired blood-brain barrier which is closely associated with various neurodegenerative diseases. However, how the albumin, the most abundant protein in the plasma, leaking through the disrupted BBB, contributes to the neuropathology remains poorly understood. We here demonstrated that mouse serum albumin-activated microglia induced astrocytes to A1 phenotype to remarkably increase levels of Elovl1, an astrocytic synthase for very long-chain saturated fatty acids, significantly promoting VLSFAs secretion and causing neuronal lippoapoptosis through endoplasmic reticulum stress response pathway. Moreover, MSA-activated microglia triggered remarkable tau phosphorylation at multiple sites through NLRP3 inflammasome pathway. Intracerebroventricular injection of MSA into the brains of C57BL/6J mice to a similar concentration as in patient brains induced neuronal apoptosis, neuroinflammation, increased tau phosphorylation, and decreased the spatial learning and memory abilities, while Elovl1 knockdown significantly prevented the deleterious effect of MSA. Overall, our study here revealed that MSA induced tau phosphorylation and neuron apoptosis based on MSA-activated microglia and astrocytes, respectively, showing the critical roles of MSA in initiating the occurrence of tauopathies and cognitive decline, and providing potential therapeutic targets for MSA-induced neuropathology in multiple neurodegenerative disorders.


Assuntos
Apoptose , Camundongos Endogâmicos C57BL , Neurônios , Albumina Sérica , Tauopatias , Animais , Humanos , Masculino , Camundongos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Astrócitos/metabolismo , Astrócitos/patologia , Astrócitos/efeitos dos fármacos , Elongases de Ácidos Graxos/metabolismo , Microglia/metabolismo , Microglia/efeitos dos fármacos , Microglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Neurônios/efeitos dos fármacos , Albumina Sérica/metabolismo , Albumina Sérica/farmacologia , Proteínas tau/metabolismo , Tauopatias/patologia , Tauopatias/metabolismo
18.
J Stroke Cerebrovasc Dis ; 33(6): 106578, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38636320

RESUMO

BACKGROUND: Notch1 signaling inhibiton with N-[N-(3,5-difluorophenacetyl)-1-alanyl]-S-phenylglycine t-butylester] (DAPT) treatment could promote brain recovery and the intervention effect is different between striatum (STR) and cortex (CTX), which might be accounted for different changes of glial activities, but the in-depth mechanism is still unknown. The purpose of this study was to identify whether DAPT could modulate microglial subtype shifts and astroglial-endfeet aquaporin-4 (AQP4) mediated waste solute drainage. METHODS: Sprague-Dawley rats (n=10) were subjected to 90min of middle cerebral artery occlusion (MCAO) and were treated with DAPT (n=5) or act as control with no treatment (n=5). Two groups of rats underwent MRI scans at 24h and 4 week, and sacrificed at 4 week after stroke for immunofluorescence (IF). RESULTS: Compared with control rats, MRI data showed structural recovery in ipsilateral STR but not CTX. And IF showed decreased pro-inflammatory M1 microglia and increased anti-inflammatory M2 microglia in striatal lesion core and peri-lesions of STR, CTX. Meanwhile, IF showed decreased AQP4 polarity in ischemic brain tissue, however, AQP4 polarity in striatal peri-lesions of DAPT treated rats was higher than that in control rats but shows no difference in cortical peri-lesions between control and treated rats. CONCLUSIONS: The present study indicated that DAPT could promote protective microglia subtype shift and striatal astrocyte mediated waste solute drainage, that the later might be the major contributor of waste solute metabolism and one of the accounts for discrepant recovery of STR and CTX.


Assuntos
Aquaporina 4 , Astrócitos , Dipeptídeos , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média , Microglia , Ratos Sprague-Dawley , Receptor Notch1 , Recuperação de Função Fisiológica , Transdução de Sinais , Animais , Aquaporina 4/metabolismo , Receptor Notch1/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Microglia/metabolismo , Microglia/efeitos dos fármacos , Microglia/patologia , Dipeptídeos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/patologia , Corpo Estriado/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Fatores de Tempo , Fármacos Neuroprotetores/farmacologia , AVC Isquêmico/metabolismo , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/fisiopatologia , AVC Isquêmico/patologia
19.
eNeuro ; 11(4)2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38604775

RESUMO

A sublethal ischemic episode [termed preconditioning (PC)] protects neurons in the brain against a subsequent severe ischemic injury. This phenomenon is known as brain ischemic tolerance and has received much attention from researchers because of its robust neuroprotective effects. We have previously reported that PC activates astrocytes and subsequently upregulates P2X7 receptors, thereby leading to ischemic tolerance. However, the downstream signals of P2X7 receptors that are responsible for PC-induced ischemic tolerance remain unknown. Here, we show that PC-induced P2X7 receptor-mediated lactate release from astrocytes has an indispensable role in this event. Using a transient focal cerebral ischemia model caused by middle cerebral artery occlusion, extracellular lactate levels during severe ischemia were significantly increased in mice who experienced PC; this increase was dependent on P2X7 receptors. In addition, the intracerebroventricular injection of lactate protected against cerebral ischemic injury. In in vitro experiments, although stimulation of astrocytes with the P2X7 receptor agonist BzATP had no effect on the protein levels of monocarboxylate transporter (MCT) 1 and MCT4 (which are responsible for lactate release from astrocytes), BzATP induced the plasma membrane translocation of these MCTs via their chaperone CD147. Importantly, CD147 was increased in activated astrocytes after PC, and CD147-blocking antibody abolished the PC-induced facilitation of astrocytic lactate release and ischemic tolerance. Taken together, our findings suggest that astrocytes induce ischemic tolerance via P2X7 receptor-mediated lactate release.


Assuntos
Astrócitos , Precondicionamento Isquêmico , Ácido Láctico , Camundongos Endogâmicos C57BL , Transportadores de Ácidos Monocarboxílicos , Receptores Purinérgicos P2X7 , Animais , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Precondicionamento Isquêmico/métodos , Ácido Láctico/metabolismo , Ácido Láctico/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Masculino , Transportadores de Ácidos Monocarboxílicos/metabolismo , Basigina/metabolismo , Isquemia Encefálica/metabolismo , Simportadores/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Modelos Animais de Doenças , Proteínas Musculares/metabolismo , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Camundongos , Células Cultivadas , Encéfalo/metabolismo , Camundongos Knockout
20.
Nat Commun ; 15(1): 3661, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38688901

RESUMO

Optochemistry, an emerging pharmacologic approach in which light is used to selectively activate or deactivate molecules, has the potential to alleviate symptoms, cure diseases, and improve quality of life while preventing uncontrolled drug effects. The development of in-vivo applications for optochemistry to render brain cells photoresponsive without relying on genetic engineering has been progressing slowly. The nucleus accumbens (NAc) is a region for the regulation of slow-wave sleep (SWS) through the integration of motivational stimuli. Adenosine emerges as a promising candidate molecule for activating indirect pathway neurons of the NAc expressing adenosine A2A receptors (A2ARs) to induce SWS. Here, we developed a brain-permeable positive allosteric modulator of A2ARs (A2AR PAM) that can be rapidly photoactivated with visible light (λ > 400 nm) and used it optoallosterically to induce SWS in the NAc of freely behaving male mice by increasing the activity of extracellular adenosine derived from astrocytic and neuronal activity.


Assuntos
Adenosina , Núcleo Accumbens , Receptor A2A de Adenosina , Sono de Ondas Lentas , Animais , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/fisiologia , Masculino , Receptor A2A de Adenosina/metabolismo , Receptor A2A de Adenosina/genética , Camundongos , Adenosina/metabolismo , Adenosina/farmacologia , Regulação Alostérica , Sono de Ondas Lentas/fisiologia , Sono de Ondas Lentas/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Luz , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Humanos , Agonistas do Receptor A2 de Adenosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...